Nutrition and Gene Expression (PDFDrive)
Nutrition and Gene Expression (PDFDrive)
Nutrition and Gene Expression (PDFDrive)
and
Gene
Expression
Edited by
This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been
made to publish reliable data and information, but the author and publisher cannot assume responsibility for the
validity of all materials or the consequences of their use. The authors and publishers have attempted to trace the
copyright holders of all material reproduced in this publication and apologize to copyright holders if permission to
publish in this form has not been obtained. If any copyright material has not been acknowledged please write and let
us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or
utilized in any form by any electronic, mechanical, or other means, now known or hereafter invented, including pho-
tocopying, microfilming, and recording, or in any information storage or retrieval system, without written permission
from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://
www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA
01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registration for a variety of
users. For organizations that have been granted a photocopy license by the CCC, a separate system of payment has
been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for
identification and explanation without intent to infringe.
Publisher’s Note
The publisher has gone to great lengths to ensure the quality of this reprint but points out that some imperfections in
the original copies may be apparent.
Disclaimer
The publisher has made every effort to trace copyright holders and welcomes correspondence from those they have
been unable to contact.
Visit the Taylor & Francis Web site at http://www.taylorandfrancis.com and the
CRC Press Web site at http://www.crcpress.com
PREFACE
The science of nutrition has changed radically since its inception. It has
evolved from the discovery of nutrients which cured well-described diseases
to a recognition that appropriate food choices can delay the development of
a variety of degenerative diseases. In the past, nutritionists worked hard to
devise recommendations to the public for nutrient intakes that would ensure
the absence of deficiency diseases. In the future, nutritionists will make
nutrient intake recommendations not on such characteristics as sex, height,
weight or age, but on the basis of the individual's phenotypic expression for
health while suppressing that individual's phenotypic expression for disease.
We are a long way from being able to make these recommendations. However,
as a first step we are beginning to recognize the interaction of specific nutrients
with the genetic code possessed by all nucleated cells. This book and its
sequels will provide the reader with some of the insights gained by foremost
scientists in the field of nutrient-gene interactions. Some of these interactions
have been researched in depth whereas others are in their infancy. As the
reader proceeds through the book, one hopes that the imagination and dedi-
cation of the scientists who have contributed to this volume will be appre-
ciated. To these authors who struggled to meet our stringent deadline, and
who wrote of their life's work, we give our thanks.
Chapter 1
Nutrient Receptors and Gene Expression ................................. .
James L. Hargrove and Carolyn D. Berdanier
Chapter 2
Lactose Intolerance and Regulation of Small Intestinal
Lactase Activity .......................................................... 23
Robert K. Montgomery, Hans A. Buller, Edmond H. H. M. Rings,
Jan Dekker, and Richard J. Grand
Chapter 3
Regulation of Gastrointestinal Lipase Gene Expression by
Dietary Lipids ............................................................ 55
Catherine Wicker-Pianquart and Antoine Puigserver
Chapter 4
Nutritional Control of Gastrointestinal Hormone
Gene Expression ......................................................... 91
Pauline Kay Lund
Chapter 5
Calcium Homeostasis, Endoplasmic Reticular Function, and the
Regulation of mRNA Translation in Mammalian Cells .................. 117
Margaret A. Brostrom and Charles 0. Brostrom
Chapter 6
Tissue-Specific Regulation of Glucokinase .............................. 143
Mark A. Magnuson and Thomas L. Jetton
Chapter 7
Dietary and Hormonal Regulation of L-Type Pyruvate Kinase
Gene Expression ........................................................ 169
Tamio Noguchi and Takehiko Tanaka
Chapter 8
Glucose-6-Phosphate Dehydrogenase: Diet and Hormonal
Influences on De Novo Enzyme Synthesis .............................. 187
Rolf F. Kletzien and Carolyn D. Berdanier
Chapter 9
Nutritional and Honnonal Regulation of Genes Encoding Enzymes
Involved in Fat Synthesis ................................................ 207
Hei Sook Sui, Naima Moustaid, Kenji Sakamoto, Cynthia Smas,
Nick Gekakis, and Ann Jerkins
Chapter 10
Regulation of Hepatic Gene Expression by Dietary Fats: A Unique
Role for Polyunsaturated Fatty Acids .................................... 227
Steven D. Clarke and Donald B. Jump
Chapter 11
Dietary Fat, Gene Expression, and Carcinogenesis ...................... 247
Howard P. Glauert
Chapter 12
Regulation of Acetyl CoA Carboxylase and Gene Expression ........... 269
John B. Allred and Diana F. Bowers
Chapter 13
Apolipoprotein B mRNA Editing ........................................ 297
Nicholas 0. Davidson
Chapter 14
Regulation of the 68 kDa Hepatic Camitine
Palmitoyltransferase ..................................................... 319
PaulS. Brady, Roderick A. Barke, and Linda J. Brady
Chapter 15
Long-Term Regulation of Hepatic Glutaminase and the Urea
Cycle Enzymes .......................................................... 335
Malcolm Watford
Chapter 16
Effects of Dietary Protein on Gene Expression .......................... 353
David K.-C. Chan and James L. Hargrove
Chapter 17
Vitamin D-Dependent Calcium Binding Protein, Calbindin-D:
Regulation of Gene Expression .......................................... 377
Rajbir K. Gill and Sylvia Christakos
Chapter 18
Vitamin D Regulation of Osteoblast Growth and Differentation ......... 391
Jane B. Lian and Gary S. Stein
Chapter 19
Retinoic Acid Regulation of Gene Expression in Adipocytes ............ 431
Donald B. Jump, Gerald J. Lepar, and
Ormond A. MacDougald
Chapter 20
The Role of Vitamin K in Clotting Factor Biosynthesis ................. 455
J. W. Suttie
Chapter 21
Role of Ascorbate in Regulating the Collagen Pathway ................. 483
Richard I. Schwarz
Chapter 22
Dietary Regulation of Metallothionein Expression ....................... 507
Neil F. Shay and Robert J. Cousins
Chapter 23
Iron-Dependent Regulation of Ferritin Synthesis ........................ 525
H. N. Munro, Z. Kikinis, and R. S. Eisenstein
Chapter 24
Vitamin B6 Modulation of Steroid Induced Gene Expression ............ 547
Douglas B. Tully, Victoria E. Allgood, and John A. Cidlowski
TABLE OF CONTENTS
I. Introduction ........................................................ 2
References ................................................................ 19
0-8493-6961-41931$0.00 +$.50
© 1993 by CRC Press. Inc. 1
2 Nutrition and Gene Expression
I. INTRODUCTION
site for sterols such as 25-hydroxy D3 that is thought to be encoded at the 5'
end of the gene on exons 1 and 2. The protein is multifunctional, and a region
that binds actin may be encoded on exon 10. 7 Vitamin D-binding protein is
thought to be related to albumin and a-fetoprotein. Ray et al. have proposed
that the latter proteins may have been generated from an ancestral gene that
resembled the gene for vitamin D-binding protein. 7
The rise of multicellular organisms permitted functionally distinct groups
of cells to be formed, including some that participated in storing or mobilizing
nutrient reserves. Functional specialization required that means for commu-
nication among groups of cells be developed, so that a signaling system would
permit nutrients to be withdrawn as needed by metabolically active cells.
Although secretion of metabolites and other small molecules may have served
for communication at first, finer controls eventually developed that gave rise
to paracrine and endocrine signals, intracellular signal transduction, and the
specialization of cells to coordinate the transfer of information.
Even though endocrine and neural mechanisms have supplanted nutrients
as primary signals in mammalian signaling networks, nutrient availability still
represents the primary signal for activating or inactivating most genetic net-
works in unicellular organisms. It is also likely that neural and endocrine
signals in multicellular organisms have not entirely replaced the early genetic
mechanisms, because neurotransmitters and hormones cooperate with nu-
trients in directing nutrient utilization. For these reasons, it is pertinent to
review the components of genetic feedback systems and the means by which
nutrients regulate genetic activity in unicellular organisms.
B
Effector Pathway Response Pathway
Receptor
v •
Signal
Transduction Altered
Metabolis
"/
Second (]) Activation
Messenger
·-~ t
DNA-Binding
~ Inactivation
or
Protein ~ Degradation
Translation
Transport
Processing
Transcription
FIGURE 1. (A) Components of a typical, mammalian gene include the promoter element,
which binds RNA polymerase II and related transcription factors; enhancer and silencer elements;
and the structural gene, which is divided into exons (the mRNA coding segments) and introns
(segments that are removed during processing of the primary transcript as mature mRNA is
formed). Trans-acting factors include proteins (and possibly RNA molecules) that modify the
rate of transcription by binding to regulatory elements in the gene. (B) Responses of genes to
nutrients require an effector pathway (left side of figure) that confers specificity of response
according to cell type and kind of nutrient, and a response pathway (right side of figure) that
may be affected at any of several levels by the nutrient or its metabolites.
most genes are subject to multiple controls. However, genes that are fully
active in only one or a few tissues tend to be transcribed at a higher basal
rate, contain a TATA element, and respond actively to signal molecules such
as hormones and second messengers. Such genes are said to be "inducible",
6 Nutrition and Gene Expression
and the processes by which enzymes and mRNAs increase to new levels are
called induction. This process occurs in response to proteins that bind to
regulatory portions of genes that may be adjacent to the promoter, but can
also occur at long distances away in either direction. These regulatory seg-
ments are termed enhancer elements if they increase transcription, and silencer
elements if they decrease transcription. By affecting proteins that bind to these
parts of genes, nutrients and hormones may provide specificity of genetic
responses.
interact with parts of the ligands have been identified. For instance, the
arabinose-binding protein will complex with arabinose, glucose, or fucose by
forming hydrogen bonds with sugar hydroxyl groups; the protein binds the
sugars through functional groups of amino acids in its primary structure,
including amides, hydroxyls, and carboxyl groups. The ligand is further
stabilized by stacking between hydrophobic residues that hold the sugar ring
in place. Interaction with ligands alters conformation of the binding proteins,
which are thought to interact with other proteins in the plasma membrane that
initiate active transport of the nutrient or affect flagellar motility or chemo-
taxis. At the atomic level, similar kinds of interactions might be expected to
occur in mammalian proteins that bind particular nutrients.
Intracellular binding proteins mediate the effects of sugars on bacterial
gene expression with the same high degree of specificity that is typical of
other protein-ligand interactions such as enzymatic catalysis. In the case of
the lactose operon, a protein called the lac repressor normally prevents tran-
scription from this promoter. The lac repressor is a tetrameric protein that
binds the inducing sugar with high affinity, and also has two binding sites
that recognize a specific DNA sequence present in the lac promoter. 21 When
the inducing metabolite, allolactose, binds to the repressor, the repressor
dislodges from promoter DNA, and transcription of the gene by RNA po-
lymerase may begin. Because the repressor mediates the genetic effect, it
could be considered a nutrient receptor for activation of the lac operon.
It might be argued that genes in most mammalian cells differ from those
in bacteria because they do not respond directly to nutrients, but instead are
controlled by hormones or second messengers that are generated in response
to nutrients. However, bacteria have similar mechanisms by which nutritional
status is encoded by a signal molecule. For example, dissociation of the lac
repressor by itself is ineffective in increasing transcription from the lac operon.
Also needed is a positive regulator called the catabolite activator protein
(CAP). Metabolism of glucose inactivates adenylyl cyclase and causes levels
of cAMP to decline, and the lac operon can only be activated if the complex
of cAMP and the activator protein is bound to the promoter DNA. Low levels
of cAMP indicate that glucose is being utilized, and that there is no need to
make use of lactose. The production of cAMP in mammalian liver may
indicate low glucose status in the whole organism as reflected by output of
insulin and glucagon by the pancreas. However, it should be expected that
many genetic responses of mammals might not involve unmodified nutrients,
but a signal related to nutrient utilization. For instance, glycolysis is regulated
by availability of several compounds of low molecular weight, including
glucose-6-phosphate, fructose-2, 6-(bis)phosphate, citrate, NAD+, and phos-
phorylation state of the cytosol (ratio of ATP to ADP + AMP + P;)Y It is
feasible that one or more of these compounds could serve as a metabolic
signal that activates transcription of genes whose products are needed for
integrating glycolysis with other metabolic pathways.
Hargrove and Berdanier 9
G~~~~P)
~Galactose +
Activation
Repression ++ Repression
Glucose:
GAL10 GAL1
FIGURE 2. Elements of a genetic switch for regulation of galactose utilization by the yeast,
Saccharomyces cerevisiae, are shown. The switch contains an upstream activation sequence
(shaded bar) that controls transcription from two genes, GALl and GALlO (indicated by right
and left arrows, respectively). Transcription is inhibited by two major inhibitory elements:
(I) Multiple copies of the operators 01 to 06 are present, which prevent transcription when
glucose is available; (2) The inhibitory protein, ga180p, binds to a transcriptional activator unless
galactose is present. An inducer is then thought to be produced that causes ga180p to dissociate
from ga14p. When ga14p binds to multiple activating-elements (boxes 1 to 4), transcription is
stimulated by several orders of magnitude compared to the repressed rate. Other activating
elements, such as GAEl and 2, are present in this complex promoter; the figure was drawn
based on data presented by Finley et al. 25 and Yun et al. 26
the induction process, the protein, gal4p, becomes phosphorylated, and evi-
dence suggests that a protein kinase is necessary for derepression of glucose-
repressible genes. It is of considerable interest that the product of the TUPl
locus, which is needed for glucose repression, is structurally similar to mam-
malian G proteins that participate in signal transduction. 28
Although yeast do possess adenyl cyclase, the role of cAMP in glucose
repression is uncertain. However, addition of glucose to derepressed cells
results in production of cAMP and a cascade of protein phosphorylation,
which causes a transition to the repressed state. Transport of glucose is not
sufficient to generate cAMP; the cells must also contain a hexokinase or
glucokinase. It is not certain whether the sugar must be phosphorylated, or
if another function is served by the hexose kinases and/or transporters. At
least two genes encode hexose transport systems, which are homologous to
the mammalian glucose-transport proteins and contain leucine zipper motifs
that might have potential for binding to DNA. It has been postulated that they
may function as glucose sensors. 29 In addition, the glucose-induced signal
requires a RAS protein that is a member of a proto-oncogene family, and a
CDC protein that forms a complex with the RAS protein and adenyl cyclase. 30
Hargrove and Berdanier 11
Metabolic Behavioral
Integration Integration
Pituitary
+ C rt
o ex
""";'"r;".""/
+.'
Hypothalamus Autonomic
Integration
·~"'"'"h"' "'
~----~
Receptors in
Area Postrema
Nutrient Sensors in
Abdominal Organs
FIGURE 3. Neural and endocrine systems for sensing nutrients communicate between the gut,
brain, and peripheral organs. Receptors for glucose and amino acids are thought to be present
on sensory endings of afferent fibers in the vagus nerve (right side of figure). These fibers pass
to the nucleus of the tractus solitarius, which is a major relay for chemical sensation and
baroreflexes; it sends information to the hypothalamus and also controls efferent activity of the
sympathetic and parasympathetic nervous system. Nutrient receptors are also present in endocrine
cells of the gut and glands such as the pancreas, which secrete numerous peptide hormones in
response to dietary constituents (left side of figure). Blood passing to the liver, peripheral organs,
and brain contains nutrients and hormones that may alter gene expression. The blood-brain barrier
prevents much of this information from reaching the brain directly, but circumventricular organs,
such as the area postrema, lack this barrier. They contain receptors for nutrients and hormones
and relay information to other parts of the brain. Small molecules such as glucose pass through
the blood-brain barrier and may directly stimulate neurons in the hypothalamus. These interactions
serve to integrate autonomic function, metabolic activity, and behavior.
14 Nutrition and Gene Expression
contains secretory nerve endings that are not thought to convey information
to other parts of the brain.
Some neurons in the brain are sensitive to nutrients, particularly glucose
and amino acids. Glucose inhibits the rate at which action potentials are
generated by neurons in the area postrema and in the ventromedial hypo-
thalamus, indicating a possible chemosensory function. 34 Amino acids in the
bloodstream are capable of affecting nerve function in the circumventricular
organs; for instance, large doses of glutamate cause cell death in the area
postrema. 46 In contrast, neurons that are protected by the blood-brain barrier
are spared, but utilize amino acids as transmitters. Receptors for excitatory
amino acids in the circumventricular organs could function as nutrient sensors,
but the gating function of the blood-brain barrier makes this less likely for
neurons that are sited deeper in the brain.
No matter how indirect the route, information concerning nutrient status
alters genetic activity in the brain. Messenger RNAs corresponding to neu-
ropeptide Y, cholecystokinin, and pituitary hormones are affected by fasting
and dietary composition. 44 .45 Recent work by Oomura and colleagues47 has
identified a striking effect of feeding or glucose administration on release of
a protein from ependymal cells that line the third ventricle of the lateral
hypothalamus. Owing to its growth-promoting activity, the protein is named
fibroblast growth factor (FGF), but this may be misleading. The concentration
of the protein in the cerebrospinal fluid (CSF) increases as much as 10,000-
fold when glucose concentrations in the CSF increase 2- to 4-fold after a
meal. FGF suppressed the activity of glucose-sensitive neurons in the lateral
hypothalamic area, but not in the ventromedial hypothalamus, and suppressed
feeding. When antibodies to FGF were injected bilaterally into the third
ventricle, nocturnal food intake increased. This evidence suggests that epen-
dymal cells may relay information concerning nutrient status to neurons in-
volved in ingestive behavior, and possibly to neurons that produce releasing
factors involved in output of pituitary hormones that respond to nutrient status.
. ,• ... is
G)
.: 110 .~ B
>
A E
~ 100
•
l!
Add Hormone f 6
·;: ~
Q
90
0 g
c 80
•
'04
Gl
..
Gl
70 ••• 0
E
••
LL
>.
0
••
.
Gl
2
.
.<:
60 Minus Hormone 0
0.. u
0
.<: 50 "'
00
D.. 0 10 20 30 40 0 20 40
50 60 80
Time (Minutes) Time (Minutes)
2 40 '2
~·;: c 0
Oi
::I
•••••••••••••••••••••••• 30
:.::
.. 1:
0 .5 ~
~Q
. •.•. •.•. -~· · ·-· ... 1:
>-o
~0
Ill
;: . •.•. 20 .J:J
:a
0
1:
·-
. •·····
•••·•.((-- Enzyme Activity
0..111
C( 10 w.,
o...,
................ ·········•
z
a: Gl
E t1:
0 ~~~-.--~---r--~--.---~-.--~---.--~--+o
0 2 4 6 8 10 12
Time (Hours)
FIGURE 4. The time scale of genetic responses is much slower than metabolic control exerted
by phosphorylation/dephosphorylation in response to the same initiating stimulus. (A) Response
of hepatic glycogen phosphorylase to addition of epinephrine (arrow) or glucagon; 48 (B) output
of glucose from liver slices after addition of glucagon; 49 (C) response of the gene for PEPCK
in glucose-fed rats to addition of cAMP or starvation gives rise to elevated mRNA and enzyme
(only the increase in enzyme level, and not the total concentration, is plotted). 50 •51 Note that
phosphorylase is maximally activated within 5 min after addition of hormone, compared to
several hours needed for transcription to fully alter enzyme levels. Panels A through C were
drawn from data in the references cited.
c (I)
Transcription
ks1
.:....-.......
~
NUCLEOTIDES mRNA Pool
kd1 Rt Rt
mRNA Degradation
Translation
kd2Pt Pt
Protein Degradation
FIGURE 5. Basis for a quantitative model that shows why gene products accumulate slowly
in response to altered rates of transcription. Specific mRNAs are synthesized from nucleotides
at relatively constant rates unless regulatory molecules alter their transcription; the rate is not
proportional to mRNA concentration. In contrast, the rate of mRNA decay is usually proportional
to mRNA concentration. This gives rise to a model based on zero-order synthesis and first-order
decay, which predicts that a period equal to the half-life of an mRNA or protein must pass before
induction can be 50% complete. Supporting data are discussed elsewhere. 53
REFERENCES
I. Shils, M. E. and Young, V. R., Modern Nutrition in Health and Disease, 7th edition,
Lea and Febiger, Philadelphia, 1988.
2. Young, V. R. and Marchini, J. S., Mechanisms and nutritional significance of metabolic
responses to altered intakes of protein and amino acids, with reference to nutritional
adaptation in humans, Am. J. Clin. Nutr., 51,270, 1990.
3. Eisenstein, R. S. and Harper, A. E., Relationship between protein intake and hepatic
protein synthesis in rats, J. Nutr., 121, I 581, 1990.
4. Shay, N. F., Nick, H, R., and Kilberg, M. S., Molecular cloning of an amino acid-
regulated mRNA (amino acid starvation-induced) in rat hepatoma cells, J. Bioi. Chern.,
265, 17844, 1990.
S. Munro, S. and Pelham, H. R. B., An Hsp70-like protein in the ER: identity with 78 kd
glucose-regulated protein and immunoglobulin heavy chain binding protein, Cell, 46,
291, 1986.
6. Hoyle, G. W. and Hill, R. L., Structure of the gene for a carbohydrate-binding receptor
unique to rat Kupffer cells, J. Bioi. Chern., 266, 1850, 1991.
7. Ray, K., Wang, X., Zhao, M., and Cooke, N. E., The rat vitamin D binding protein
(Gc-globulin) gene. Structural analysis, functional and evolutionary correlations, J. Bioi.
Chern., 266, 6221, 1991.
20 Nutrition and Gene Expression
8. Wingender, E., Transcription regulating proteins and their recognition sequences, Crit.
Rev. Eukaryotic Gene Expr., 1, 11, 1990.
9. Boyer, T. G. and Maquat, L., Minimal sequence requirements for the initiation of
transcription from an atypical, TATATAA box-containing housekeeping promoter, J. Bioi.
Chern., 265, 20524, 1990.
10. Dynan, W. S. and Tjian, R., Control of eukaryotic messenger RNA synthesis by
sequence-specific DNA-binding proteins, Nature (London), 316, 774, 1985.
II. Betz, H., Ligand-gated ion channels in the brain: the amino acid receptor superfamily,
Neuron, 5, 383, 1990.
12. Verdoorn, T. A., Burnashev, N., Monyer, H., Seeburg, P. H., and Sakmann, B.,
Structural determinants of ion flow through recombinant glutamate receptor channels,
Science, 252, 1715, 1991.
13. Durnam, D. M. and Palmiter, R. D., Transcriptional regulation of the mouse metal-
lothionein-1 gene by heavy metals, J. Bioi. Chern., 256, 5712, 1981.
14. Thiel, E. C., Regulation of ferritin and transferrin receptor mRNAs, J. Bioi. Chern.,
265, 4771, 1990.
15. Matschinsky, F., Glucokinase as glucose sensor and metabolic signal generator in pan-
creatic [3-cells and hepatocytes, Diabetes, 39, 647, 1990.
16. Hoenig, M., Lee, R. J., and Ferguson, D. C., Glucose inhibits the high-affinity (Ca+ 2 -
Mg+2)-ATPase in the plasma membrane of a glucose-responsive insulinoma, Biochim.
Biophys. Acta, 1022, 333, 1990.
17. Monod, J., The phenomenon of enzymatic adaptation and its bearing on problems of
genetics and cellular differentiation, Growth, II, 223, 1947.
18. Jacob, F. and Monod, J., On the regulation of gene activity, Cold Spring Harbor Symp.
Quant. Bioi., 26, 193, 1960.
19. Quiocho, F. A., Vyas, N. K., Sack, J. S., and Vyas, M. N., Atomic protein structures
reveal basic features of binding of sugars and ionic substrates, and calcium cation, Cold
Spring Harbor Symp. Quant. Bioi., 52, 453, 1987.
20. Vyas, N. K., Vyas, M. N., and Quiocho, F. A., Sugar and signal-transducer binding
sites of the Escherichia coli galactose chemoreceptor protein, Science, 242, 1290, 1988.
21. Pace, H. C., Lu, P., and Lewis, M., lac repressor: crystallization of intact tetramer
and its complexes with inducer and operator DNA, Proc. Nat!. Acad. Sci. U.S.A., 87,
1870, 1990.
22. Pilkis, S. J., El-Maghrabi, M. R., and Claus, T. H., Hormonal regulation of hepatic
gluconeogenesis and glycogenolysis, Annu. Rev. Biochem., 57, 755, 1988.
23. Strathern, J, N., Jones, E. W., and Broach, J. R., Eds., The Molecular Biology of
the Yeast Saccharomyces. Metabolism and Gene Expression, Cold Spring Harbor Lab-
oratory, New York, 1982.
24. Entian, K.-D., Glucose repression: a complex regulatory system in yeast, Microbial.
Sci .. 3, 366, 1986.
25. Finley, R. L., Jr., Chen, S., Ma, J., Byrne, P., and West, R. W., Jr., Opposing
regulatory functions of positive and negative elements in UASG control transcription of
the yeast GAL genes, Mol. Cell. Bioi., 10, 5663, 1990.
26. Yun, S.-J., Hiraoka, Y., Nishizawa, M., Takio, K., Titani, K., Nogi, Y., and Fukusawa,
T., Purification and characterization of the yeast negative regulatory protein GAL80, J.
Bioi. Chern., 266, 693, 1991.
27. Bhat, P. J. and Hopper, J, E., The mechanism of inducer formation in ga/3 mutants
of the yeat galactose system is independent of normal galactose metabolism and mito-
chondrial respiratory function, Genetics, 128, 233, 1991.
28. Williams, F. E. and Trumbly, R. J., Characterization of TUP!, a mediator of glucose
repression in Saccharomyces cerevisiae, Mol. Cell. Bioi., 10, 6500, 1990.
29. Lewis, D. A. and Bisson, L. F., The HXTI gene product of Saccharomyces cerevisiae
is a new member of the family of hexose transporters, Mol. Cell. Bioi., II, 3804, 1991.
Hargrove and Berdanier 21
30. Van Aelst, L., Jans, A. W. H., and Thevelein, J. M., Involvement of the CDC25
gene product in the signal transmission pathway of the glucose-induced RAS-mediated
cAMP signal in the yeast, Saccharomyces cerevisiae, J. Gen. Microbial., 137, 341, 1991.
31. Zhou, K. and Kohlhaw, G., Transcriptional activator LEU3 of yeast. Mapping of the
transcriptional activation function and significance of activation domain tryptophans, J.
Bioi. Chern., 265, 17409, 1990.
32. Mei, N., Intestinal chemosensitivity, Physiol. Rev., 65, 211, 1985.
33. Niijima, A., Glucose-sensitive afferent nerve fibers in the liver and their role in food
intake and blood glucose regulation, J. Autonomic Nervous Syst., 9, 207, 1983.
34. Oomura, Y. and Yoshimatsu, H., Neural networks of glucose monitoring system, J.
Autonomic Nervous Sysr., 10, 359, 1984.
35. Vigas, M., Tatar, P., Jurcovicova, J., and Jezova, D., Glucoreceptors located in
different areas mediate the hypoglycemia-induced release of growth hormone, prolactin,
and adrenocorticotropin in man, Neuroendocrinology, 51, 365, 1990.
36. Ewart, W. R., Medullary integration of afferent information from the gastrointestinal
tract, in Brain-Gut Interactions, Tache, Y. and Wingate, D., Eds., CRC Press, Boca
Raton, FL, 1991, 109.
37. Rehfeld, J, F., Cholecystokinin, in Handbook of Physiology, Seeton 6: The Gastroin-
testinal System, Vol. II, Schultz, S., Makhlouf, G. M., and Rauner, B. B., Eds., 1989,
337.
38. Brown, J. C., Buchan, A.M. J., Mcintosh, C. H. S., and Pederson, R. A., Gastric
inhibitory peptide, Brain-Cut Interactions, Tache, Y. and Wingate, D., Eds., CRC Press,
Boca Raton, FL, 1991, 109.
39. Taylor, I. L., Pancreatic polypeptide family: pancreatic polypeptide, neuropeptide Y,
and peptide YY, Brain-Gut Interactions, Tache, Y. and Wingate, D., Eds., CRC Press,
Boca Raton, FL, 1991,109.
40. Miselis, R. R., Shapiro, R. E., and Hyde, T. M., The area postrema, in Circumven-
tricular organs and body fluids, Vol. II, Gross, P. M., Ed., CRC Press, Boca Raton,
FL, 1987, 185.
41. Liddle, R. A., Carter, J.D., and McDonald, A. R., Dietary regulation of rat intestinal
cholecystokinin gene expression, J. Clin. Invest., 81, 2015, 1988.
42. Czyzyk-Krzeska, M. F., Bayliss, D. A., Seroogy, K. B., and Millhorn, D. E., Gene
expression for peptides in neurons of the petrosal and nodose ganglia of the rat, Exp.
Brain Res., 83, 411, 1991.
43. Adachi, A., Kobashi, M., Miyoshi, N., and Tsukamoto, G., Chemosensitive neurons
in the area postrema of the rat and their possible functions, Brain Res. Bull., 26, 137,
1991.
44. Brady, L. S., Smith, M.A., Gold, P. W., and Herkenham, M., Altered expression
of hypothalamic neuropeptide mRNAs in food-restricted asnd food-deprived rats, Neu-
roendocrinology, 52,441, 1990.
45. Chua, S. C., Jr., Liebel, R. L., and Hirsch, J., Food deprivation and age modulate
neuropeptide gene expression in the murine hypothalamus and adrenal gland, Mol. Brain
Res., 9, 95, 1991.
46. Phelix, C. F. and Hartle, D. K., Systemic glutamate induces degeneration of a sub-
population of serotonin-immunoreactive neurons in the area postrema of rats, Neurosci.
Lett., 117, 31, 1990.
47. Oomura, Y., Sasaki, K., Suzuki, K., Muto, T., Li, A., Ogita, A.-I., Hanai, K.,
Tooyama, I., Kimura, H., and Yanaihara, N., A new brain glucosensor and its phys-
iological significance, Am. J. Clin. Nutr., 55, 278S, 1992.
48. Sutherland, E. W., The effect of the hyperglycemic factor and epinephrine on enzyme
systems of liver and muscle, Ann. N.Y Acad. Sci., 54, 693, 1951.
49. Rail, T. W., Sutherland, E. W., and Wosilait, W. D., The relationship of epinephrine
and glucagon to liver phophorylase, J. Bioi. Chern., 218, 483, 1956.
22 Nutrition and Gene Expression
50. Hopgood, M. F., Ballard, F. J., Reshef, L., and Hanson, R. W., Synthesis and
degradation of phosphoenolpyruvate carboxylase in rat liver and adipose tissue. Changes
during a starvation-re-feeding cycle, Biochern. J.. 134, 445, 1975.
51. Beale, E. G., Hartley, J. L., and Granner, D. K. N6, 0 2 '-dibutyryl cAMP and glucose
regulate the amount of messenger RNA coding for hepatic phosphoenolpyruvate car-
boxykinase (GTP), J. Bioi. Chern., 257, 2022, 1982.
52. Goldstein, A., Aronow, L., and Kalman, S.M., Principles of Drug Action, 2nd ed.,
Harper and Row, New York, 1976, 280.
53. Hargrove, J. L., Hulsey, M.G., and Beale, E. G., The kinetics of mammalian gene
expression, BioEssays, 13, 667, 1991.
54. Moore, R. E., Goldsworthy, T. L., and Pitot, H. C., Turnover of 3' -po1yadenylate-
containing RNA in livers from aged, partially hepatectomized, neonatal, and Morris
5123C hepatoma-bearing rats, Cancer Res., 40, 1449, 1980.
55. Shapiro, D. J., Blume, J. E., and Nielsen, D. A., Regulation of messenger RNA
stability in eukaryotic cells, BioEssays, 6, 221, 1987.
56. Granner, D., Andreone, T., Sasaki, K., and Beale, E., Inhibition of transcription of
the phosphoenolpyruvate carboxykinase gene by insulin, Nature, 305, 549, 1983.
57. Sasaki, K., Cripe, R., Koch, S., Andreone, T. L., Petersen, D. D., Beale, E. G.,
and Granner, D. K., J. Bioi. Chern., 259, 15242, 1984.
58. Sasaki, K. and Granner, D. K., Regulation of phosphoenol-pyruvate carboxykinase
gene transcription by insulin and cAMP: reciprocal actions on initiation and elongation,
Proc. Nat/. Acad. Sci. U.S.A., 85, 2954, 1988.
59. Sehgal, P., Derman, E., Molloy, G. R., Tamm, I., and Darnell, J. E., 5,6-Dich1oro-
1-f3-D-ribofuranosylbenzimidazole inhibits initiation of nuclear heterogeneous RNA chains
in HeLa cells, Science, 194, 431, 1976.
60. Chrapkiewicz, N. B., Beale, E. G., and Granner, D. K., Induction of the messenger
ribonucleic acid coding for phosphoenolpyruvate carboxykinase in H4IIE cells. Evidence
for a nuclear effect of cAMP, J. Bioi. Chern., 257, 14428, 1982.
61. Palmiter, R. D., Quantitation of parameters that determine the rate of ovalbumin syn-
thesis, Cell, 4, 189, 1975.
62. Thompson, K. S. and Towle, H. C., Localization of the carbohydrate response element
of the rat L-type pyruvate kinase gene, J. Bioi. Chern., 266, 8679, 1991.
63. McGrane, M. M., Yun, J. S., Patel, Y. M., and Hanson, R. W., Metabolic control
of gene expression: in vivo studies with transgenic mice, Trends Biochern. Sci., 17, 40,
1992.
Chapter 2
TABLE OF CONTENTS
Acknowledgments ........................................................ 47
References ................................................................ 47
The use of milk as food for adults has its origins in prehistory. The
average North American or Northern European has been taught to think of
milk (usually cow's milk) as the perfect food. This attitude is so pervasive
that an inability to drink milk is considered a serious abnormality. Although
the terminology of milk or lactose intolerance and lactase deficiency is well
established in the literature, they are misnomers and misleading, as will be
discussed below. Milk intolerance can be due to allergies to milk proteins or
to the inability to digest milk sugar or lactose; the remainder of this review
will be confined to the latter: lactose intolerance. Lactose, the major carbo-
hydrate in milk, is hydrolyzed to glucose and galactose by the small intestinal
enzyme, lactase-phlorizin hydrolase (E.C. 3.2.1.23; 3.2.1.62), subsequently
referred to as lactase. Lactose intolerance is due to reduced levels of lactase
in the small intestine. The clinical aspects of lactose intolerance and nutrition, 1
and the population distribution of high and low lactase levels, have been
published in detaiF and will be reviewed only briefly. The major part of the
discussion will focus on recent progress in understanding the structure and
function of lactase and its regulation.
It is now clear that although milk intolerance is a frequent problem in
human adults, it is not a disease, nor an abnormality, but the normal condition
for most humans. 3 •4 Although milk intolerance was first described in 190 I , 5
a clear understanding of the digestion and absorption of lactose depended on
the availability of reliable tests. The development of the glucose oxidase
method by Dahlqvist6 provided a rapid, accurate method for assaying lactase
in tissue samples, including intestinal biopsies. In combination with mea-
surements of blood glucose levels following ingestion of a standardized lactose
dose, this method enabled investigators to link intestinal lactase activity levels
with lactose absorption and lactose intolerance. 7 Numerous studies using these
methods have delineated the distribution of lactase along the small intestine,
during human development, and in different human populations throughout
the world (summarized in References 2 and 8 through 10).
Montgomery et al. 25
from the stomach to the cecum may be as rapid as 10 min. 23 Finally, fecal
flora are known to adapt to ingested carbohydrate, and it is clear that non-
absorbed lactose can be salvaged by the colonic flora by fermentation to short
chain fatty acids, hydrogen, methane, water, carbon dioxide, and the pro-
duction of energy. 24 If lactose is provided slowly over a long period of time
in many ''intolerant'' people, the flora may adapt to the load and symptoms
produced by gas and acid in the colon may be reduced or eliminated. 1 This
mechanism of lactose tolerance in people with low lactase levels accounts for
the discrepancy in some studies in which "lactase deficient patients" are not
lactose intolerant. 25 Colonic distension may also vary among lactose intolerant
people, providing another variable in the expression of symptoms after lactose
ingestion.
A. DIAGNOSIS
The diagnosis of lactose malabsorption and its pathogenesis is based on
a combination of clinical findings and the results of appropriate tests. ' 9·26.29
The use of screening tests for lactose malabsorption in the diagnosis of lactose
intolerance has received wide attention. 19 ·26
E. COMPARISON OF TESTS
Studies are available which have compared intestinal histology, lactase
activity, and breath hydrogen test results in children with chronic diarrhea or
abdominal pain. 25 •27 In patients with abnormal intestinal histology, approxi-
mately 75% had abnormal lactose breath hydrogen values (sensitivity 75%).
However, in patients with normal histology, the lactose breath hydrogen test
was only 54% specific, reflecting the presence of a patchy villus lesion. Very
similar values were found when lactase activities on biopsy were compared
with results of lactose breath hydrogen tests. 25 •29 Accordingly, an abnormal
lactose breath hydrogen test is of value in identifying those patients who will
be symptomatic after lactose ingestion. In children less than 5 years of age,
an abnormal lactose breath hydrogen test always signifies abnormal intestinal
mucosa, which usually needs further definition with a small intestinal biopsy.
A normal lactose breath hydrogen test does not rule out an intestinal mucosal
abnormality and should not be used to avoid an intestinal biopsy in the
diagnosis of suspected mucosal disease (e.g., gluten sensitive enteropathy)Y
Since lactose is a key nutrient, lactase plays a critical role in the nutrition
of mammalian neonates. As mentioned above, human congenital lactase de-
ficiency, present from birth, is extremely rare and inherited as an autosomal
recessive gene. 10 In adulthood, milk drinking is considered normal among
populations of Northern European extraction and a few other groups. How-
ever, for most humans and all other adult mammals, significant milk ingestion
results in mild to severe gastrointestinal symptoms, caused by the inability
to digest lactose, 2 due to low levels of intestinal lactase. The initial human
studies were performed in adult patients of Northern European origin who
had elevated lactase. The data obtained led to the assumption that high lactase
30 Nutrition and Gene Expression
TABLE 1 TABLE 2
Distribution of Lactase Distribution of Lactase Phenotypes in
Phenotypes in Selected Selected European Populations
Populations in the
United States Low lactase
Country Population (%)
Low lactase
Sweden Swedes
Population (%)
Netherlands Dutch 0
Austria Austrians 20
Northern European 7
France French 32
Whites 22
Southern French 44
Blacks 65
Italy Northern Italians 50
American Indians 95
Southern Italians 72
Vietnamese 100
Sicilians 71
that patients, who on biopsy were lactase deficient, did not report symptoms
of lactose intolerance. 45 .4 6 Another major source of confusion was the dis-
appearance of symptoms of lactose malabsorption after prolonged periods of
lactose ingestion in lactose intolerant patients. In addition to clinical obser-
vations, the induction hypothesis was supported by studies which showed that
lactase activity increased in adult rats after very high lactose intake. 47•48 How-
ever, the changes in lactase activity were relatively small compared to the
magnitude of decrease that occurs around the weaning period, and high glu-
cose intake produced comparable effects on lactase activity. In the studies by
Lebenthal et a!., 48 lactase activity was higher during prolonged nursing than
in controls, but the decline of lactase activity around weaning was not pre-
vented by these experimental conditions.
Kretchmer4 1 described Nigerian medical students with clinically proven
lactose intolerance who remained unable to digest lactose, when tested by
lactose tolerance tests, after 6 months of daily lactose intake (50 g). Never-
theless, the students gradually adjusted to this large lactose intake and did
not show any symptoms of intolerance. It is now clear that the explanation
for this adaptive response was the emergence of fecal flora capable of fer-
menting the nonabsorbed lactose to short chain fatty acids. 49 Thus, there is
no compelling evidence for adaptive changes in small intestinal lactase activity
in humans.
The genetic hypothesis was first put forward in 1966 when Bayless and
co-workers showed that 70% of a group of black Americans examined in
Baltimore were lactose malabsorbers, while only 8% of the white subjects
were malabsorbers. 9 Many subsequent studies correlated racial and ethnic
distribution of lactase enzyme levels and lactose intolerance in adults (sum-
marized in References 2, 4, 50). The necessary genetic analysis was provided
by Sahi, who showed Mendelian inheritance of lactose malabsorption as an
autosomal recessive trait in a large family study from Finland. 51 Similar studies
performed subsequently support the genetic hypothesis. 52 The available data
indicate that persistence of high levels of lactase enzyme is probably an
autosomal dominant trait. 52 The persistence in adulthood of the capacity for
lactose digestion in a few human groups is probably a recent evolutionary
development, while the majority of the world's population, and all placental
mammals studied, show a reduction of lactase activity in adulthood. It has
been hypothesized that elevated lactase levels in adults emerged over a span
estimated to be a minimum of 10,000 years in several loci around the world,
probably coincident with the development of dairying. 50
B. LACTASE DEVELOPMENT
In rats and rabbits, the two mammals which so far have been studied in
greatest detail, the specific activity of lactase (expressed in units per mg
protein) exhibits a similar developmental pattern. 53 Lactase activity is un-
Montgomery et al. 33
detectable until a few days before birth; then there is a late gestational rise,
with a peak in specific activity shortly after birth, and a fall during weaning
to the low levels which are seen in adulthood. 53 This pattern is partially a
reflection of the short gestation and immaturity of these mammals. The guinea
pig, which has a gestation of 68 days and is born in a more mature state,
shows detectable lactase activity at 30 days of gestation and a steady increase
until shortly before birth when levels fall slightly. 54 Guinea pigs are also
reported to show very little decrease in lactase levels from the time of birth
to adulthood. 55 In rats, the initial appearance of enzyme activity coincides
with the initial morphogenesis of the enterocyte, which occurs about three
days prior to birth. 53 ·56
In contrast to these patterns, the human intestine begins to synthesize
lactase at approximately the lOth week of gestation, but levels remain low
until 27 to 32 weeks, when enzyme levels increase rapidly. 5 7 In view of the
relatively low lactase levels in infants born prematurely (28 to 32 weeks), it
is of interest that few develop clinical signs of carbohydrate intolerance. It
has been demonstrated 58 - 60 that lactose is malabsorbed in these infants and
reaches the colon where it is salvaged by colonic flora.
In the majority of the world's population who develop "late onset lactase
deficiency'' in mid-childhood, the pattern is similar to that in other mammals;
the late gestational rise is followed by persistence of lactase specific activity
until approximately 5 to 7 years with a fall thereafter to low adult levels. 20
In the Caucasian population, especially those peoples from or derived from
Northern Europe, and certain localized clusters of other racial origins, lactase-
specific activity rises late in gestation and remains at, or slightly below, this
level throughout adult life-in other words, the maintenance of a juvenile
trait (summarized in References 2, 4).
comer and McGill 45 showed that both normal and deficient human adults
displayed this pattern, with the "normal" subjects having markedly elevated
activities along the intestine, except at the most proximal and most distal
points It should be emphasized that in adult humans as well as adult rats and
rabbits, even though lactase activity levels are decreased significantly from
levels found in sucklings, there are still measurable levels of activity. At the
time of weaning, lactase activity is not decreased to zero nor "switched off".
Animal studies indicate that thyroxine is a major regulator of the devel-
opmental pattern of lactase at the time of weaning in rats. 65 Hypophysectomy
and thyroidectomy retard the decrease in lactase activity during the third
postnatal week, while thyroxine replacement restores the normal pattern. 65
Glucocorticoids enhance rat lactase activity in the first weeks of life. 66 Recent
detailed studies in rats demonstrated cooperative effects of thyroxine and
glucocorticoid hormones in modulating the postnatal development of lactase. 67
Malo and Menard68 observed an increase in lactase-specific activity following
injection of epidermal growth factor (EGF); Foltzer-Jourdainne and Raul 69
have recently reported on the effects of EGF on intestinal enzymes. In hy-
pophysectomized animals, the decrease is retarded but still occurs. 65 Studies
of transplanted fetal intestine have shown that the decrease in lactase activity
occures according to the age of the tissue, not of the host. 70 Thus, the un-
derlying pattern appears to be programmed or intrinsic to the tissue, not
dependent on hormonal changes.
There are few data available on hormonal regulation of lactase in human
intestine. Experiments with explants of 12- to 14-week gestation human in-
testine showed that hydrocortisone induced an increase in lactase levels. 71
However, the major increase in human lactase occurs in the last several weeks
of fetal life. A late gestational upsurge in fetal serum cortisol levels has been
described in humans, 72 but whether or not this has a regualtory role in humans
is currently unknown.
Intracellular MVM
ER
cc
195 kDa 205
Nascent High
Mannose
'I' High Mannose & Complex Processed < Active Site
i' Processed High Mannose • 0-Linked
the cell surface enzyme to a 160-kDa form very similar to the mature form
found in normal intestine. The authors feel, however, that because the COS
cells are not epithelial cells, the processing of lactase may be aberrant. A
schematic diagram summarizing the data from studies in the rat on lactase
synthesis and processing is shown in Figure 1.
Studies of intestinal microvillus membrane composition have demon-
strated a change at the time of weaning from high sialic acid to high fucose
content. 98 Rat intestinal MVM glycoproteins undergo sialylation during the
suckling period, while fucosylation begins at weaning, indicating develop-
mental alterations in glycosylation. Concurrent with this change, the enzymes
mediating fucosylation and sialylation also change in activity. 99 Based on
lectin binding studies, Nsi-Emvo et al. 100 proposed that the postweaning de-
cline of lastase activity is due to changes in glycosylation with age, leading
to formation of an unprocessed, inactive precursor. These data remain to be
confirmed.
So far, no other studies have reported an inactive, high molecular weight
precursor form of lactase in adult intestine, although a recent study from
Gray's laboratory describes the accumulation of an inactive 100-kDa pro-
cessed form of the enzyme. 101 On the contrary, lactase immunoprecipitated
with a monoclonal antibody shows comparable protein forms in suckling and
adult animals. 102 When the immunoprecipitated enzyme is analyzed, suckling
and adult forms show identical specific activities as well as Km values. 73
Recent studies of the age-specific changes in glycosylation of rat lactase
38 Nutrition and Gene Expression
indicate that the core structure, consisting of both N-linked and 0-linked
oligosaccharides, remains constant during development, although terminal
sugars shift from predominantly sialic acid during the suckling period to fucose
in adulthood. This alteration in glycosylation of the protein occurs in a dif-
ferent pattern from the postweaning decline in lactase-specific activity. Con-
sequently, age-dependent changes in glycosylation are unlikely to result in
an inactive precursor nor account for the decrease in lactase-specific activity
observed during development. 103
r
oc
r
F.bb
l
F.c
r
~F.·
r; . ~;l;i
D EDDCCCYC
I cm.,IOO AA
H• Conserved AA at active site (residue)
Y• N-Glycosylation site
FIGURE 2. Comparison of derived amino acid sequences from human, rabbit, and rat lactase-
phlorizin hydrolase. Region II begins at approximately residue 360; region III at approximately
882; and region IV at approximately 1370. (Prepared by Dr. 1. Dekker; data from References
93 and 104.)
Studies of chromosome loss in cell hybrids indicate that the gene for human
lactase is located on chromosome 2; 106 there are no data available on the
chromosomal location of rat or rabbit lactase. Hybridization of the human
lactase probe to human DNA digested with a panel of restriction enzymes in
a search for restriction fragment length polymorphisms (RFLP) revealed a
common Mspl polymorphism. This RFLP may provide a marker for the lactase
gene in genetic analysis. 107
It has recently been shown that intestinal isomaltase exhibits sequence
homology with lysosomal alpha-glucosidase, suggesting a common evolu-
tionary origin. ~ A similar relationship might be postulated for lactase and
10
the enzyme structure is the result of duplication of the gene. This provides a
link between a bacterial enzyme and the three mammalian enzymes which
have been analyzed, but does not address the issue of intermediate steps or
how the two may be related.
The major focus of interest in lactase has been the mechanism which
regulates the decrease in lactase activity at weaning. As discussed above, the
Montgomery eta/. 41
most obvious explanation would be that lactase levels depend on the level of
the substrate lactose, so that when lactose decreased in the diet, lactase
decreased also. However, it is now clear that enzyme activity levels do not
depend upon ingested lactose. The available data on lactase structure, func-
tion, size, and glycosylation provide no compelling evidence for changes in
any of these attributes which occur with weaning and account for the decrease
in enzyme-specific activity. On the contrary, the available evidence indicates
that the same protein is present before and after weaning, or in lactase suf-
ficient and deficient humans. Those individuals with low levels of activity
have less lactase protein. Although a number of other mechanisms have been
suggested and investigated, no consensus has been reached and the subject
is still controversial.
Tsuboi eta!. 110 suggested that the decline in the specific activity of lactase
was dependent upon a change in the rate of enterocyte turnover, and that its
synthesis was constant, resulting in a decrease in specific activity. At the
time of weaning, as shown by others and confirmed by Tsuboi et a!., llO cell
migration rates do increase while lactase-specific activity is decreased. How-
ever, a recent study by Yeh showed that corticoid injection induced both an
increase in cell migration and an increase in lactase levels, not a decrease,
in suckling rats, 111 suggesting that a change in enterocyte migration rate is
not sufficient to account for the decrease in lactase activity. In contrast, Jonas
et al. 112 found that lactase synthesis fell in parallel with the decline seen in
lactase-specific activity during development, indicating a decrease in lactase
synthesis as a mechanism. Smith and James 113 suggested that a two-stage
process was involved: early in the postnatal period, the fall in the specific
activity of lactase was due to increased cell turnover, and later was dependent
upon a decreased rate of synthesis. Another mechanism which has received
little attention is increased degradation of lactase. Studies of dietary effects
on adult lactase levels suggest that these may occur at least partially through
modifications of lactase degradation. 114 It is known that the level of luminal
proteases, which modulate lactase turnover, increases with weaning. 115 How-
ever, there is currently no direct evidence of increased turnover in the decline
in lactase-specific activity.
The studies described all measured lactase-specific activity, which may
lead to deceptive conclusions. The studies in our laboratory 112 indicated sub-
stantial synthesis of lactase even in adult rats, suggesting that the whole
concept of lactase decline at weaning needed reexamination. In fact, our
recent work 73 shows that total lactase activity remains nearly constant from
day 12 to adulthood during development of rat intestine, although the specific
activity decreases over time. This is due to a substantial increase in total
cellular protein and a probable reduction in the number of lactase enzyme
molecules per cell. These findings are in agreement with studies by Ekstrom
et a!., 116 demonstrating significant total lactase activity in pigs past the time
of weaning, as well as observations by Moog of significant postweaning
42 Nutrition and Gene Expression
levels of lactase in mice. 117 This persistence of lactase activity into adulthood
challenges previous concepts of the regulation of the enzyme and alters the
interpretation of previous data in the literature suggesting a marked loss of
enzyme activity during maturation. It also implies that this enzyme might
maintain important function after the weaning period.
Lactase protein levels reflect the synthetic rate of the protein, which in
tum is dependent upon the steady-state level of mRNA. There is presently
no convincing evidence of a significant role for posttranslational mechanisms,
such as modifications of glycosylation, as an overall mechanism for the
developmental pattern of lactase activity. Increased degradation may play a
major role, but there is currently little supportive evidence for such a step.
As described below, the primary regulation of lactase levels in the small
intestine is probably transcriptional, as has been found for most other mam-
malian proteins. 118
B Total Activity
~ mRNA
?d 14d 21d AM LF
FIGURE 3. Coordinate changes in total lactase mRNA and total lactase enzyme activity during
development of rat intestine. The time shown along the horizontal axis refers to postnatal age
in days. AM refers to adult males; LF refers to lactating females. The increased lactase activity
seen in LF is probably a posttranscriptional event. (Data from Reference 105.)
TABLE 3
Distribution of Sucrase/Lactase Activity
Ratios and Presence of Lactase mRNA
( +) in Human Intestinal Biopsy Samples
in Three Racial Groups
(Number of subjects)
Data Orientals Blacks White
Ratio> 2 12 4 6
Ratio< 2 0 2 7
Total 12 6 13
Lactase mRNA ~ 12 4 6
and ratio> 2
Lactase mRNA + 0 2 7
and ratio< 2
VIII. CONCLUSIONS
Although the literature still frequently refers to "lactose intolerance" and
"lactase deficiency" as if these were abnormalities, it is clear that low levels
Montgomery et al. 45
23456789 10 H 12 B 14 15
Lactose mRNA
Actin mRNA
60 r--------------------------------.
6
LACTASE/ACTIN mRNA
FIGURE 4. Comparison of lactase activity and mRNA levels in biopsies from human intestine.
(A) The Northern blot shows results from 5 Oriental, 3 black, and 7 white patients. Four 11-g of
total human intestinal RNA were prepared and hybridized to an appropriately labeled human
eDNA probe (h-LPH-cDNA-1 ). 125 The blots at the bottom of the figure represent hybridization
to a human 13-actin probe. (B) Lactase mRNA content/actin mRNA content were obtained by
densitometry and a ratio calculated; this was compared to lactase specific activity for each patient:
(•) Oriental patients, (6) black patients, (O) white patients. (Data from Reference 125; reprinted
with permission of the Rockefeller University Press, New York.)
46 Nutrition and Gene Expression
of lactase, and the associated intolerance for significant intake of lactose, are
the normal condition for most of the world's human population. An extensive
literature has correlated the occurrence of lactose intolerance with decreased
levels of lactase in the small intestine. However, it is worth reiterating that,
with the exception of frank disease states, humans (as well as most mammals
examined) have measurable levels of lactase in their small intestine. As dis-
cussed above, "lactase" may actually have other functions in the adult in-
testine, explaining why it is still present. In this sense, rats and rabbits are
useful models for studying the decline in lactase levels in humans. The cur-
rently available evidence suggests that the decreased expression of lactase
protein is a reflection of decreased expression of lactase mRNA.
The overall pattern in the human population thus appears similar to that
described in rats, where enzyme levels and mRNA levels are coordinate in
development. 105 Elevated lactase levels in specific population groups, which
have been correlated with the presence of an autosomal dominant gene, appear
due to an elevated transcription of lactase mRNA. The molecular control of
this elevated expression remains to be elucidated.
The regulation of the decline in lactase levels is probably a complex
combination of factors, including decreased mRNA transcription, the resulting
decreased enzyme synthesis, and increased enzyme degradation. The unique
and more interesting question is what mechanism controls the persistence in
some human groups of high juvenile levels of lactase into adulthood.
IX. SUMMARY
The small intestinal enzyme lactase-phlorizin hydrolase, which hydrolyzes
lactose, the sole carbohydrate in milk, plays a critical role in the nutrition of
the mammalian neonate. Milk is also an important source of nutrients, es-
pecially calcium, for many adults, particularly those of Northern Europe and
North America. Lactose intolerance, which can preclude significant milk
intake, is common in adult humans, usually due to low levels of small intestinal
lactase. Low lactase levels result from either intestinal injury, or, in the
majority of the world's adult population, alterations in the genetic expression
of lactase. Although the mechanism of decreased lactase levels has been the
subject of intensive investigation, no concensus has yet emerged. Recent
studies have begun to define the cellular and molecular biology of this enzyme.
In animals and humans, a glycosylated precursor is proteolytically cleaved
to yield the mature enzyme on the microvillus membrane of the enterocyte,
bound to the lipid bilayer only by a hydrophobic anchor sequence. The enzyme
hydrolyzes lactose, phlorizin, and glycosylceramides. A decline in lactase-
specific activity occurs at the time of weaning in most mammalian species;
in the majority of humans, who have low lactase activity as adults, the decline
Montgomery eta!. 47
ACKNOWLEDGMENTS
Portions of this chapter are reprinted from the FASEB Journal with per-
mission of the Federation of American Societies for Experimental Biology,
Rockville, MD 20852.
Work in the authors' laboratories has been supported by National Institutes
of Health research grant DK-32658; a grant from the Whitaker Health Sciences
Fund; grant P30 DK 34928 to the Center for Gastroenterology Research on
Absorptive and Secretory Processes, New England Medical Center Hospitals;
a grant from Nutricia, Zoetermeer, The Netherlands; and a NATO collabo-
rative research grant.
REFERENCES
I. Paige, D. M. and Bayless, T. M., Eds., Lactose Digestion, The Johns Hopkins Uni-
versity Press, Baltimore, 1981.
2. Scrimshaw, N. S. and Murray, E. B., The acceptability of milk and milk products in
populations with a high prevalence of lactose intolerance, Am. J. Clin. Nutr., 48, 1083,
1988.
3. Johnson, J, D., Kretchmer, N., and Simoons, F. J,, Lactose malabsorption: its biology
and history, in Advances in Pediatrics, Vol 21, Schulman, 1., Ed., Year Book Medical
Publishers, Chicago, 197, 1974.
4. Flatz, G., Genetics of lactose digestion in humans, in Advances in Human Genetics,
Harris, H. and Hirschhorn, K., Eds., Plenum Press, New York, I, 1987.
5. Jacobi, A., Milk sugar in infant feeding, Trans. Am. Pediatr. Soc., 13, 150, 1901.
6. Dahlqvist, A., Method for assay of intestinal disaccharidases, Anal. Biochem., 7, 18,
1964.
7. Dahlqvist, A., Hammond, J, B., Crane, R. K., Dunphy, J, V., and Littman, A.,
Intestinal lactase deficiency and lactose intolerance in adults, Gastroenterology, 45, 488,
1963.
8. Cuatrecasas, P., Lockwood, D. H., and Caldwell, J, R., Lactase deficiency in the
adult, a common occurrence, Lancet, I, 14, 1965.
9. Bayless, T. M. and Rosensweig, N. S., A racial difference in the incidence of lactase
deficiency: a survey of milk intolerance and lactase deficiency in healthy males, lAMA,
197, 968, 1966.
10. Savilahti, E., Launiala, K., and Kuitunen, P., Congenital lactase deficiency: a clinical
study on 16 patients, Arch. Dis. Child, 58, 246, 1983.
11. Mendel, L. B. and Mitchell, P. H., Chemical studies on growth: the inverting enzymes
of the alimentary tract, especially in the embryo, Am. J. Physiol., 20, 81, 1907.
48 Nutrition and Gene Expression
12. Plimmer, R. H. A., On the presence of lactase in the intestine of animals and on the
adaptation of the intestine to lactose, J. Physiol. (London). 35, 81, 1906.
13. Gilat, T., Russo, S., Gelman-Malachi, E., and Aldor, T. A. M., Lactase in man, a
non-adapwble enzyme, Gastroenterology, 62, 1125, 1972.
14. Kogut, M.D., Donnell, G. N., and Shaw, K. N. F., Studies of lactose absorption in
patients with galactosemia, J. Pediatr., 71, 75, 1967.
15. Palmiter, R. D., What regulates lactose content in milk, Nature (London), 221, 912,
1969.
16. Kretchmer, N. and Sunshine, P., Intestinal disaccharidase deficiency in the sea lion,
Gastroenterology, 53, 123, 1967.
17. Crisp, E. A., Czolij, R., and Messer, M., Absence of beta-galactosidase (lactase)
activity from intestinal brush borders of suckling macropods: implications for mechanism
of lactose absorption, Comp. Biochem. Physiol., 88B, 923, 1987.
18. Jenness, R., Regehr, E. A., and Sloan, R. E., Comparative biochemical studies of
milks. II. Dialyzable carbohydrates, Comp. Biochem. Physiol., 13, 339, 1964.
19. Newcomer, A. D., McGill, D. B., Thomas, P. J,, and Hofmann, A. F., Prospective
comparison of indirect methods for detecting lactase deficiency, N. Engl. J. Med., 293,
1232, 1975.
20. Welsh, J.D., Poley, J. R., Bhatia, M., and Stevenson, D. E., Intestinal disaccharidase
activities in relation to age, race, and mucosal damage, Gastroenterology, 75, 847, 1978.
21. Mobassaleh, M., Montgomery, R. K., Biller, J, A., and Grand, R. J,, Development
of carbohydrate absorption in the fetus and neonate, Pediatrics, 75, 160, 1985.
22. Martini, M. C. and Savaiano, D. A., Reduced intolerance symptoms from lactose
consumed during a meal, Am. J. Clin. Nutr., 47, 57, 1988.
23. Bond, J, H. and Levitt, M.D., Investigation of small bowel transit time in man utilizing
pulmonary hydrogen (H 2 ) measurements, J. Lab. Clin. Med., 85, 546, 1975.
24. Levitt, M. D., Production and excretion of hydrogen gas in man, N. Engl. J. Med.,
281, 122, 1969.
25. Hyams, J, S., Stafford, R. J,, Grand, R. J., and Watkins, J, B., Correlation of
lactose breath hydrogen test, intestinal morphology, and lactase activity in young children,
J. Pediatr., 97, 609, 1980.
26. Newcomer, A. D., Screening tests for carbohydrate malabsorption, J. Pediatr. Gas-
troentero/. Nutr., 3, 6, 1984.
27. Barr, R. G., Perman, J, A., Schoeller, D. A., and Watkins, J, B., Breath test in
pediatric gastrointestinal disorders: new diagnostic opportunities, Pediatrics, 62, 393,
1978.
28. Douwes, A. C., Fernandes, J,, and Degenhardt, H. J,, Improved accuracy of lactose
tolerance test in children, using expired H2 measurement, Arch. Dis. Child., 53, 939,
1978.
29. Barr, R. G., Levine, M.D., and Watkins, J, B., Recurrent abdominal pain of childhood
due to lactose intolerance: a prospective study, N. Engl. J. Med., 300, 1449, 1979.
30. Ostrander, C. R., Cohen, R. S., Hopper, A. 0., and Stevenson, D. K., Breath
hydrogen analysis: a review of the methodologies and clinical applications, J. Pediatr.
Gastroenterol. Nutr., 2, 525, 1983.
31. Perman, J, A., Clinical application of breath hydrogen measurements, Can. J. Physiol.
Pharmacal., 69, Ill, 1991.
32. Queen, P. M. and Henry, R. R., Growth and nutrient requirements of children, in
Pediatric Nutrition: Theory and Practice, Grand, R. J., Sutphen, J. L., and Dietz, W. H.,
Eds., Butterworths, Boston, 1987, 341.
33. Rosado, J, L., Solomons, N. W., Lisker, R., and Bourges, H., Enzyme replacement
therapy for primary adult lactase deficiency, Gastroenterology, 87, 1072, 1984.
34. Newcomer, A. D. and McGill, D. B., Clinical importance of lactase deficiency, N.
Engl. J. Med., 310, 42, 1984.
Montgomery et al. 49
35. Bayless, T. M., Rothfeld, B., Massa, C., Wise, L., Paige, D. M., and Bedine, M.S.,
Lactose and milk intolerance: clinical implications, N. Engl. J. Med., 292, 1156, 1975.
36. Stephenson, L. S. and Latham, M. C., Lactose intolerance and milk consumption: the
relation of tolerance to symptoms, Am. J. Clin. Nutr., 27, 296, 1974.
37. Lisker, R., Aguilar, L., and Zavala, C., Intestinal lactase deficiency and milk drinking
capacity in the adult, Am. J. Clin. Nutr., 31, 1499, 1978.
38. Kocian, J., Skala, I., and Bakos, K., Calcium absorption from milk and lactose-free
milk in healthy subjects and patients with lactose intolerance, Digestion, 9, 317, 1973.
39. Griessen, M., Cochet, B., Infante, F., Jung, A., Bartholdi, P., Donath, A., Loizeau,
E., and Courvoisier, B., Calcium absorption from milk in lactase-deficient subjects,
Am. J. Clin. Nutr., 49, 377, 1989.
40. Cavalli-Sforza, L. T., Strata, A., Barone, A., and Cucurachi, L., Primary adult
lactose malabsorption in Italy: regional differences in prevalence and relationship to
lactose intolerance and milk consumption, Am. J. Clin. Nutr., 45, 748, 1987.
41. Kretchmer, N., Memorial lecture: lactose and lactase-a historical perspective, Gas-
troenterology, 61, 805, 1971.
42. Biiller, H. A. and Grand, R. J,, Lactose intolerance. Annu. Rev. Med., 41, 141, 1990.
43. Bolin, T. D. and Davis, A. E., Primary lactase deficiency: genetic or acquired?, Am.
J. Dig. Dis., 15, 679, 1970.
44. Rosensweig, N. S., Adult lactase deficiency: genetic control or adaptive response?.
Gastroenterology, 60, 464, 1971.
45. Newcomer A. D. and McGill, D. B., Distribution of disaccharidase activity in the small
bowel of normal and lactase-deficient subjects, Gastroenterology, 51, 481, 1966.
46. Welsh, J. D., Isolated lactase deficiency in humans: report on 100 patients, Medicine,
49, 257, 1970.
47. Bolin, T. D., Pirola, R. C., and Davis, A. E., Adaptation of intestinal lactase in the
rat. Gastroenterology, 60, 432, 1969.
48. Lebenthal, E., Sunshine, P., and Kretchmer, N., Effect of prolonged nursing on the
activity of intestinal lactase in rats, Gastroenterology, 64, 1136, 1973.
49. Bond, J, H. and Levitt, M. D., Quantitative measurement of lactose absorption, Gas-
troenterology, 70, 1058, 1976.
50. Simoons, F. J., The geographic hypothesis and lactose malabsorption, Dig. Dis. Sci.,
23, 963, 1978.
51. Sahi, T., Isokoski, M., Jussila, J., Launiala, K., and Pyoriilii, K., Recessive inher-
itance and adult type-lactose malabsorption, Lancet, 2, 823, 1973.
52. Sahi, T. and Launiala, K., More evidence for the recessive inheritance of selective
adult type lactose malabsorption, Gastroenterology, 73, 231, 1977.
53. Doell, R. G. and Kretchmer, N., Studies of small intestine during development. I.
Distribution and activity of I)-galactosidase, Biochim. Biophys. Acta., 62, 353, 1962.
54. Bailey, D. S., Cook, A., McAllister, G., Moss, M., and Mian, N., Structural and
biochemical differentiation of the mammalian small intestine during fetal development,
J. Cell Sci.. 72, 195, 1984.
55. Blaxter, K. L., Lactation and the growth of the young. in Milk: The Mammary Gland
and Its Secretion, Vol. II, Kon, S. K. and Cowie, A. T., Eds., Academic Press, New
York. 1961. 305.
56. Montgomery, R. K., Kothe, M. J. C., Biiller, H. A., and Grand, R. J., Rat lactase
mRNA appears concurrently with development of columnar epithelial cells in fetal in-
testine, Gastroenterology, 98, A423(Abstr.). 1990.
57. Antonowicz, I., Chang, S. K., and Grand, R. J,, Development and distribution of
lysosomal enzymes and disaccharidases in human fetal intestine. Gastroenterology, 67,
51, 1974.
50 Nutrition and Gene Expression
58. MacLean, W. C. and Fink, B. B., Lactose malabsorption by premature infants: mag-
nitude and clinical significance, J. Pediatr., 97, 383, 1980.
59. Kien, C. L., Liechty, E. A., Myerberg, D. Z., and Mullett, M. D., Dietary carbo-
hydrate assimilation in the premature infant: evidence for a nutritionally significant bac-
terial ecosystem in the colon, Am. J. Clin. Nutr., 46, 456, 1987.
60. Murray, R. D., Boutton, T. W., Klein, P. D., Gilbert, M., Paule, C. L., and
MacLean, W. C., Comparative absorption of [13C] glucose and [13C] lactose by pre-
mature infants, Am. J. Clin. Nutr., 51, 59, 1990.
61. Miller, D. and Crane, R. K., Digestive function of the epithelium of the small intestine.
II. Localization of disaccharide hydrolysis in the isolated brush border portion of intestinal
epithelial cells, Biochim. Biophys. Acta, 52, 293, 1961.
62. Nordstrom, C., Dahlqvist, A., and Josefsson, L., Quantitative determination of en-
zymes in different parts of the villi and crypts of rat small intestine, J. Histochem.
Cytochem., 15, 713, 1968.
63. Gordon, J. I., Intestinal epithelial differentiation: new insights from chimeric and trans-
genic mice, J. Cell Bioi., 108, 1187, 1989.
64. Auricchio, S., Rubino, A., and Murset, G., Intestinal glycosidase activites in the
human embryo, fetus and newborn, Pediatrics, 35, 944, 1965.
65. Yeh, K. Y. and Moog, F., Intestinal lactase activity in the suckling rat: influences of
hypophysectomy and thyroidectomy, Science, 183, 77, 1974.
66. Koldovsky, 0. and Sunshine, P., Effect of cortisone on developmental pattern of the
neutral and acid beta-galactosidase of the small intestine of the rat, Biochem. J., 117,
467, 1970.
67. Yeh, K. Y., Yeh, M., and Holt, P.R., Thyroxine and cortisone cooperate to modulate
postnatal intestinal enzyme differentiation in the rat, Am. J. Physiol., 260, G371, 1991.
68. Malo, C. and Menard, D., Influence of epidermal growth factor on the development
of suckling mouse intestinal mucosa, Gastroenterology, 83, 28, 1982.
69. Foltzer-Jourdainne, C. and Raul, F., Effect of epidermal growth factor on the expres-
sion of digestive hydrolases in the jejunum and colon of newborn rats, Endocrinology,
127, 1763, 1990.
70. Montgomery, R. K., Sybicki, M. A., and Grand, R. J., Autonomous biochemical
and morphological differentiation in fetal rat intestine transplanted at 17 and 20 days of
gestation, Dev. Bioi., 87, 76, 1981.
71. Arsenault, P. and Menard, D., Influence of hydrocortisone on human fetal small
intestine in organ culture, J. Pediatr. Gastroenterol. Nutr., 4, 893, 1985.
72. Murphy, B. E. P., Human fetal serum cortisol levels related to gestational age: evidence
of midgestational fall and a steep late gestational rise, independent of sex or mode of
delivery, Am. J. Obstet. Gynecol., 144, 276, 1982.
73. Buller, H. A., Wassenaer, A. G. van, Raghavan, S., Montgomery, R. K., Sybicki,
M.A., and Grand, R. J., New insights into the lactase and glycosylceramidase activities
of rat microvillus membrane lactase-phlorizin hydrolase, Am. J. Physiol., 257, G616,
1989.
74. Asp, N. G. and Dahlqvist, A., Human small intestine 13-galactosidases: specific assay
of three different enzymes, Anal. Biochem., 47, 527, 1972.
75. Gray, G. M. and Santiago, N. A., Intestinall3-galactosidases. I. Separation and char-
acterization of 3 enzymes in normal human intestine, J. Clin. Invest., 48, 716, 1969.
76. Brady, R. 0., Gal, A. E., Kanfer, J. N., and Bradley, R. M., The metabolism of
glucocerebrosides, III, Purification and properties of glucosyl- and galactosylceramide
cleaving enzyme from rat intestinal tissue, J. Bioi. Chern., 240, 3766, 1965.
77. Kobayashi, T. and Suzuki, K., The glycosylceramidase in the murine intestine, J. Bioi.
Chern., 256, 7768, 1981.
Montgomery et at. 51
78. Leese, H. J. and Semenza, G., On the identity between the small intestinal enzymes
phlorizin hydrolase and glycosylceramidase, J. Biol.Chern .. 248, 8170, 1973.
79. Birkenmeier, E. and Alpers, D. H., Enzymatic properties of rat lactase-phlorizin hy-
drolase, Biochirn. Biophys. Acta, 350, 100, 1974.
80. Haringsma, J., Biiller, H. A., Matsudaira, P. T., Montgomery, R. K., and Grand,
R. J., Human lactase-phlorizin hydrolase, function and active sites, Eur. J. Clin.Invest.,
20 A26 (Abstr.), 1990.
81. Legler, G., Untersuchungen zum wirkungsmechanismus glycosidspaltender enzyme. III.
Markierung des aktiven zentrums einer beta-glucosidases aus aspergillus wentii mit con-
duritol-b-epoxid, Z. Physiol. Chern .. 349, 767, 1968.
82. Dinur, T., Osiecki, K. M., Legler, G., Gatt, S., Desnick, R. J,, and Grabowski,
G. A., Human acid 13-glucosidase: isolation and amino acid sequence of a peptide con-
taining the catalytic site, Proc. Nat!. Acad. Sci. U.S.A.. 83, 1660, 1986.
83. Quaroni, A. and Semenza, G., Partial amino acid sequences around the essential car-
boxylate in the active sites of the sucrase-isomaltase complex, J. Bioi. Chern .• 251, 3250,
1976.
84. Rousset, M., Trugnan, G., Brun, J, L., and Zweibaum, A., Inhibition of the post-
translational processing of microvillar hydrolases is associated with a specific decrease
of sucrase-isomaltase and an increased turnover of glucose in CaCo-2 cells treated with
monensin, FEES Lett.. 208, 34, 1986.
85. Danielsen, E. M. and Cowell, G. M., Biosynthesis of intestinal microvillar proteins:
processing of N-linked carbohydrate is not required for surface expression, Biochern. J ..
240, 777. 1986.
86. Biiller, H. A., Montgomery, R. K., Sasak, W. V., and Grand, R. J., Biosynthesis,
glycosylation, and intracellular transport of intestinal lactase-phlorizin hydrolase in rat,
J. Bioi. Chern .. 262, 17206, 1987.
87. Biiller, H. A., Rings, E. H. H. M., Montgomery, R. K., Sasak, W. V., and Grand,
R. J,, Further studies of glycosylation and intracellular transport of lactase-phlorizin
hydrolase in rat small intestine, Biochern. J., 263, 249, 1989.
88. Hubbard, C. S. and Ivatt, R. J,, Asparagine-linked oligosaccharides, Annu. Rev.
Biochern .. 50, 555, 1981.
89. Hauri, H. P., Sterchi, E. E., Bienz, D., Fransen, J, A.M., and Marxer, A., Expres-
sion and intracellular transport of microvillus membrane hydrolases in human intestine,
J. Cell Bioi.• 101, 838, 1985.
90. Nairn, H. Y., Sterchi, E. E., and Lentze, M. J,, Biosynthesis and maturation of lactase-
phlorizin hydrolase in the human small intestinal epithelial cells, Biochern. J.. 241,
427, 1987.
91. Skovbjerg, H., Danielsen, E. M., Noren, 0., and Sjostrom, H., Evidence for bio-
synthesis of lactase-phlorizin hydrolase as a single-chain high molecular weight precursor,
Biochirn. Biophys. Acta, 798, 247, 1984.
92. Danielsen, E. M., Skovbjerg, H., Noren, 0., and Sjostrom, H., Biosynthesis of
intestinal microvillar proteins, intracellular processing of lactase-phlorizin hydrolase,
Biochern. Biophys. Res. Cornrn .• 122, 82, 1984.
93. Mantei, N., Villa, M., Enzler, T., Wacker, H., Boll, W., James, P., Hunziker, W.,
and Semenza, G., Complete primary structure of human and rabbit lactase-phlorizin
hydrolase: implications for biosynthesis, membrane anchoring and evolution of the en-
zyme, EMBO J., 7, 2705, 1988.
94. Hauri, H. P., Quaroni, A., and Isselbacher, K. J,, Biogenesis of intestinal plasma
membrane: posttranslational route and cleavage of sucrase-isomaltase, Proc. Nat!. Acad.
Sci. U.S.A., 76, 5183, 1979.
52 Nutrition and Gene Expression
95. Montgomery, R. K., Sybicki, M.A., Forcier, A. G., and Grand, R. J., Rat intestinal
microvillus membrane sucrase-isomaltase is a single high molecular weight protein and
fully active enzyme in the absence of luminal factors, Biochirn. Biophys. Acta, 661, 346,
1981.
96. Yeh, K., Yeh, M., Pan, P., and Holt, P. R., Post-translational cleavage of rat intestinal
lactase occurs at the luminal side of the brush border membrane, Gastroenterology, 1OJ,
312, 1991.
97. Nairn, H. Y., Lacey, S. W., Sambrook, j. F., and Gething, M.-J., Expression of a
full-length eDNA coding for human intestinal lactase-phlorizin hydrolase reveals an un-
cleaved, enzymatically active, and transport-competent protein, J. Bioi. Chern., 266,
12313, 1991.
98. Srivastava, 0. P., Steele, M. I., and Torres-Pinedo, R., Maturational changes in
terminal glycosylation of small intestinal microvillar proteins in the rat, Biochirn. Biophys.
Acta, 914, 143, 1987.
99. Chu, S. W. and Walker, W. A., Developmental changes in the activities of sialyl- and
fucosyltransferases in rat small intestine, Biochirn. Biophys. Acta, 883, 496, 1986.
100. Nsi-emvo, E., Launay, J, F., and Raul, F., Is adult-type hypolactasia in the intestine
of mammals related to changes in the intracellular processing of lactase, Cell Mol. Biol.,
33, 335, 1987.
101. Quan, R., Santiago, N. A., Tsuboi, K. K., and Gray, G. M., Intestinal lactase, shift
in intracellular processing to altered, inactive species in the adult rat, J. Bioi. Chern.,
265, 15882, 1990.
102. Quaroni, A., Pre- and postnatal development of differentiated functions in rat intestinal
epithelial cells, Dev. Bioi., Ill, 280, 1985.
103. Biiller, H. A., Rings, H. H. M., Pajkrt, D., Montgomery, R. K., and Grand, R. j.,
Glycosylation of lactase-phlorizin hydrolase in rat small intestine during development,
Gastroenterology, 98, 667, 1990.
104. Duluc, 1., Boukamel, R., Mantei, N., Semenza, G., Raul, F., and Freund, J.,
Sequence of the precursor of intestinal lactase-phlorizin hydrolase from fetal rat, Gene,
103, 275, 1991.
105. Biiller, H. A., Kothe, M. J, C., Goldman, D. A., Grubman, S. A., Sasak, W. V.,
Matsudaira, P. T., Montgomery, R. K., and Grand, R. J., Coordinate expression of
lactase-phlorizin hydrolase mRNA and enzyme levels during development, J. Bioi. Chern.,
265, 6978, 1990.
106. Kruse, T. A., Bolund, L., Grzeschik, K. H., Ropers, H. H., Sjostrom, H., Noren,
0., Mantei, N., and Semenza, G., The human lactase-phlorizin hydrolase gene is located
on chromosome 2, FEBS Lett., 240, 123, 1988.
107. Kruse, T. A., Bolund, L., Byskov, A., Sjostrom, H., Noren, 0., Mantei, N., and
Semenza, G., Mapping of the human lactase-phlorizin hydrolase gene to chromosome
2, Cytogenet. Cell Genet., 51, 1026, (Abstr.), 1989.
108. Hoefsloot, L. H., Hoogeveen-Westerveld, M., Kroos, M. A., Beeumen, J, van,
Reuser, A. J. J., and Oostra, B. A., Primary structure and processing of lysosomal
alpha-glucosidase; homology with the intestinal sucrase-isomaltase complex, EMBO J.,
7, 1697, 1988.
109. Oshima, A., Tsuji, A., Nagao, Y., Sakuraba, H., and Suzuki, Y., Cloning, sequenc-
ing, and expression of eDNA for human beta-galactosidase, Biochem. Biophys. Res.
Comm., 157, 238, 1988.
110. Tsuboi, K. K., Kwong, L. K., D'Harlingue, A. E., Stevenson, D. K., Kerner, J, A.,
and Sunshine, P., The nature of maturational decline of intestinal lactase activity, Biochim
Biophys. Acta, 840, 69, 1985.
Ill. Yeh, K., Yeh, M., and Holt, P. M., Intestinal lactase expression and epithelial cell
transit in hormone-treated suckling rats, Am. J. Physiol., 260, 0379, 1991.
Montgomery eta/. 53
112. Jonas, M. M., Montgomery, R. K., and Grand, R. J., Intestinal lactase synthesis
during postnatal development in the rat, Pediatr. Res., 19, 956, 1985.
113. Smith, M. W. and James, P. S., Cellular origin of lactase decline in post weaned rats,
Biochim. Biophys. Acta, 905, 503, 1987.
114. Goda, T., Bustamante, S., and Koldovsky, 0., Dietary regulation of intestinal lactase
and sucrase in adult rats: quantitative comparison of effect of lactose and sucrose, f.
Pediatr. Gastroenterol. Nutr., 4, 998, 1985.
115. Seetharam, B., Yeh, K. Y., and Alpers, D. H., Turnover of intestinal brush border
proteins during postnatal development in rat, Am. f. Physiol., 239, 0524, 1980.
116. Ekstrom, K. E., Benevenga, N.J., and Grummer, R. H., Changes in the intestinal
lactase activity in the small intestine of two breeds of swine from birth to 6 weeks of
age, J. Nutr., 105, 1032, 1975.
117. Moog, F., Denes, A. E., and Powell, P.M., Disaccharidases in the small intestine of
the mouse: normal development and influence of cortisone, actinomycin D, and cyclo-
heximide, Dev. Bioi., 35, 143, 1973.
118. Johnson, P. F. and McKnight, S. L., Eukaryotic transcriptional regulatory proteins,
Anna. Rev. Biochem., 58, 799, 1989.
I 19. Sebastio, G., Villa, M., Sartorio, R., Guzzetta, V., Poggi, V., Auricchio, S., Boll,
W., Mantei, N., and Semenza, G., Control of lactase in human adult-type hypolactasia
and in weaning rabbits and rats, Am. J. Hum. Genet., 45, 489, 1989.
120. Freund, J. N., Duluc, I., and Raul, F., Lactase expression is controlled differently in
the jejunum and ileum during development in rats, Gastroenterology, 100, 388, 1991.
121. Sterchi, E. E., Mills, P.R., Fransen, J. A.M., Hauri, H. P., Lentze, M. J., Nairn,
H. Y., Ginsel, L., and Bond, J., Biogenesis of intestinal lactase-phlorizin hydrolase in
adults with lactose intolerance, J. Clin. Invest., 86, 1329, 1990.
122. Witte, J., Lloyd, M., Lorenzsonn, V., Korsmo, H., and Olsen, W., The biosynthetic
basis of adult lactase deficiency, J. Clin. Invest., 86, 1338, 1990.
123. Maiuri, L., Raia, V., Potter, J., Swallow, D., Ho, M. W., Fiocca, R., Finzi, G.,
Cornaggia, M., Capella, C., Quaroni, A., and Auricchio, S., Mosaic pattern oflactase
expression by villous enterocytes in human adult-type hypolactasia, Gastroenterology,
100, 359,1991.
124. Schmidt, G. H., Wilkinson, M. M., and Ponder, B. A. J., Cell migration pathway
in the intestinal epithelium: an in situ marker system using mouse aggregation chimeras,
Cell, 40, 425, 1985.
125. Escher, J. C., de Koning, N. D., Engen, C. G. J. van, Arora, S., Biiller, H. A.,
Montgomery, R. K., and Grand, R. J., Molecular basis of lactase levels in adult
humans, J. Clin. Invest., 89, 480, 1992.
126. Lloyd, M., Mevissen, G., Fischer, M., Olsen, W., Goodspeed, D., Genini, M., Boll,
W., Semenza, G., and Mantei, N., Regulation of intestinal lactase in adult hypolactasia,
J. Clin. Invest., 89, 524, 1992.
127. Biiller, H. A., unpublished data.
Chapter 3
TABLE OF CONTENTS
I. Introduction ....................................................... 56
0-8493-6%1-41931$0.00 +$.50
© 1993 by CRC Press, Inc. 55
56 Nutrition and Gene Expression
References ................................................................ 83
I. INTRODUCTION
A. PREDUODENAL LIPASES
As early as in 1946, Schonheyder and colleagues demonstrated that the
human stomach was able to digest dietary lipids and reported the presence of
a lipase enzyme in gastric juice. 3 Preduodenallipases have been detected in
most animal species and are widely distributed. Their synthesis and secretion
differ from one species to another. Depending on the species considered,
preduodenallipase is almost entirely produced by the tongue as in the rat and
mouse, 4 •5 pharynx as in the calf and the sheep, 6 ·7 or stomach as in the man,
rabbit, dog, hog, horse, cat, guinea pig, baboon, and macaque. 3 •8 - 11
In the rat, lingual lipase is located in the acinar cells of serous von Ebner
glands and in the lingual mucous gland. 12 ·13 By means of specific polyclonal
antibodies to human and rabbit gastric lipase, human gastric lipase has been
demonstrated to be exclusively located in the chief cells of the fundic mucosa
of the human stomach, 14 - 16 whereas the cells producing rabbit gastric lipase
are restricted in the upper base of the gastric fundic gland. 10 • 17 Lipase activity
in human gastric content is 15 to 75 U/mP 8 or 4700 U/g of fresh mucosa in
men under 50 years. 15 Gastric lipolytic activity decreases in aged persons, to
reach 700 U/g of fresh mucosa in subjects over 60 years of age. 15
Most of the biochemical studies have been performed with human and
rabbit gastric lipases. Preduodenal lipases are acid enzymes (the pH optima
for preduodenal lipases generally fall in the range of 2 to 62 ), which may
work in extreme pH conditions. They can act in the stomach, where the pH
is 4.5 to 5.5 after a meal intake, to initiate the hydrolysis and digestion of
dietary fat. Human gastric lipase does not hydrolyze a tributyrin emulsion in
TABLE 1
Classification of Digestive (Phospho)lipases IJl
01:1
Tongue (Von Ebner glands) Exocrine pancreas (acinar cells) Human milk Exocrine pancreas (acinar cells)
Pharynx Exocrine pancreas (acinar cells) Intestine (Paneth cells)
Stomach (chief cells)
Other Names
Lingual lipase Pancreatic lipase (EC 3.1.1.3) Bile salt stimulated lipase (BSSL) Phospholipase A2 (EC 3 .I. 1.4)
Salivary lipase Carboxylic ester hydrolase (CEH)
Pharyngeal lipase Cholesterol esterase (CE) =<:
~
Gastric lipase Carboxyl ester lipase (CEL) ~·
Pregastric lipase Monoglyceride lipase c;·
;::
Nonpancreatic lipase Nonspecific lipase
>:l
Preduodenal lipase Lysophospholipase ;:::
l:l...
CJ
Substrate Specificity ~
~
sn-3 Position of glycerides sn-l And sn- 3 positions of Cholesterol esters sn-2 Position of L-o.-glycero- ~
(also sn-l and sn-2 positions glycerides Fat-soluble vitamin esters phospholipids ~
Glycerides ~
of glycerides)
Glycerophospholipids
"'"'
c·
;:::
Primary, secondary, and tertiary
carboxylic esters
Adapted from Reference 2.
Wicker-Planquart and Puigserver 59
the absence of bile salts or alimentary proteins. The presence of most of the
bile salts (taurocholate, glycocholate, taurodeoxycholate, glycodeoxycholate,
or bile salts mixture), which must be added prior to the enzyme in order to
prevent irreversible lipase denaturation, activates human gastric lipase on the
tributyrin substrate. 2 This can be explained by the fact that human gastric
lipase activity is restricted to a triacylglycerol!water interfacial tension range
of 8 to 13 dyn!cm. Interfacial tension at tributyrin/water interface is 15 dyn!cm,
which rapidly inactivates human gastric lipase. The fact that the enzyme has
a single disulfide bridge 19 can explain the rapid interfacial denaturation of
human gastric lipase. Naturally occurring bile salts like taurodeoxycholate in
the concentrations prevailing in the upper small intestine reduce the interfacial
tension to about 9 dyn/cm. Other amphiphiles, like most alimentary proteins,
which decrease the tributyrin/water interfacial tension to values between 8 to
13 dynlcm, also allow the expression of lipase activity.
Prepyloric lipases can hydrolyze a variety of lipid substrates including
milk fat globule triacylglycerols. They are essential for the digestion of milk
fat in the newborn. It was thought until recently that intragastric digestion
was specific for short and medium chain triglycerides and had very little effect
on long chain triglycerides, the major constituents of dietary fat. However,
using purified human gastric lipase, Gargouri et a!. 19 have shown that human
gastric lipase catalyzes the hydrolysis of both long and short chain triacyl-
glycerols at comparable rates. Sn-3 positions of ester bonds in triglycerides
are preferentially hydrolyzed by the enzyme. An important role for gastric
lipolysis is to prepare triacylglycerols for further intraduodenal digestion.
B. PANCREATIC LIPASES
Pancreatic lipase (triacylglycerol acyl hydrolase, EC 3.1.1.3) is, along
with phospholipase A2 , the predominant pancreatic lipolytic enzyme. It amounts
to 4.5% of the total proteins in pancreatic juice. 20 It is synthesized and secreted
by pancreatic acinar cells and is present in duodenum aspirate. The optimum
pH for pancreatic lipase is at around pH 8 to 9. 21 Pancreatic lipase irreversibly
loses its activity below pH 5.0. 21
Pancreatic function is immature in neonates of most species (man, dog,
rat). However, a pancreatic lipase activity, although weak, is detectable during
the last week of fetal development in the rat. 22 Lipase level is still reduced
in the rat during the period from birth to weaning, lipase concentration reach-
ing 40% of that in adult at the end of weaning. 22 However, in healthy adults,
pancreatic lipase is considered to be of prime importance in the digestion of
dietary lipids.
Pancreatic lipase (52 kDa for the pig enzyme) hydrolyzes long chain
triacylglycerols (preferentially by attacking the external chains at positions
sn-1 and sn-3 of glycerides), producing first diacylglycerol, then 2-mono-
acylglycerol and free fatty acids. Pancreatic lipase, similarly to the gastric
enzyme, displays almost no activity when substrate is in the monomeric state. 23
60 Nutrition and Gene Expression
1. Intragastric Lipolysis
Lipids are mixed and dispersed with the other nutrients in the stomach.
Hydrolysis of alimentary lipids, catalyzed by one of the preduodenallipases
secreted from the serous glands of the tongue or from the gastric mu-
cosa,2·3.11 takes place in the stomach. It has been shown that these lipases are
particularly stable under the acidic conditions prevailing in the stomach (pH 1
to 3) and are activated by various dietary proteins. 33 During in vivo gastric
lipolysis, triacylglycerol is hydrolyzed to primarily a,f3-diacylglycerol and
free fatty acid. Production of free fatty acids facilitates emulsification of
triglycerides and diglycerides. 34 The amount of hydrolysis products (digly-
cerides and free fatty acids) increases over time following ingestion to reach
a maximum corresponding to 10 to 30% 17 to 65% 35 of total hydrolysis. The
lack of complete hydrolysis is most likely due to preduodenal lipolysis in-
hibition by long chain free fatty acids. 33 ·36
2. Intraduodenal Lipolysis
Pancreatic lipase and colipase are discharged into the duodenum and
associate first to bile lipid micelles and vesicles prior to adsorption to emul-
sified particles from the stomach. 37 Triglyceride-rich emulsion particles con-
taining 2 to 3% triglycerides are thus covered by a mixed lipid monolayer
Wicker-Planquart and Puigserver 61
The gastric phase of fat digestion is also thought to play an essential role
in milk digestion in preterm or fullterm neonates, where bile salt levels 46 and
pancreatic lipase secretions 47 -49 are quite low. Functional immaturity of ex-
ocrine pancreatic function, although associated with excretion of up to 25%
of ingested fat, does not appear to have a major clinical impact. 50 •51 It has
been shown that 14 to 60% of milk lipid is hydrolyzed to diglyceride, mon-
oglyceride, and free fatty acid in the first 30 to 60 min following ingestion. 35
Subsequent hydrolysis is achieved by a milk lipase (optimum pH = 8.5),
which acts in the intestine and requires bile salts to be active. This bile salt
stimulated lipase (BSSL) is identical to carboxyl ester hydrolase found in
pancreatic juice. 52 In vivo, the enzyme hydrolyzes monoglyceride to free
glycerol and fatty acid. Prior intragastric digestion greatly enhances BSSL
activity where 30% hydrolysis by BSSL is achieved, compared to 5% hy-
drolysis without prior exposure. 35 Thus, concerted action of the two enzymes
(preduodenallipase and BSSL) explains the efficiency of fat digestion in the
suckling neonate.
Isolated lipase deficiency, 53 - 55 co lipase deficiency, 56 and combined lipase-
co1ipase deficiencies 57•58 have been reported. These genetic diseases and pan-
creatic lipase deficiency caused by cystic fibrosis or chronic alcoholism are
commonly associated with fat excretion in the stools54 •58 but are not related
to growth deficit or malnutrition. Dietary fat of 50 to 70% in severe pancreatic
insufficiency59 or in isolated lipase and colipase deficiency 58 is indeed ab-
sorbed. In these situations. gastric enzyme plays an essential role in the
digestion, acting first in the stomach and then in the upper small intestine
where the postprandial pH is still acidic. 60
A. PREDUODENAL ADAPTATION
1. Gastrointestinal Transit
Inhibitory effect of fat in the small intestine on the rate of gastric emp-
tying has been reported. 61 However, the importance of the recent dietary
history in small bowel transit time has been emphasized. 62 Both gastric
emptying and mouth to cecum transit time are significantly faster after con-
sumption of a high fat diet for 14 days than after consumption of a low fat
diet in human 62 (half-time for gastric emptying = 98 vs. 147 min, mouth to
cecum transit time = 240 vs. 360 min, respectively). According to the
authors, the changes observed in gastrointestinal transit time could be related
to the release or sensitivity to humoral transmitters such as cholecystokinin
(CCK) and peptide YY (PYY). These peptides have been shown to be
released by the presence of lipid in the small intestine and to delay gastric
and small intestinal transit. 63 - 65
Wicker-Planquart and Puigserver 63
2. Prepyloric Lipases
In contrast with pancreatic lipase, which level is very low in the new-
bom,4749 the concentration of human gastric lipase at birth is quite comparable
to that found in the adult. 18 In the rat, lingual lipase activity was found to
raise exponentially immediately after birth. 66 This suggests that ingestion of
milk might be of major importance in eliciting preduodenal lipase activities
during the first days of postnatal life.
Only a few studies have clearly established that preduodenallipases were
able to undertake an adaptive response to dietary fat. The first preliminary
observation was made in 1978 by Hamosh, 4 who showed that rat lingual lipase
content in animals fed a high fat diet (22% com oil) for 2 weeks was 1.4-
fold higher than that of animals fed a similar purified diet containing 4% com
oil. A more detailed examination of the effect of dietary fat on lingual lipase
activity established that even 10% fat in the diet (composed of a mixture of
half sunflower oil and half lard), when given to rats for 3 weeks, was able
to induce the specific activity of lingual lipase by a 1.23-fold factor, the
maximum increase (1.30-fold) being reached when rats were fed a 20% fat
dietY Adding more fat to the diet (up to 30%) did not further enhance lingual
lipase activity. Maximum values of enzyme-specific activities were measured
after 1 week of feeding a 20% fat diet, and no additional change in lingual
lipase specific activity occurred when rats were fed the 20% fat diet for
4 weeks. Whatever the type of fat added in the diet (lard, sunflower oil, olive
oil, peanut oil, soybean oil, com oil, salmon oil, or butter), no significant
differences in lipase induction factor could be measured, i.e., the adaptive
response of lingual lipase to fat was not affected either by the chain length,
by the number, or the position of the double bonds in the fatty acid molecule,
even when dietary fat was provided in a moderate amount (10% by weight)Y
Evidence of an adaptive response to dietary fat has also been described for
the gastric enzyme in the rabbit. 68 Similar findings concerning the amount of
fat inducing a maximum increase in gastric lipase activity (6% fat in the diet
for fundus lipase and 12% for cardia lipase) or the lack of a specific effect
of the fatty acid composition of dietary triglyceride molecules on lipase ad-
aptation were reported. 68 However, feeding rabbits a 12% dietary fat diet for
1 week did not change the specific activity of lipase in the cardia or fundus
mucosae, as compared with feeding a 2.7% dietary fat diet. A 2-week period
was necessary to achieve adaptation of gastric enzyme.
acid chains, the other components of the diet, the species studied, and the
age of animal. This can explain the differences measured in the extent of
induction of pancreatic lipase.
In the rat, pancreatic lipase concentration is high at birth but falls after
1 d. 48 Emergence of lipase is quite slow since it represents 40% of the value
in adult rats just before weaning. 22 This suggests that an increase in pancreatic
lipase before weaning is not a direct consequence of milk fat intake during
suckling. Low pancreatic lipase secretions have also been reported in the
human infant. 49 . 51 and in the neonatal dog. 35 According to Snook, 48 lipase
accumulation before weaning occurs mainly in response to a developmental
clock that may be affected only indirectly by the type of food eaten. In fact,
weaning rats on a high lipid diet (50% com oil) does not increase the rate of
lipase accumulation beyond that observed in rats weaned on to a high starch
diet during the first 25 d after birth. 70 By contrast, 30-d-old rats on the high
fat diet have higher lipase content than 30-d-old rats on the high starch diet. 70
Thus, adaptation of pancreatic lipase to nutritional components only occurs
after the usual weaning period (about 3 weeks in the rat).
b. Chain Length
According to Sabb et al., 74 no influence of chain length (from C 12 to C 18)
was seen on lipase activity in weanling rats (40 to 60 g) fed high fat diets
(29% lipid, i.e., 67% kcal as fat) for 1 week since coconut oil, the dietary
~
TABLE 2 ~
Fatty Acid Composition of Diets, Induction Factors for Lipase and Colipase in Response
to the Ingestion of Fat Diets
Induction Factors
Main FA• Unsaturated Lipase Colipase
Diet (%) FA(%) Deschodt-Lanckman et al. 70 Sabb et al. 74 Saraux et al. 73 Saraux et al. 73
"'"'c:;·
;::
Wicker-Planquart and Puigsen'er 67
fat with the greatest percentage (45%) of cl2 chain length used in their study,
stimulated lipase similarly to the other fats (corn oil, lard, safflower oil,
butter, olive oil) (Table 2). Regarding the effects of lard (99% fatty chain
superior to cl2:0: 2% cl4:0> 29% cl6:0> 23% CI8:0> 2% cl6:1> 33% cl8:1> 10%
cl8:2) and coconut oil (20% c8:0 to CIO:O> 45% cl2:0> 18% cl4:0> 10.9% cl6:0>
9.8% C 18 ,0 ) in adult rats (180 g) fed 40% fat diet for 3 weeks, Saraux et al. 73
observed that lipase was stimulated to the same extent (about twofold). How-
ever, lipase- and colipase-specific activities were less efficiently increased
(only 1.5- and 1.3-fold, respectively) by diets containing medium chain tri-
glycerides (Table 2). The fact that absorption of the medium chain fatty acids
resulting from hydrolysis of medium chain triglycerides occurs via the portal
route, the fatty acids being rapidly transported to the liver and oxidized/0
might explain their relative inaction with respect to inducing maximal pan-
creatic lipase content. Unfortunately, in Sabb's and Saraux's studies, all
experimental diets, except the medium chain triglyceride one, contained vary-
ing amounts of unsaturated lipids, which, in turn, may influence induction
factor values for pancreatic lipase. Indeed, Deschodt-Lanckman et al. 70 re-
ported that a 50% (w:w) fat diet with tricaprylin as the lipid source, given to
adult rats (180 to 220 g) for 5 d, poorly increased lipase content, when
compared to other lipid diets. This effect seemed to be related to the degree
of saturation rather than to the chain length of the dietary fat since tricaprylin
(98% C 8 ,0 ) and tristearin (96% C 180) diets led to the same induction of lipase-
specific activity in pancreas homogenates (l.8-fold)1° (Table 2).
b. Colipase-Specific Activity
As far as the existence of an adaptation of pancreatic colipase to nutritional
lipids is concerned, contradictory findings have been reported. No adaptation
of colipase to dietary lipids was observed in either rats or mice by Vander-
meers-Piret eta!. ,92 whereas several studies have described colipase adaptation
TABLE 3 ~
r,
Specific Tissue Content in Lipase and Colipase Activity and Relative Synthesis of Lipase in
~....
Pancreatic Lobules in Response to Changes in Lipid in the DieC5
~
IS"
;::
Enzyme-specific activity in ~
~
~
TABLE 4 -...1
0
Experimental Procedures and Type of Dietary Fat Related to the Amount of Lipids Necessary to Induce
Minimal and Maximal Lipase Response in Rat Pancreatic Tissue
Experimental procedures Dietary fat Butter Lard Sunflower oil/lard Com oil Sunflower oil
Polyunsaturated/saturated 0.03 0.13 2.6 5.0 5.19
(from Ref. 67) adaptation 2 weeks 3 weeks 3 weeks I week IOd
period
Minimum amount of % By weight of dietary fat 34.6 23 20 23.2 10
fat-inducing lipase % Dietary calories as fat 55 45.4 39.3 54 25
adaptation Resulting induction factor 1.2 1.57 1.89 2.4 1.38
for lipase
Maximum induction of % Dietary calories as fat 77 73.2 52.7 67 62
lipase activity Maximum inducation factor 1.83 2.15 2.15 2.85 2.48
<:
:::
~·
c;·
:::::
:::,
:::::
:::::..
,a
~
tl1
-8
~
t;
c;·
:::::
Wicker-Planquart and Puigserver 71
to dietary fat in the raf 3 •75 •93 •94 as well as in the pig. 78 These conflicting
findings may be explained by quantitative and qualitative differences in dietary
protein ingestion. 73
The effect on rat colipase activity of changing relative proportions of
dietary fat and carbohydrate while keeping the diet isonitrogenous (22% pro-
tein, the daily protein intake being around 4 g per day) and isocaloric79 is
described in Table 3. In contrast with lipase, colipase activity was found to
exhibit a significant enhancement (1 .3-fold) in the pancreas of rats fed the
lipid-free diet, as compared to rats fed the standard diet. Between 3 and 25%
fat in the diet, a 2.3-fold increase in colipase-specific activity was measured
in the pancreas, which was lower than that of pancreatic lipase. Colipase
activity then slightly declined between 25 and 30% dietary fat but was still
significantly higher (I .9-fold) than that in control rats. Due to the differential
adaptation of lipase and its cofactor, the colipase/lipase ratio progressively
decreased from 0.93 in control animals to a minimal value of 0.73 in 30%
lipid-fed rats. Nonparallel adaptations of lipase and colipase, where colipase/
lipase ratio dropped from 0.78 to 0.51, have also been reported in the rat94
when dietary fat increased from 3 to 19.5% and in the pig, 78 where the ratio
decreased from 2.35 in 25% fat-fed pigs to 1.77 in 5% fat-fed animals. In
hyperlipidic diet-fed rats, intrinsic colipase level is therefore not high enough
to allow full activation of pancreatic lipase when bile salts are present.
Saraux et a!. 73 showed that lipid intake modulates co lipase in the rat only
when the protein intake is between 3.5 and 6.0 g per day. When protein
ingestion is below 3.5 g, colipase does not adapt to dietary lipids (from 2 to
40%, by weight), while above 6.0 g protein intake, colipase level is maximal,
even with 2% lipid in the diet. Between 3.5 and 6.0 g, lipid intake modulates
colipase and lipase in a parallel way, colipase/lipase ratio being always around
0. 7. In their conditions, a correlation coefficient of co lipase with fat intake
of 0.64 was reported. The correlation of colipase with protein intake was
even stronger (0.85). 73 The particular sensitivity of colipase to nutritional
proteins might explain that their lipidic diets did increase colipase content to
the same extent as lipase since, unfortunately, their experimental diets were
not isonitrogenous, an increase in the protein content of the diet (from 17 to
37%) accompanying the rise in fat (from 0. 7 to 45%, by weight, corresponding
to 1.7 to 73.2% of total energy). Nevertheless, these findings suggest that
there is a cooperative effect on colipase activity between lipid and protein in
the rat in the range of 3.5 to 6.0 g daily protein intake. 73 •
c. Lipase Biosynthesis
There is little information on the role of dietary lipids in the regulation
of the earlier steps in the secretory pathway of acinar cells, especially their
effect on protein synthesis. Data concerning lipase biosynthesis, as influenced
by dietary lipids, are quite scarce. 71 •75 •95 Snook, 71 when directly measuring
protein synthesis in vitro by the rate of incorporation of 14C-valine into pan-
creas slices from rats fed a high fat diet (providing more than 55% of the
72 Nutrition and Gene Expression
dietary calories as fat) or a control diet, reported that slices from rats fed the
high fat diet incorporated more label into pancreatic proteins. We found that
the increase in the synthesis of total pancreatic secretory proteins was about
twofold in magnitude 75 by incubating pancreatic lobules from rats fed high
fat (25 and 30% sunflower oil) and standard (3% sunflower oil) diets in the
presence of 35 S-methionine for 30 min and measuring incorporation of labeled
methionine into the pancreatic secretory proteins.
The increase in pancreatic lipase content by the high fat diets was directly
caused by an enhanced synthesis of the enzyme, as indicated by the good
correlation between the increases in lipase-specific activity and relative syn-
thesis. The specific enhancement in lipase fractional synthetic rate (expressed
as the percentage of radioactivity incorporated into pancreatic lipase, com-
pared to radioactivity incorporated into the total mixture of exocrine proteins)
was about twofold in the high fat diet fed animals (Table 3), a plateau in
lipase biosynthesis being reached when rats were given the 20% fat diet.
+
+
•+
+ +
1 400
l
f
..!..
300
+
•
I
1
200
•
• 100
.i..:I 0
0 1
D A Y 8
J
•
FIGURE 1. Short-term dietary adaptation oflipase (empty blocks) and colipase (shaded blocks)
activities in rat pancreatic tissue, as a function of the ingestion period (0 to 5 d) of a high fat
diet (25% sunflower oil). 95 Each point represents the mean value of five determinations. The
vertical bars indicate the SE. The P values, calculated from Student's t-test comparing 25%
lipidic diet to the control diet (3% lipid), are <0.05( + ), <0.01( + + ), <0.001( + + + ).
b. Colipase Activity
Short-term adaptation of colipase does not resemble that of lipase. Co-
lipase responds more slowly than lipase; its content in the pancreas decreases
initially and then increases gradually in response to dietary fat (Figure 1).
Coenzyme level on day 5 is quite comparable to that of rats fed the standard
diet. 95 Increasing the adaptation period to 10 d leads to a twofold induction
in colipase activity. 75
Although the alterations in food composition change the lipase and co-
lipase content, the mechanisms for the changes are probably distinct, as
suggested by the differential short-term adaptation responses of both pan-
creatic proteins.
DAY S
0 2 3 5
A
L
PCP
SP
low fat fed rats ( 11% energy as fat corresponding to 5% fat) cultured in low
glucose medium for 48 h, when the antecedent diet is a semipurified diet,
prepared with well-defined components. 102 The magnitude of lipase induction
in these conditions is relatively small (33%). On the other hand, when the
antecedent diet, providing 10% energy as fat, is prepared from crude com-
ponents, no increase in cellular lipase activity was observed. 102 According to
the authors, 102 their results support a role for the ketones in the regulation of
pancreatic lipase; however, ketones are not the sole mediator and might act
in conjunction with other food components.
2. Hormonal Regulators
Hormonal factors are thought to be involved in lipase response to dietary
fat. Fat hydrolysis products are assumed to act on the intestinal mucosa (more
likely on the proximal part of the small intestine 103 ), leading to a secretion
of peptide hormones. In fact, the presence of lipid in the small intestine leads
to a release of CCK and secretin in man, 63 dog, 64 •85 •86 and ratHl4 but not in
the pig. 105 CCK does not seem to be the mediator of lipase adaptation since
intravenous administration of caerulein, a synthetic analog of CCK, does not
modify lipase synthesis. 106 On the other hand, secretin is a possible inter-
mediate between ingested fat and pancreatic lipase regulation since the hor-
mone exerts stimulatory effects on lipase concentration in pancreatic tissue 107
and on total protein 108 and lipase 109 synthesis. On the other hand, secretin
does not alter the lipase content of cultured pancreatic cells. 110 Injection of
secretin for 15 days increases the pancreatic weight, RNA amount, and lipase
content in the rat pancreas. 107 These findings have been confirmed and studied
more extensively by others. 108 • 109 Infusion of synthetic secretin in conscious
rats (16 clinical units per kg and h) elicits total protein synthesis after a 2-
to 3-h lag period, a maximal twofold enhancement being obtained after a 12-
h infusion. 108 When measuring rates of synthesis of individual exocrine pro-
teins by following the incorporation of a mixture of 15 14 C-labeled amino
acids into proteins, a pronounced increase (by a 2.5 factor) in the relative
synthetic rate of lipase was observed. 109 This leads to a calculated 4. 9- and
4.1-fold change with time, after 12- and 24-h stimulation, respectively, in
lipase absolute synthetic rate. These values are very close to those obtained
from high fat fed rats/ 5 ·95 as far as relative and absolute synthesis rates of
lipase are concerned. Therefore it can be concluded that secretin remains a
very possible mediator in lipase adaptation. Whether other hormones in the
secretin family such as glucagon, vasoactive intestinal polypeptide (VIP), and
gastric inhibitory polypeptide (GIP) also have a stimulatory effect on lipase
synthesis has not yet been demonstrated.
Finally, other nonpeptidic hormones, such as steroid ones, are not re-
sponsible for nutritional changes of lipase synthesis.
76 Nutrition and Gene Expression
l
ex on intron exon
Transcription
hn RNA
l
Processing -cap structure
NUCLEUS -poly A tailing
-adenylate methylation
-splicing
ACXNAIR. mature mRNA
------------------------------------------------ ~ Transport-------------------------------
CELL mature mRNA
~ mRNA
translationJ
Proteint
mRNA have been reported, 122 which allow. with due precaution. satisfactory
preparation of pure, biologically active material. Quantitation of specific
mRNA expression has been then rendered possible thanks to the establishment
of lipase and colipase eDNA clones. The cloning and characterization of
pancreatic colipase eDNA has been performed in man, 123 · 124 rat, 125 and dog, 126
those of pancreatic lipase eDNA in man, 127 dog. 128 and rat recently. 129 We
have chosen, in the assay of lipase and colipase mRNAs, to hybridize mRNA
immobilized on filters to the corresponding synthetic labeled eDNA. This is
a more sensitive method than the translational one in a cell-free system.
a. Lipase mRNA
The effects of a 10-d ingestion of 10% or 25% sunflower oil on lipase
mRNA level result in a 2.2- and 3.9-fold increase in the lipase messenger,
respectively. 130 These data support a pretranslational mechanism by which
triglycerides regulate pancreatic adaptation.
When we examined the kinetics of the cellular mRNA concentration to
dietary stimulation, two distinct responses were obtained. 95 · 131 Lipase mRNA
increased in a biphasic manner in adapted rats. 95 This may have been the
result of the presence of a rather large amount of sodium bicarbonate (18.5%
by weight) in the experimental diet (25% sunflower oil). Indeed, we had
emphasized in another study 130 the possible interference of sodium bicarbonate
with the lipase response, since sodium bicarbonate had been shown to be
responsible for a variety of side effects, such as sodium overload. impaired
absorption of calcium, and an increase in plasma CCK level. On the other
hand, lipase mRNA was found to be maximally induced as soon as the second
day of lipid diet intake (by twofold) when no sodium bicarbonate was present
in the 25% sunflower diet and did not further increase up to 10 d. 131 Never-
theless, all these data suggest that the mechanism of lipase adaptation by
dietary lipids is mostly pretranslational. Changes in lipase mRNA levels
consecutive to lipid diet ingestion probably result from an increased tran-
scriptional rate of lipase gene. At present, we cannot exclude yet other mech-
anisms, such as RNA processing. regulation of lipase messenger transport,
and changes in mRNA stability.
The rise in lipase mRNA was consistent with the increase in the measured
values for lipase relative synthesis. 130 · 131 However, when the total increase
in pancreatic protein synthesis was taken into account. lipase absolute syn-
thesis rates increased more than the mRNA level. It is therefore possible that
a control mechanism acting at the translational level favors the translation of
lipase mRNA among the different pancreatic mRNAs. There is now a great
deal of evidence that such translational discriminations between different
mRNAs exist. 117• 132
The effect of a high fat diet on lipase mRNA levels in pancreatic tissue
allows us to eliminate the steroid hormones as possible regulators in diet
80 Nutrition and Gene Expression
b. Colipase mRNA
The level of colipase mRNA, as measured by dot-blot hybridization with
the corresponding eDNA probe, moderately increases, by a 1.5-fold factor,
in the pancreas of high fat adapted rats (25% sunflower oil), a plateau being
reached on the 3rd day of diet ingestion. 125 This finding suggests that ad-
aptation of pancreatic colipase to dietary lipid is genetically regulated. It is
nevertheless difficult to correlate colipase mRNA level and cofactor activity
in pancreatic tissue, since alterations in cofactor synthesis and secretion may
occur. The magnitude of the rise in colipase cytoplasmic mRNA concentra-
tion, although significant, is less than that in lipase mRNA under the same
experimental conditions. 131 This can explain the differences in induced activity
levels of both proteins in adapted rats. 75 •95
TABLE 5
Changes in Lipase Relative Synthesis, mRNA Levels, and
Transcriptional Rates, Induced by a Lipid-Rich Diet 131
Days on diet
Changes 0 2 3 5
Biosynthesis' 6.1 ± 0.5 6.7 ± 0.6 7.8 ± 0.8 8.9 ± 0.5 9.6 ± 0.7
mRNA• I 1.4 ± 0.08 1.79 ± 0.28 1.83 ± 0.16 1.99 ± 0.33
Transcriptional rate' 3.3 4.7 ND ND 5.7
not suitable for the study of the canine gene. Another possibility is that
regulatory elements are positioned in another region of the gene. It would be
of interest to test the 5' flanking region of the rat gene, when rat lipase gene
cloning will be performed, and analyze the upstream region to help to define
genetic elements that mediate the induction of the lipase gene by nutritional
agents and secretin.
In the case of other pancreatic genes, it has been possible, when con-
structing expression recombinant plasmids containing the 5' flanking region
of rat chymotrypsin gene linked to the coding sequence of a reporter gene
such as chloramphenicol acetyltransferase (CAT), to obtain specific expres-
sion of chymotrypsin gene recombinant in the rat exocrine pancreas tumor
line cells AR4-2J, but not in HIT cells, a transformed line from hamster
pancreatic endocrine cells. 114 Careful examination of the DNA sequences
upstream for nine rat pancreatic genes (amylase, carboxypeptidases Al and
A2, chymotrypsin B, elastases I and II, ribonuclease, trypsins I and II) coupled
with deletion experiments, have led to the identification of a consensus se-
quence: GTCACCTGTGCTITTCCCTG located -89 to -213 bp from the
cap site; this confers preferential expression of these genes in pancreatic
exocrine cells. 116 These regulatory sequences are similar to other enhancers
discovered so far in that they work whether they are located before or after
the genes and whether they are in the normal orientation or are inverted. 116
Enhancers are believed to work through the binding of pancreatic nuclear
protein, which leads to a change in the structure of the chromosome in the
vicinity of the gene. Indeed, two teams independently have demonstrated
binding of pancreatic nuclear protein with the enhancer consensus sequence
of mouse amylase gene 139 and rat chymotrypsin gene. 140 The specific factor
for amylase gene consensus element isolated from rat pancreatic nuclei in-
teracts also with elastase I gene consensus sequence, as indicated by com-
petition experiments for protein binding to the amylase sequences. 139 Site-
directed mutagenesis of the amylase sequence results in concomitant loss of
protein binding and enhancer activity, which indicates that the nuclear protein
is a positive regulator, controlling the activity of the amylase gene enhancer. 139
Another factor, isolated from 266-6 and HeLa cells, having the DNA-binding
specificity of MLTF (major late transcription factor of adenovirus), has been
shown to bind to the chymotrypsin sequence in vitro. 140 Site-directed muta-
genesis of chymotrypsin sequence into a perfect MLTF site results in the loss
of enhancer activity in acinar cells, but not of the binding of MLTF-like
factor. 140 This finding seems to indicate that MLTF-like factor acts as a
negative regulator of chymotrypsin enhancer function. Such transcriptional
repressions have also been described in other systems. 14 I. 142
Finally, another exciting new area of active research is the generation of
transgenic mice which can be analyzed and tested under a variety of physi-
ological circumstances. Such studies have already led to pancreas-specific
expression of amylase/CAT construct in transgenic lines and to the study of
Wicker-Planquart and Puigserver 83
insulin dependence of the hybrid amylase gene 143 or to the study of islet-
specific expression of human insulin and its regulation by glucose and amino
acids. 144
All these techniques constitute powerful tools which will be exploited in
the future and will open up other sets of experimental possibilities for the
study of lipase and colipase gene regulation.
REFERENCES
1. Lairon, D. and Borel, P., La regulation nutritionnelle des enzymes de Ia digestion des
lipides, Cah. Nutr. Diet., 24, 413, 1989.
2. Gargouri, Y., Moreau, H., and Verger, R., Gastric lipases: biochemical and physio-
logical studies, Biochem. Biophys. Acta, 1006, 255, 1989.
3. Schonheyder, F. and Volquartz, K., The gastric lipase in man, Acta Physiol. Scand.,
11,349,1946.
4. Hamosh, M., Rat lingual lipase: factors affecting enzyme activity and secretion, Am. J.
Physiol., 235, E416, 1978.
5. Field, R. B. and Scow, R. 0., Purification and characterization of rat lingual lipase, J.
Bioi. Chern., 258, 14563, 1983.
6. Sweet, B. J., Matthews, L. C., and Richardson, T., Purification and characterization
of pregastric esterase from calf, Arch. Biochem. Biophys., 234, 144, 1984.
7. Bernback, S., Hernell, 0., and Bliickberg, L., Purification and molecular character-
ization of bovine pregastric lipase, Eur. J. Biochem., 148, 233, 1985.
8. Cohen, M., Morgan, R. G. H., and Hofmann, A. F., Lipolytic activity of human
gastric and duodenal juice against medium and long-chain triglycerides, Gastroenterology,
60, 1, 1971.
9. Perret, J. P., Lipolyse gastrique chez le lapereau. Origine et importance physiologique
de Ia lipase, J. Physiol. (Paris). 78. 221, 1982.
10. DeNigris, S. J., Hamosh, M., Kasbekar, D. K., Lee, T. C., and Hamosh, P., Lingual
and gastric lipases: species differences in the origin of prepancreatic digestive lipases and
in the localization of gastric lipase, Biochim. Biophys. Acta, 959, 38, 1988.
II. Moreau, H., Gargouri, Y., Lecat, D., Junien, J, L., and Verger, R., Screening of
preduodenallipases in several mammals, Biochem. Biophys. Acta, 959, 247, 1988.
12. Roberts, I. M. and Jaffe, R., Lingual lipase: immunocytochemical localization in the
rat von Ebner gland, Gastroenterology, 90, 1170, 1986.
13. Roberts, I. M., Nochomovitz, L. E., Jaffe, R., Hanel, S. I., Rojas, M., and Agostini,
R. A., Immunocytochemical localization of lingual lipase in serous cells of the developing
rat tongue, Lipids, 22, 764, 1987.
14. Abrams, C. K., Hamosh, M., Lee, T. C., Ansher, A. F., Collen, M. J., Lewis,
J. H., Benjamin, S. B., and Hamosh, P., Gastric lipase: localization in the human
stomach, Gastroenterology, 95, 1460, 1988.
15. Moreau, H., Laugier, R., Gargouri, Y., and Verger, R., Human gastric lipase is
entirely of gastric fundic origin, Gastroenterology, 95, 1221, 1988.
16. Moreau, H., Bernadac, A., Gargouri, Y., Benkouka, F., Laugier, R., and Verger,
R., lmmunocytolocalization of human gastric lipase in chief cells of the fundic mucosa,
Histochemistry, 91,419, 1989.
84 Nutrition and Gene Expression
17. Moreau, H., Bernadac, A., Tn!tout, N., Gargouri, Y., Ferrato, F., and Verger, R.,
Immunocytochemical localization of rabbit gastric lipase and pepsinogen, Eur. J. Cell
Bioi., 51, 165, 1990.
18. Fredrickzon, B. and Hernell, 0., Role of feeding on lipase activity in gastric contents,
Acta Paediatr. Scand., 66, 479, 1977.
19. Gargouri, Y., Pieroni, G., Riviere, C., Sauniere, J, F., Lowe, P. A., Sarda, L., and
Verger, R., Kinetic assay of human gastric lipase on short- and long-chain triacylglycerol
emulsions, Gastroenterology, 91, 919, 1986.
20. Schick, J,, Verspohl, R., Kern, H., and Scheele, G., Two distinct adaptative responses
in the synthesis of exocrine pancreatic enzymes to inverse changes in protein and car-
bohydrate in the diet, Am. J. Physiol., 247, G611, 1984.
21. Verger, R., Pancreatic lipases, in Lipases, Borgstrom, B. and Brockman, H. L., Eds.,
Elsevier, Amsterdam, 1984, 84.
22. Saraux, B. and Girard-Globa, A., Development of pancreatic enzymes in fetal and
suckling rats with emphasis on lipase and colipase, J. Dev. Physiol., 4, 121, 1982.
23. Sarda, L. and Desnuelle, P., Action de la lipase pancreatique sur les esters en emulsion,
Biochem. Biophys. Acta, 30, 513, 1958.
24. Semeriva, M. and Desnuelle, P., Pancreatic lipase and colipase. An example of het-
erogeneous biocatalysis, Adv. Enzymol., 48, 319, 1979.
25. Maylie, M.-F., Charles, M., Gache, C., and Desnuelle, P., Isolation and partial
identification of a pancreatic colipase, Biochem. Biophys. Acta, 229, 286, 197!.
26. Chapus, C., Sari, H., Semeriva, M., and Desnuelle, P., Role of colipase in the
interfacial adsorption of pancreatic lipase at hydrophobic interfaces, FEBS Lett., 58, 155,
1975.
27. Patton, J, S., Donner, J,, and Borgstrom, B., Lipase-colipase interactions during gel
filtration high and low affinity binding situations, Biochem. Biophys. Acta, 529, 67,
1978.
28. Sauve, P. and Desnuelle, P., Interactions of pancreatic colipase with taurodeoxycholate-
oleate mixtures above the critical micellar concentrations, FEBS Lett., 122, 91, 1980.
29. Borgstrom, B. and Hildebrand, H., Lipase and colipase activities of human small
intestinal contents after a liquid test meal, Scand. J. Gastroenterol., 10, 585. 1975.
30. Patton, J, S., Albertsson, P.-A., Erlanson, C., and Borgstrom, B., Binding of porcine
pancreatic lipase and colipase in the absence of substrate studied by two-phase partition
and affinity chromatography, J. Bioi. Chern., 253, 4195, 1978.
3!. Staggers, J. E., Hernell, 0., Stafford, R. J,, and Carey, M. C., Physical-chemical
behavior of dietary and biliary lipids during intestinal digestion and absorption. I. Phase
behavior and aggregation states of model lipid systems patterned after aqueous duodenal
contents of healthy adult human beings, Biochemistry, 29, 2028, 1990.
32. Hernell, 0., Staggers, J, E., and Carey, M. C., Physical-chemical behavior of dietary
and biliary lipids during intestinal digestion and absorption. II. Phase analysis and ag-
gregation states of luminal lipids during duodenal fat digestion in healthy adult human
beings, Biochemistry, 29, 2041, 1990.
33. Gargouri, Y., Pieroni, G., Lowe, P. A., Sarda, L., and Verger, R., Human gastric
lipase. The effect of amphiphiles, Eur. J. Biochem., 156, 305, 1986.
34. Borgstrom, B., Importance of phospholipids, pancreatic phospholipase A2 and fatty acid
for the digestion of dietary fat, Gastroenterology, 78, 954, 1980.
35. Iverson, S. J., Kirk, C. L., Hamosh, M., and Newsome, J., Milk lipid digestion in
the neonatal dog: the combined actions of gastric and bile salt stimulated lipases, Biochim.
Biophys. Acta, 1083, 109, 1991.
36. Hamosh, M., Fat digestion in the newborn: role of lingual lipase and preduodenal
digestion, Pediatr. Res., 13, 615, 1979.
37. Lairon, D., Nalbone, G., Lafont, H., Leonardi, J,, Domingo, N., Hauton, J. C.,
and Verger, R., Possible roles of bile lipids and colipase in lipase adsorption, Biochem-
istry, 17, 5263, 1978.
Wicker-Planquart and Puigserver 85
38. Miller, K. W. and Small, D. M., The phase behavior of triolein, cholesterol and
emulsions, J. Colloid Int. Sci., 89, 466, 1982.
39. Carey, M. C., Small, P. M., and Bliss, C. M., Lipid digestion and absorption, Annu.
Rev. Physiol., 45, 651, 1983.
40. Hofman, A. and Borgstrom, B., Physical-chemical state of lipids in intestinal content
during their digestion and absorption, Fed. Proc., 21, 43, 1962.
41. Patton, J, S. and Carey, M. C., Watching fat digestion, Science, 204, 145, 1979.
42. Lindstrom, M., Ljusberg-Wahren, H., and Larsson, K., Aqueous lipid phases of
relevance to intestinal fat digestion and absorption, Lipids, 16, 749, 1981.
43. Clark, B. S., Lawergren, B., and Martin, J, V., Regional intestinal absorptive ca-
pacities for triolein: an alternative to markers, Am. J. Physiol., 225, 574, 1973.
44. Tso, P., Pitts, V., and Granger, D. N., Role of lymph flow in intestinal chylomicron
transport, Am. J. Physiol., 249, G21, 1985.
45. Plucinski, T. M., Hamosh, M., and Hamosh, P., Fat digestion in rat: role of lingual
lipase, Am. J. Physiol., 237, E541, 1979.
46. Watkins, J. B., Ingall, D., and Szczepanik, P., Bile salt metabolism in the newborn,
New Engl. J. Med., 288, 431, 1973.
47. Robberecht, P., Deschodt-Lanckman, M., Camus, J., Bruylands, J., and Christophe,
J., Rat pancreatic hydrolases from birth to weaning and dietary adaptation after weaning,
Am. J. Physiol., 221, 376, 1971.
48. Snook, J. T., Effect of diet on development of exocrine pancreas of the neonatal rat,
Am. J. Physiol., 221,1388,1971.
49. Zoppi, G., Andreotti, G., Pajno-Ferrara, F., Njai, D. M., and Gaburro, D., Exocrine
pancreas function in premature and full term neonates, Pediatr. Res., 6, 880, 1972.
50. Fomon, S. J., Ziegler, E. R., Thomas, L. N., Jensen, R. L., and Filer, L. J.,
Excretion of fat by normal full term infants fed various milks and formulae, Am. J. Clin.
Nutr., 23, 1299, 1970.
51. Katz, L. and Hamilton, J. R., Fat absorption in infants of birth weight less than 1300 g,
J. Pediatr., 85, 608, 1974.
52. Blackberg, L., Lombardo, D., Hernell, 0., Guy, 0., and Olivecrona, T., Bile-salt-
stimulated lipase in human milk and carboxyl ester hydrolase in pancreatic juice. Are
they identical enzymes?, FEES Lett., 136, 284, 1981.
53. Sheldon, W., Congenital pancreatic lipase deficiency, Arch. Dis. Child., 39, 268, 1964.
54. Larbre, F., Hartman, E., Cotton, J. B., Mathieu, M., Charrat, A., and Moreau,
P., Diarrhee chronique par absence de lipase pancreatique, Pediatric, 24, 807, 1969.
55. Figarella, C., DeCaro, A., Leupold, D., and Poley, J., Congenital pancreatic lipase
deficiency, J. Pediatr., 96, 412, 1980.
56. Hilderbrand, H., Borgstrom, B., Bokassy, A., Erlanson-Aibertsson, C., and Helin,
I., Isolated colipase deficiency in two brothers, Gut, 23, 243, 1982.
57. Fayez, K., Ghishan, J., Moran, R., Durie, P. R., and Greene, H. L., Isolated
congenitallipase-colipase deficiency, Gastroenterology, 86, 1580, 1984.
58. Ligumsky, M., Granot, E., Branski, D., Stankiewicz, H., and Goldstein, R., Isolated
lipase and colipase deficiency in two brothers, Gut, 31, 1416, 1990.
59. Ross, C. A. C. and Sammons, H. G., Nonpancreatic lipase in children with pancreatic
fibrosis, Arch. Dis. Child., 30, 428, 1955.
60. Abrams, C. K., Hamosh, M., VanHubbard, S., Dutta, S. K., and Hamosh, P.,
Lingual lipase in cystic fibrosis: quantitation of enzyme activity in the upper small intestine
of patients with exocrine pancreatic insufficiency, J. Clin. Invest., 73, 374, 1984.
61. Cooke, A. R., Control of gastric emptying and motility, Gastroenterology, 68, 804,
1975.
62. Cunningham, K. M., Daly, J., Horowitz, M., and Read, N. W., Gastrointestinal
adaptation to diets of differing fat composition in human volunteers, Gut, 32, 483, 1991.
86 Nutrition and Gene Expression
84. Ballestra, M. C., Manas, M., Mataix, F. J., Martinez-Victoria, E., and Seiquer,
I., Long-term adaptation of pancreatic response by dogs to dietary fats of different degrees
of saturation: olive and sunflower oil, Br. J. Nutr., 64, 487, 1990.
85. Kim, Y. C., Faichney, A., and Ky, L., Endogenous release of secretin by sodium oleate
in dog, Gastroenterology, 78, 1195, 1980.
86. Faichney, A., Chey, W. Y., Kim, Y. C., Lee, K. Y., Kim, M. S., and Chang, T. M.,
Effect of sodium oleate on plasma secretin concentration and pancreatic secretion in dogs,
Gastroenterology, 81, 458, 1981.
87. Fink, A. S., Taylor, I. L., Luxemburg, M., and Meyer, J. H., Pancreatic polypeptide
release by intraluminal fatty acids, Metabolism, 32, 1063, 1983.
88. Sommer, H. and Kasper, H., Dietary effects on pancreatic exocrine function. Exper-
iments on the isolated perfused rat pancreas, Ann. Nutr. Metab., 25, 38I, 1981.
89. Belleville, J., Prost, J., and Gillet, M., Effets des regimes riches en triglycerides et
en phospholipides sur les activites de !a lipase et de la phospholipase A du sue pancreatique
et du pancreas de rat, Arch. Int. Physiol. Biochim., 86, 631, 1978.
90. Ouagued, M., Saraux, B., Girard-Globa, A., and Bourdel, G., Differential regulation
of lipase and colipase in the rat pancreas by dietary fat and proteins, J. Nutr., 110, 2302,
1980.
91. Robberecht, P., Deschodt-Lanckman, M., Camus, J., Bruylands, J., and Christophe,
J., Rat pancreatic hydrolases from birth to weaning and dietary adaptation after weaning,
Am. J. Physiol., 221, 376, 1971.
92. Vandermeers-Piret, M. C., Vandermeers, A., Wijns, W., Rathe, J., and Christophe,
j., Lack of adaptation of pancreatic colipase in rats and mice, Am. J. Physiol., 232,
E131, 1977.
93. Girard-Globa, A. and Simond-Cote, E., Nutritional and circadian variations in lipase
activity and colipase saturation in rat pancreas, Ann. Bioi. Anim. Biochim. Biophys.,
17, 539, 1977.
94. Lairon, D., Lacombe, C., Borel, P., Corraze, G., Nibbelink, M., Chautan, M.,
Chanussot, F., and Lafont, H., Beneficial effects of wheat germ on circulating lipo-
proteins and tissue lipids in rats fed a high fat, cholesterol-containing diet, J. Nutr.,
117, 838, I 987.
95. Wicker, C. and Puigserver, A., Changes in mRNA levels of rat pancreatic lipase in
the early days of consumption of a high-lipid diet, Eur. J. Biochem., I80, 563, I989.
96. Stabile, B. E., Borzatta, M., Stubbs, R. S., and Debas, H. T., Intravenous mixed
aminoacids and fats do not stimulate exocrine pancreatic secretion, Am. J. Physiol., 246,
G274. I984.
97. Burns, G. P. and Stein, T. A., Pancreatic enzyme secretion during intravenous fat
infusion, J. Parenteral Enteral Nutr., I I, 60, 1987.
98. Edelman, K. and Valenzuela, J. E., Effect of intravenous lipid on human pancreatic
secretion, Gastroenterology, 85, 1063, I983.
99. Konturek, S. J., Tasler, J., Cieszkowski, M., Jaworek, J.,and Konturek, J., Intra-
venous amino acids and fat stimulate pancreatic secretion, Am. J. Physiol., 236, E678,
I979.
100. Lavan, M., Bazin, R., and Herzog, J., Comparative effects of oral and parenteral
feeding on pancreatic enzymes in the rat, J. Nutr., 104, 1432, I974.
101. Bazin, R., Lavan, M., and Herzog, J., Pancreatic lipase and ketogenic conditions,
Biomedicine, 28, 160, 1978.
102. Hirschi, K. K., Sabb, j. E., and Brannon, P. M., Effects of diet and ketones on rat
pancreatic lipase in cultured acinar cells, J. Nutr., 121, 1129, 1991.
I 03. Simoes Nunes, C., Lack of pancreatic enzyme adaptation to diet carbohydrates and lipids
after proximal small intestine bypass in the pig, Digestion, 25, I08, I982.
I04. Douglas, B. R., Woutersen, R. A., Jansen, J. B. M. J., De Jong, A. J. L., and
Lamers, C. B. H. W., The influence of different nutrients on plasma cholecystokinin
levels in the rat, Experimentia, 44, 2I, I 988.
88 Nutrition and Gene Expression
105. Corring, T. and Chayvialle, J, A., Diet composition and the plasma levels of some
peptides regulating pancreatic secretion in the pig, Reprod. Nutr. Develop., 27, 967, 1987.
106. Schick, J., Kern, H., and Scheele, G., Hormonal stimulation in the exocrine pancreas
results in coordinate and anticoordinate regulation of protein synthesis, J. Cell Bioi., 99,
1569, 1984.
107. Solomon, T. E., Petersen, H., Elashoff, I., and Grossman, M. J., Interaction of
caerulein and secretin on pancreatic size and composition in the rat, Am. J. Physiol.,
235, E714, 1978.
108. Rausch, U., Vasiloudes, P., Riidiger, K., and Kern, H. F., In-vivo stimulation of rat
pancreatic acinar cells by infusion of secretin. I. Changes in enzyme content, pancreatic
fine structure and total rate of protein synthesis, Cell Tissue Res., 242, 633, 1985.
109. Rausch, U., Vasiloudes, P., Riidiger, K., and Kern, H. F., In-vivo stimulation of rat
pancreatic acinar cells by infusion of secretin. II. Changes in individual rates of enzyme
and isoenzyme biosynthesis, Cell Tissue Res., 242, 641, 1985.
I 10. Kemper, K. A. and Brannon, P. M., Effects of secretin on enzymes of cultured
pancreatic acinar cells, FASEB J., 2, A731, 1988.
Ill. Brown, D. D., Gene expression in eukaryotes, Science, 211,667, 1981.
112. Darnell, J, E., Variety in the level of gene control in eukaryotic cells, Nature, 297, 365,
1982.
113. Mitchell, P. J. and Tjian, R., Transcriptional regulation in mammalian cells by sequence-
specific DNA binding proteins, Science, 245, 371, 1989.
114. Walker, M., Edlund, T., Boulet, A., and Rutter, W., Cell specific expression con-
trolled by the 5' flanking region of insulin and chymotrypsin genes, Nature, 306, 557,
1983.
115. Swift, G., Hammer, R., MacDonald, R., and Brinster, R., Tissue specific expression
of the pancreatic elastase I gene in transgenic mice, Cell. 38, 639, 1984.
116. Boulet, A. M., Erwin, C. R., and Rutter, W. J., Cell-specific enhancers in the rat
exocrine pancreas, Proc. Nat!. Acad. Sci. U.S.A., 83, 3599, 1986.
117. Austin, S. A. and Kay, J. E., Translational regulation of protein synthesis in eukaryotes,
Essays Biochem., 18, 79, 1982.
118. Ross, R. J., Messenger RNA turnover in eukaryotic cells, Mol. Bioi. Med., 5, I, 1988.
119. Jamieson, J.D. and Palade, G. E., Synthesis, intracellular transport and discharge of
secretory proteins in stimulated pancreatic exocrine cells, J. Cell Bioi., 50, 135, 1971.
120. Blobel, G. and Dobberstein, B., Transfer of proteins across membranes. Presence of
proteolytically processed and unprocessed nascent immunoglobulin light chains on mem-
brane-bound ribosomes of murine myeloma, J. Cell Bioi., 67, 835, 1975.
121. Barnard, E. A., Biological function of pancreatic ribonuclease, Nature, 221, 340, 1969.
122. Chirgwin, J, M., Przybyla, A. E., MacDonald, R. J., and Rutter, W. J., Isolation
of biologically active ribonucleic acid from sources enriched in ribonuclease, Biochem-
istry, 18, 5294, 1979.
123. Lowe, M. E., Rosenblum, J. L., McEven, P., and Strauss, A. W., Cloning and
characterization of the human colipase eDNA, Biochemistry, 29, 823, 1990.
124. Renaud, W. and Dagorn, J, C., eDNA sequence and deduced amino acid sequence of
human preprocolipase, Pancreas, 6, 157, 1991.
125. Wicker, C. and Puigserver, A., Rat pancreatic colipase mRNA: nucleotide sequence
of a eDNA clone and nutritional regulation by a lipidic diet, Biochem. Biophys. Res.
Commun., 167, 130, 1990.
126. Fukuoka, S.-1., Taniguchi, Y., Kitagawa, Y., and Scheele, G., Full length eDNA
sequence encoding canine pancreatic colipase, Nucleic Acids Res., 18, 5549, 1990.
127. Lowe, M. E., Rosenblum, J. L., and Strauss, A. W., Cloning and characterization of
human pancreatic lipase eDNA, J. Bioi. Chern., 264, 20042, 1989.
Wicker-Planquart and Puigserver 89
128. Kerfelec, B., LaForge, K. S., Puigserver, A., and Scheele, G., Primary structures of
canine pancreatic lipase and phospholipase A2 messenger RNAs, Pancreas, I, 430, 1986.
129. Wicker-Planquart, C. and Puigserver, A., Primary structure of rat pancreatic lipase
mRNA, FEBS Lett., 296, 61, 1992.
130. Wicker, C., Scheele, G. A., and Puigserver, A., Pancreatic adaptation to dietary lipids
is mediated by changes in lipase mRNA, Biochirnie, 70, 1277, 1988.
131. Wicker, C. and Puigserver, A., Expression of rat pancreatic lipase gene is modulated
by a lipid-rich diet at a transcriptional level, Biochern. Biophys. Res. Cornrnun., 166,
358, 1990.
132. Kern, H. F., Rausch, U., and Scheele, G., Regulation of gene expression in pancreatic
adaptation to nutritional substrates or hormones, Gut, 28, 89, 1987.
133. Beaudoin, A. R., Grondin, G., St-Jean, P., Vacherear, A., Cabana, C., and Grossman,
A., Steroids and secretory function of the exocrine pancreas, Endocrinology, 119, 2106,
1986.
134. Duan, R.-D. and Erlanson-Aibertsson, C., The anticoordinate changes of pancreatic
lipase and colipase activity to amylase activity induced by adrenalectomy in normal and
diabetes rats, Int. J. Pancreatol., 6, 271, 1990.
135. Duan, R.-D., Poensgen, J., and Erlanson-Albertsson, C., Adrenalectomy anticoor-
dinately changes the synthesis of rat pancreatic enzymes, Digestion, 43, 138, 1989.
136. Wicker, C., Puigserver, A., Rausch, U., Scheele, G., and Kern, H., Multiple-level
caerulein control of the gene expression of secretory proteins in the rat pancreas, Eur.
J. Biochern., 151,461, 1985.
137. Puigserver, A., Wicker, C., and Gaucher, C., Aspects moleculaires de !'adaptation
des enzymes pancreatiques et intestinales au regime alimentaire, Reprod. Nutr. Dev., 25,
787, 1985.
138. Mickel, F. S., Weidenbach, F., Swarovsky, B., LaForge, K. S., and Scheele, G. A.,
Structure of the canine pancreatic lipase gene, J. Bioi. Chern., 264, 12895, 1989.
139. Howard, G., Keller, P. R., Johnson, T. M., and Meisler, M. H., Binding of a
pancreatic nuclear protein is correlated with amylase enhancer activity, Nucleic Acids
Res., 17, 8185, 1989.
140. Meister, A., Weinrich, S. L., Nelson, C., and Rutter, W. J,, The chymotrypsin
enhancer core. Specific factor binding and biological activity, J. Bioi. Chern., 264, 20744,
1989.
141. Nir, U., Walker, M.D., and Rutter, W. J., Regulation of rat insulin I gene expression:
evidence for negative regulation in nonpancreatic cells, Proc. Natl. Acad. Sci. U.S.A.,
83, 3180, 1986.
142. Zhang, Z. X., Kumar, V., Rivera, R. T., Chisholm, J,, and Biswas, D. K., Cis-
acting negative regulatory element of prolactin gene, J. Bioi. Chern., 265, 4785, 1990.
143. Osborn, L., Rosenberg, M. P., Keller, S. C., Ting, C.-N., and Meisler, M. H.,
Insulin response of a hybrid amylase/CAT gene in transgenic mice, J. Bioi. Chern., 263,
16519, 1988.
144. Selden, R. F., Skoskiewicz, M. J,, Burke Howie, K., Russell, P. S., and Goodman,
H. M., Regulation of human insulin gene expression in transgenic mice, Nature (London),
321,525, 1986.
Chapter 4
TABLE OF CONTENTS
I. Introduction ....................................................... 92
0-8493-6961-41931$0.00 + $.50
© 1993 by CRC Pres>, Inc. 91
92 Nutrition and Gene Expression
I. INTRODUCTION
gut region by altering the rate of stem cell division and the rate of renewal
of the mucosal lining. Such alterations in mucosal mass are termed adaptive
growth of the GI tract. For example, fasting results in hypoplasia and atrophy
of the gut mucosa while hyperphagia results in mucosal hyperplasia. 4 •5 It is
not clear whether alterations in nutrient load or type affect the rate of turnover
of GI endocrine cells to the same degree as other mucosal cell types. Indeed,
there is evidence that some GI endocrine cells tum over much more slowly
than other epithelial cells. 6 Changes in the concentrations of GI hormone
mRNAs may therefore be secondary to differential effects of nutrient status
on relative mass of endocrine and "non-endocrine" cell types and may not
reflect the effect of nutrient on gene expression per se. Histological analyses
of the mucosa performed in parallel with quantification of GI hormone mRNAs
could help discern if apparent alterations in the levels of GI hormone mRNAs
are secondary to alterations in· relative mass of endocrine cells. To date, such
combined studies have been performed very infrequently.
Chemical and mechanical stimuli-Ingested nutrients present a complex
stimulus to the GI tract. Mechanical stimulation results from distention of the
gut by food. Chemical stimulation occurs via nutrients within the lumen of
the GI tract or via altered concentrations of absorbed nutrients in the circu-
lation. Gut responses to these nutritional stimuli can involve long neural
reflexes mediated by the central nervous system and short neural reflexes
mediated by the intrinsic neuronal plexuses in the gut wall. Mechanical and
chemical stimuli may stimulate secretion of GI hormones into the circulation
or local secretion into the extracellular fluid. Nutrient effects on GI hormone
gene expression therefore may be mediated by direct effects of nutrients on
the GI endocrine cells, by indirect effects of neuronal reflexes, or by indirect
effects of other GI hormones acting in an endocrine and paracrine manner.
Thus, careful experimental design is required to elucidate precisely which of
these multiple nutrient-mediated events underlies a nutrient-dependent change
in GI hormone gene expression.
Hormone expression-A given GI hormone may be expressed in mul-
tiple regions of the gut or accessory tissues. A nutrient-mediated effect ob-
served in one gut region or cell type may not necessarily reflect effects in
other regions or cells.
Cis- and trans-acting factors-Cell-specific and regulated expression of
most genes appear to be determined by cis-acting regulatory elements com-
prised of DNA sequences that flank the transcribed portions of the gene or
lie within the structural gene. 7 Trans-acting factors, usually proteins, bind to
these DNA regulatory elements and alter the rate of gene transcription. 7 It is
increasingly clear that steady-state levels of many mRNAs are regulated by
posttranscriptional mechanisms, probably via effects on RNA binding factors
that influence mRNA stability and turnover. 8 Ultimately, an understanding
of nutrient regulation of GI hormone-gene expression and synthesis will re-
Lund 95
A. BACKGROUND
Gastrin is synthesized primarily in G cells of the antrum of the stomach
and exists as two major forms of 34 (G34) and 17 (G 17) amino acids. 1 The
two forms derive from differential processing of a single precursor encoded
by a single gene and mRNA. 1 Gastrin has two major physiological roles, the
stimulation of gastric acid secretion and stimulation of mucosal growth in the
acid-secreting portion of the stomach. 1 Since gastric acid secretion is a major
determinant, if not causative factor in peptic ulcer disease, understanding the
molecular basis of nutritional effects on gastrin synthesis and release has
implications for understanding the etiology and pathophysiology of peptic
ulcer.
Fasting results in decreases in plasma gastrin, decreases in antral gastrin
concentrations, and a decrease in antral G cell density. 1•9 •10 These effects are
reversed by refeeding. 1 •9 Gastrin is released into the circulation in response
to food ingestion. 1 The chemical composition of food influences gastrin re-
lease. Proteins, digestion products of proteins, and individual amino acids
are particularly strong stimulants of gastrin secretion. 1 • 11 • 12 Neural reflexes
also influence gastrin release. 1 Recent studies, conducted primarily in rat and
dog, suggest that the effects of fasting and nutrient-induced changes in cir-
culating and antral gastrin concentrations are mediated at the level of gastrin
mRNA (Figure 1). These findings therefore indicate that nutrient status in-
fluences gastrin gene transcription of mRNA stability.
circulating
metabolites
and nutri en ts
(? )
l l' Ur ~ l
nm lr o l
I'.' I
Gastrin
I f-A
Ch:
pep tones
cell amino acid s
amin es
~ G RP
FIGURE l. Schematic to summarize nutrient effects on the gastrin cell that are implicated in
regulation of gastrin gene ex.pression: + = positive effect; - = negative effect; ? = indirect
evidence for an effect. Peptones, amino acid s. and amines appear to act via the lumen either by
cell surface receptors or after uptake into the G cell as indicated.
endings in proximity to the G cell. 1 To date, little is known about the role
of neural influences in nutrient effects on gastrin gene expression (Figure 1).
GASTRI N
GASTIU
protein or other nutrients on the CCK producing cell also cannot be excluded
(Figure 2).
Another candidate mediator of nutrient-mediated effects on CCK gene
transcription is GRP which is known to stimulate CCK secretion and is
localized in efferent nerve endings throughout the gastrointestinal tract. 1 Evi-
dence againt GRP as a mediator of increased CCK gene transcription in
response to luminal nutrient stems from studies demonstrating that intravenous
bombesin (the amphibian counterpart of mammalian GRP) transiently in-
creases plasma levels of CCK but does not influence CCK mRNA levels in
duodenum. 45 This finding is of interest in that it indicates that increases in
CCK secretion are not obligatorily linked to increases in CCK mRNA. 45 On
the other hand, a role for GRP in nutrient effects to increase CCK mRNA
cannot entirely be excluded since intravenous bombesin may not necessarily
elicit the same effects as activation of GRP efferents by nutrients within the
intestinal lumen.
The soybean trypsin inhibitor model is a well defined model to study
effects of luminal protein on CCK mRNA since overall nutrient status of the
animals is not significantly affected. 44 Using this model Kanayama and Liddle
have demonstrated recently that somatostatin can inhibit the induction of CCK
mRNA in response to soybean trypsin inhibitor. 46 This raises the possibility
that, as with gastrin, a component of nutrient effects to stimulate CCK gene
transcription may be mediated by removal of a negative influence of so-
matostatin (Figure 2). To date, however, there are no data to directly support
this possibility. Thus, the precise mediators of nutrient effects on CCK gene
transcription are not defined (Figure 2).
opment of useful model systems and progress in characterizing the CCK cis-
regulatory elements.
A. BACKGROUND
Somatostatin is a tetradecapeptide originally isolated from the hypothal-
amus. 49 It is now established that the GI tract and pancreas are major sites
of somatostatin synthesis. 1•50•51 In stomach and pancreas, like brain, the pre-
dominant form of somatostatin is a tetradecapeptide whereas a 28 amino acid
form predominates in the intestine. 5 1-53 These two forms of somatostatin derive
from differential processing of a precursor encoded by a single somatostatin
gene and mRNA. 54- 56 Somatostatin is synthesized in D cells of the pancreatic
islets and D cells within the mucosa of the stomach, small intestine, and large
intestine. 1 •50- 53 •57•58 In some species, somatostatin is also synthesized by neu-
rons within the intrinsic neuronal plexuses of the gut. 1•58
Somatostatin has widespread actions that have been reviewed elsewhere
and are beyond the scope of this chapter. 1 •50 In general, somatostatin is an
inhibitory peptide. In the GI tract, somatostatin acts to inhibit gastric acid
secretion, pancreatic exocrine secretion, and the secretion of hormones from
pancreatic islet and GI endocrine cells. 1 Available evidence suggests that rather
than acting in an endocrine manner, gastrointestinal somatostatin may act in
a paracrine manner to modulate target cell function, possibly via release of
somatostatin from cytoplasmic processes projecting from the D cells onto
target cells. 59
A number of studies suggest effects of nutrients on somatostatin concen-
trations in plasma and somatostatin content of different regions of the GI
tract, especially stomach and pancreas. 1.3.so The interpretation of such findings
is complicated by the multiple cellular sites of somatostatin biosynthesis. In
addition, since somatostatin acts in a paracrine manner, tissue content of
somatostatin may remain unchanged even in the face of local alterations in
somatostatin synthesis or secretion by specific cell populations. As well as
providing information about nutrient regulation of somatostatin gene expres-
sion, analyses of nutrient effects on somatostatin mRNA content in different
regions of GI tract may therefore aid in the interpretation of nutrient effects
on D cell function.
1-30
II G.OCAOON G.P-1 (1-37) IP-11 I PROGLUCAGON
G.ICENTIN 69
L_G._~_7~--~~~~I ____G._P-1
__ 1 __ ~
GUT
I OXYNTOM:XlU.IN
PANCREAS
Isolated fetal rat intestinal cells in culture (FRIC cells) are another promising
system which has already been used to demonstrate positive regulation of
somatostatin gene expression by cyclic AMP and phorbol esters. 64 Nonethe-
less, progress in elucidating nutrient effects on gastrointestinal somatostatin
gene expression will require a combination of information derived from these
in vitro systems and in vivo approaches such as transgenic animals.
VI. GLUCAGON/ENTEROGLUCAGON
A. BACKGROUND
The proglucagon gene is a particularly interesting and complex system
with regard to nutrient control of expression. Mammals possess a single
glucagon gene and mRNA, but the encoded precursor, proglucagon, is a
polyprotein precursor that contains three glucagon related peptides as well as
other peptides (Figure 3). 1•65 -68 A common proglucagon precursor is processed
very differently in the pancreas and intestine to result in different biologically
active peptides with different biological activities. 69 - 73 The structure of the
common pancreatic and intestinal proglucagon is shown in Figure 3. Proglu-
cagon contains the 29 amino acid pancreatic glucagon in the midregion with
amino acid extensions at amino and carboxyl termini. A long carboxyl terminal
extension contains two glucagon-like peptides (GLPs) with structural ho-
mology to glucagon, and these have been termed GLP-1 and GLP-11. Spacer
or intervening peptides (IPs) separate glucagon from GLP-1 and GLP-1 from
GLP-11.
In the pancreas, proglucagon is synthesized in pancreatic A cells. 1·74
Products of posttranslational processing of proglucagon in the pancreas are a
29 amino acid pancreatic glucagon, a 30 amino acid peptide comprised of
the amino terminus of proglucagon, and a "Major proglucagon fragment"
(Figure 3). 69- 73 The only product of pancreatic proglucagon processing with
defined biological activity is glucagon. 74 The primary biological role of pan-
creatic glucagon is well established. Glucagon regulates metabolic pathways
106 Nutrition and Gene Expression
in liver and adipose tissue and maintains euglycemia during fasting or nutrient
perturbation. 74 Pancreatic glucagon is secreted into the circulation in response
to fasting, decreases in plasma glucose, and increases in plasma amino acids
and neural influences. 71 ·74 Pancreatic glucagon secretion is suppressed by high
plasma glucose. 71 •74
In the intestine, proglucagon is processed very differently than in the
pancreas. The products of intestinal proglucagon are enteroglucagons (alter-
nately named glucagon-like immunoreactants [GUs]), GLP-I (73-77), GLP-
11, and IP-11 (Figure 3). 69- 73 Enteroglucagons or GUs include glicentin and
oxyntomodulin (Figure 3). Glicentin is composed of the first 69 amino acids
of proglucagon and includes the entire sequence of glucagon in the midregion
flanked by amino acid extensions at both amino and carboxyl termini (Figure
3). Oxyntomodulin consists of glucagon with a hexapeptide extension at the
carboxyl terminus (Figure 3). At physiological concentrations, glicentin and
oxyntomodulin appear not to share the biological actions of pancreatic glu-
cagon but appear to act primarily to inhibit gastric acid secretion and thus
are enterogastrones. 1 •71 •75 Of the other intestinal pro glucagon derived peptides,
only GLP-I has been extensively characterized in terms of biological activity.
GLP-I 7-37 is the primary form of GLP-I in intestine and is a potent stimulant
of insulin secretion and insulin gene expression. 71 •76 •77 The biological roles
of GLP-11 and IP-11 are not defined. Based on observations that plasma
concentrations of enteroglucagons are elevated in most situations of increased
cell proliferation of the intestinal mucosa, the enteroglucagons (glicentin/
oxyntomodulin) or other products of intestinal proglucagon processing are
candidate enterotrophic hormones. 68 •78
Products of intestinal proglucagon-derived peptides are secreted in re-
sponse to different stimuli than pancreatic products. Available evidence sug-
gests that all of the intestinal products of proglucagon are secreted in paral-
lel. 69- 72 Plasma enteroglucagon levels are reduced by fasting and elevated
after food ingestion. 71 Intrajejunal glucose or carbohydrate and fatty acids
are especially strong stimulants of enteroglucagon secretion. 1•79 The secretion
of enteroglucagons in response to luminal glucose but suppression of pan-
creatic glucagon by high plasma glucose indicates that the pancreatic A cell
and intestinal L cell have very different responses to glucose.
To date, the regulation of glucagon gene expression by nutrients has not
been studied extensively. The molecular basis for nutrient control of proglu-
cagon gene expression is likely to be both complex and interesting given the
different secretory responses of two different proglucagon expressing cell
types, the A cell and the L cell, in response to nutritional stimuli.
.
""' • PANCREAS
"" • • INTESTINE
% ..
FED FASTED
pM/t
.
""' • PANCREAS
• INTESTINE
FED FASTED
pt.l/g
FED FASTED
FIGURE 4. Proglucagon mRNA and peptide in ad libitum fed and fasted rats. (A) Proglucagon
mRNA in pancreas and small intestine. Proglucagon mRNA was analyzed in poly A+ RNA by
Northern hybridization and abundance assessed as concentration per f.Lg poly A+ RNA. Shown
is the abundance in pancreas or intestine of fasted animals expressed as % of abundance of fed
animals; (B) Glucagon immunoreactivity in pancreas and intestine of fed and fasted rats; (C) Plasma
enteroglucagon immunoreactivity in fed and fasted rats. * = significant difference in fasted
animals vs. fed animals (p < 0.05).
total mRNA extracted from pancreas (Figure 4). In contrast, Chen et a!.,
using in situ hybridization histochemistry, observed a significant increase in
pancreatic proglucagon mRNA in response to a 4-d fast that induced signif-
icant glucopenia. 80 It is possible that the different durations of fasting account
for these different results. However, it also seems possible that the different
results reflect the different methodologies utilized. Our inability to detect a
change in pancreatic proglucagon mRNA by Northern hybridization may be
due to the fact that pancreatic A cells represent a very small proportion of the
108 Nutrition and Gene Expression
breeding of two transgenic lines. 91 The transgenic animals used for cross-
breeding were ones with an insulin promoter used to drive the SV40 large T
antigen gene and ones with the insulin promotor linked to mouse polyoma
small T antigen. The SV40 transgenics develop pancreatic insulinoma but not
enteroendocrine tumors. 91 The polyoma transgenics do not develop tumors in
pancreas or gut. 91 On crossbreeding, however, a large proportion of animals
develop enteroendocrine tumors that express a spectrum of GI hormones.
STC-1 cell lines derived from the enteroendocrine tumors express a wide
range of GI hormone. The molecular basis for the development of enteroen-
docrine tumors in these crossbred mice is of itself very interesting since
enteroendocrine tumors are relatively rare. For those working in the field of
GI hormone expression, the cell lines derived from these tumors may represent
a very important and unique tool for future studies.
ACKNOWLEDGMENTS
This work was supported by NIH grant DK 40247. Thanks are expressed
to Dr. Rodger Liddle for extremely useful input and discussion. Blanche Jones
provided invaluable editorial and secretarial assistance.
REFERENCES
I. Walsh, J, H., Gastrointestinal hormones, in Physiology of the Gastrointestinal Tract,
Johnson, L. R., Ed., Raven Press, New York, 1987, 181.
2. Brown, J, C., Mcintosh, C. H. S., and Pederson, R. A., The gastrointestinal peptides
and nutrition, Can. J. Physiol. Pharmacal., 61, 282, 1982.
3. Yamada, T., Gut hormone release induced by food ingestion, Am. J. Clin. Nutr., 42,
1033, 1985. (DB 44)
4. Dowling, R. H., Small bowel adaptation and its regulation, Scand. J. Gastroenterol.,
74, 53, 1982.
5. Lipscomb, H. L. and Sharp, J, G., Effects of reduced food intake on morphometry
and cell production in the small intestine of the rat, Virchows Arch., 41, 285, 1982.
6. Fujimoto, S., Kimoto, K., Yamashita, S., Kawai, K., and Hattori, T., and Fugita,
S., Tritiated thymidine autoradiographic study on origin and renewal of gastrin cells in
antral area of hamsters, Gastroenterology, 79, 785, 1985.
7. Mitchell, P. J. and Tjian, R., Transcriptional regulation in mammalian cells by sequence-
specific DNA binding proteins, Science, 245, 371, 1989.
8. Raghowr, R., Regulation of messenger RNA turnover in eukaryotes, Trends Biochem.
Sci., 12, 358, 1987.
9. Lichtenberger, L. M., Lechago, J,, and Johnson, L. R., Depression of antral and
serum gastrin concentration by food deprivation in the rat, Gastroenterology, 68, 1473,
1975.
112 Nutrition and Gene Expression
10. Schwarting, H., Koop, H., Gellert, G., and Arnold, R., Effect of starvation on
endocrine cells in the rat stomach, Regul. Pep!., 14, 33, 1986.
II. Taylor, I. L., Byrne, W. J., Christie, D. L., Ament, M. E., and Walsh, J. H., Effect
of individual L-amino acids on gastric acid secretion and serum gastrin and pancreatic
polypeptide release in humans, Gastroenterology, 83, 272, 1982.
12. Lichtenberger, L. M., Importance of food in the regulation of gastrin release and
formation, Am. J. Physiol., 243, G429, 1982.
13. Wu, V., Sumii, K., Tari, A., Sumii, M., and Walsh, J. H., Regulation of rat antral
gastrin and somatostatin gene expression during starvation and after refeeding, Gastro-
enterology, 101, 1552, 1991.
14. Wu, S. V., Sumii, K., Tari, A., Mogard, M., and Walsh, J. H., Regulation of gastric
somatostatin gene expression, Metabolism, 39, 125, 1990. (DB41)
15. Brand, S. J. and Stone, D., Reciprocal regulation of antral gastrin and somatostatin
gene expression by omeprazole-induced achlorhydria, J. Clin. Invest., 82, 1059, 1988.
16. Schusdziarra, V., Harris, V., Conlon, J. M., Arimura, A., and Unger, R., Pancreatic
and gastric somatostatin release in response to intragastric and intraduodenal nutrients
and HCI in the dog, J. Clin. Invest., 62, 509, 1978.
17. Bloom, S. R., Mortimer, C. H., and Thorner, M. 0., Inhibition of gastrin and gastric
acid secretion by growth hormone release inhibitory hormone, Lancet, I, 1106, 1974.
18. Harty, R. F., Maico, D. G., and McGuigan, J. E., Somatostatin inhibition of basal
and carbachol stimulated gastrin release in rat antral organ culture, Gastroenterology,
81' 707, 1981.
19. Karnik, P. S., Monahan, S. J., and Wolfe, M. M., Inhibition of gastrin gene expression
by somatostatin, J. Clin. Invest., 83, 367, 1989.
20. Karnik, P. S. and Wolfe, M. M., Somatostatin stimulates gastrin mRNA turnover in
dog antral mucosa, J. Bioi. Chern., 265, 2550, 1990.
21. Wu, S. V., Sumii, K., and Walsh, J. H., Studies of regulation of gastrin synthesis and
post-translational processing by molecular biology approaches, Ann. N.Y. Acad. Sci.,
597, 17, 1990. (DB 18)
22. Lichtenberger, L. M., Forssman, W. G., and Ito, S., Functional responsiveness of
an isolated and enriched fraction of rodent gastrin cells, Gastroenterology, 79, 447, 1980.
23. Theill, L. E., Wiborg, 0., and Vuust, J., Cell-specific expression of the human gastrin
gene: evidence for a control element located downstream of the TATA box, Mol. Cell.
Bioi., 7, 4329, 1987.
24. Brand, S. J. and Wang, T. C., Gastrin gene expression and regulation in rat islet cell
lines, J. Bioi. Chern., 263, 16597, 1988. (DB 106)
25. Wang, T. C. and Brand, S. J., Islet cell-specific regulatory domain in the gastrin
promoter contains adjacent positive and negative DNA elements, J. Bioi. Chern., 265,
8908, 1990. (DB63)
26. Merchant, J. L., Demediuk, B., and Brand, S. J., A GC-rich element confers epi-
dermal growth factor responsiveness to transcription from the gastrin promoter, Mol.
Cell. Bioi., II, 2686, 1991. (DB61)
27. Godley, J. M. and Brand, S. J., Regulation of the gastrin promoter by epidermal growth
factor and neuropeptides, Proc. Nat/. Acad. Sci. U.S.A., 86, 3036, 1989. (DB62)
28. Takahashi, Y., Fukushige, S., Murotsu, T., and Matsubara, K., Structure of human
cholecystokinin gene and its chromosomal location, Gene, 50, 353, 1986.
29. Deschenes, R. J., Lorenz, L. J., Haun, R. S., Roos, B. A., Collier, K. J., and
Dixon, J., Cloning and sequence analysis of a eDNA encoding rat preprocholecystokinin,
Proc. Nat!. Acad. Sci. U.S.A., 81, 726, 1984.
30. Gubler, U., Chua, A. 0., Hoffman, B. J., and Collier, K. J., Cloned eDNA to
cholecystokinin mRNA predicts an identical preprocholecystokinin in pig brain and gut,
Proc. Nat/. Acad. Sci. U.S.A., 81, 4307, 1984.
Lund 113
31. Ivy, A. C. and Oldberg, E., A hom10ne mechanism for gallbladder contraction and
evacuation, Am. J. Physiol., 85, 381, 1928.
32. Harper, A. A. and Raper, H. S., Pancreozymin, a stimulant of secretion of pancreatic
enzymes in extracts of small intestine, J. Physiol., 102, 115, 1943.
33. Niederau, C., Luthen, R., Niederau, M., Strohmeyer, G., Ferrell, L. D., and Grendell,
J, H., Effects of long term CCK stimulation and CCK blockade on pancreatic and
intestinal growth, morphology and function, Digestion, 46, 217, 1990.
34. Liddle, R. A., Goldfine, I. D., Rosen, M. S., Taplitz, R. A., and Williams, J. A.,
Cholecystokinin bioactivity in human plasma, J. Clin. Invest., 75, 1144, 1985.
35. Konturek, S. J., Radecki, T., Thor, P., and Dembinski, A., Release of cholecystokinin
by amino acids, Proc. Soc. Exp. Bioi. Med., 143, 305, 1973.
36. Meyer, J, H. and Jones, R. S., Canine pancreatic responses to intestinally perfused fat
and products of fat digestion, Am. J. Physiol., 226, 1178, 1974.
37. Liddle, R. A., Green, G. M., Conrad, C. K., and Williams, J. A., Proteins but not
amino acids, carbohydrates, or fats stimulate cholecystokinin secretion in the rat, Am.
J. Physio/., 251, G243, 1986.
38. Liddle, R. A., Goldfine, I. D., and Williams, J. A., Bioassay of plasma cholecystokinin
in rats: effects of food, trypsin inhibitor, and alcohol, Gastroenterology, 87, 542, 1984.
39. Iwai, K., Fushiki, T., and Fukuoka, S.-1., Pancreatic enzyme secretion mediated by
a novel peptide: monitor peptide hypothesis, Pancreas, 3, 720, 1988.
40. Iwai, K., Fukuoka, S.-1., Fushiki, T., Tsujikawa, M., Hirose, M., Tsunasawa, S.,
and Sakiyama, F., Purification and sequencing of a trypsin-sensitive cholecystokinin
releasing peptide from rat pancreatic juice. Its homology with pancreatic secretory trypsin
inhibitor, J. Bioi. Chern., 262, 8956, 1987.
41. Fukuoka, S. and Scheele, G. A., Rapid and selective cloning of monitor peptide, a
novel cholecystokinin-releasing peptide, using minimal amino acid sequence and the
polymerase chain reaction, Pancreas, 5, I, 1990.
42. Greenstein, R. J., Isola, L., and Gordon, J., Differential cholecystokinin gene expres-
sion in brain and gut of the fasted rat, Am. J. Med. Sci., 299, 32, 1990.
43. Kanayama, S. and Liddle, R. A., Influence of food deprivation on intestinal chole-
cystokinin and somatostatin, Gastroenterology, 100, 909, 1991. (DB58)
44. Liddle, R. A., Carter, J.D., and McDonald, A. R., Dietary regulation of rat intestinal
cholecystokinin gene expression, J. Clin. Invest., 81, 2015, 1988. (DB40)
45. Kanayama, S. and Liddle, R. A., Regulation of intestinal cholecystokinin and so-
matostatin mRNA by bombesin in rats, Am. J. Physiol., 261, G71, 1991.
46. Kanayama, S. and Liddle, R. A., Somatostatin regulates duodenal cholecystokinin and
somatostatin messenger RNA, Am. J. Physiol., 258, 1990, G358. (DB22)
47. Haun, R. S. and Dixon, J, E., A transcriptional enhancer essential for the expression
of the rat cholecystokinin gene contains a sequence identical to the - 296 element of the
human c-fos gene, J. Bioi. Chern., 265, 15455, 1990.
48. Monstein, H.-J. and Folkesson, R., Phorbo\12-myristate-13-acetate (PMA) stimulates
a differential expression of cholecystokinin (CCK) and c-fos mRNA in a human neuro-
blastoma cell line, FEBS, 293, 145, 1991.
49. Brazeau, P., Vale, W., Burgos, R., Ling, N., Butcher, M., Riveir, J., and Guillemin,
R., Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary
growth hormone, Science, 179, 77, 1973.
50. Newman, J. B., Lluis, F., and Townsend, C. M, Jr., Somatostatin, in Gastrointestinal
Endocrinology, Thompson, J. C., Greeley, G. H., Jr., Rayford, P. L., and Townsend,
C. M., Jr., Eds., McGraw-Hill, Toronto, 1987,286.
51. Patel, Y. C. and Reichlin, S., Somatostatin in hypothalamus, extrahypothalamic brain,
and peripheral tissues of the rat, Endocrinology, 102, 523, 1978.
52. Pradayrol, L., Jornvall, H., Mutt, V., and Ribet, A., N-terminally extended somato-
statin: the primary structure of somatostatin-28, FEBS, 109, 55, 1980.
114 Nutrition and Gene Expression
53. Trent, D. F. and Weir, G. C., Heterogeneity of somatostatin-like peptides in rat brain,
pancreas, and gastrointestinal tract, Endocrinology, 108, 2033, 1981.
54. Shen, L.-P. and Rutter, W., Human somatostatin I: sequence of the eDNA, Proc. Nat!.
Acad. Sci. U.S.A., 224, 168, 1984.
55. Funckes, C. L., Minth, C. D., Deschenes, R., Magazin, M., Tavianini, M. A.,
Sheets, M., Collier, K., Weith, H. L., Aron, D. C., Roos, B. A., and Dixon, J. E.,
Cloning and characterization of a mRNA-encoding rat preprosomatostatin, J. Bioi. Chern.,
258, 8781, 1983.
56. Goodman, R. H., Aron, D. C., and Roos, B. A., Rat pre-prosomatostatin: structure
and processing by microsomal membranes, J. Bioi. Chern., 258, 5570, 1983.
57. Hofler, H., Childers, H., Montminy, M. R., Goodman, R. H., Leehan, R. M.,
DeLellis, R. A., Tischler, A. S., and Wolfe, H. J., Localization of somatostatin mRNA
in the gut, pancreas and thyroid gland of the rat using antisense RNA probes for in situ
hybridization, Acta Histochem., 34, 101 (Suppl.), 1987.
58. Vinik, A. I., Gaginella, T. S., O'Dorisio, T. M., Shapiro, B., and Wagner, L., The
distribution and characterization of somatostatin-like immunoreactivity in epithelial cells,
submucosa, and muscle of the rat stomach and intestine, Endocrinology, I 09, 1921, 1981.
59. Larrson, L.-1., Goltermann, N., De Magistris, L., Rehfeld, J. F., and Schwartz,
T. W., Somatostatin cell processes as pathways for paracrine secretion, Science, 205,
1393, 1979.
60. Chiba, T., Park, J., and Yamada, T., Biosynthesis of somatostatin in canine fundic
D cells, J. Clin. Invest., 81, 282, 1988.
61. Park, J., Chiba, T., Yokotani, K., DelValle, J., and Yamada, T., Somatostatin
receptors on canine fundic D-cells: evidence for autocrine regulation of gastric somato-
statin, Am. J. Physiol., 257, 0235, 1989.
62. Montminy, M. R., Sevarino, K. A., Wagner, J. A., Mandel, G., and Goodman,
R. H., Identification of a cyclic-AMP-responsive element within the rat somatostatin
gene, Proc. Nat/. Acad. Sci. U.S.A., 83, 6682, 1986.
63. Powers, A. C., Tedeschi, F., Wright, K. E., Chan, J. S., and Habener, J. F.,
Somatostatin gene expression in pancreatic islet cells is directed by cell-specific DNA
control elements and DNA-binding proteins, J. Bioi. Chern., 264, 10048, 1989.
64. Brubaker, P. L., Drucker, D. J., and Greenberg, G. R., Synthesis and secretion of
somatostatin-28 and -14 by fetal rat intestinal cells in culture, Am. J. Physiol .. 258,
0974, 1990.
65. Habener, J. F., Goodman, R. H., and Lund, P. K., Complementary DNAs encoding
precursors of glucagon and somatostatin, in Biogenetics of Neurohormonal Peptides,
Hakanson, R. and Thorell, J., Eds .. Academic Press, San Diego, 1985, 47.
66. Drucker, D. J. and Brubaker, P. L., Glucagon biosynthesis in fetal rat intestine,
Biomed. Res., 9 (Suppl. 3), 29, 1988.
67. Novak, U., Wilks, A., Buell, G., and McEwen, S., Identical mRNA for preproglucagon
in pancreas and gut, Eur. J. Biochem., 164, 553, 1987.
68. Hynes, M. A., Shiow-Lian, J., Ulshen, M. H., Simmons, J. G., and Lund, P. K.,
Characterization, localization and regulation of extra pancreatic proglucagon mRNAs,
Biomed. Res., 9 (Suppl. 3), 147, 1988.
69. Moody, A. J., Holst, J., Thim, L., and Jensen, S., Relationship of glicentin to
proglucagon and glucagon in the porcine pancreas, Nature (London), 289, 514, 1981.
70. Patzelt, C. and Chiltz, E., Conversion of proglucagon in pancreatic alpha cells: the
major endproducts are glucagon and a single peptide, the major proglucagon fragment,
that contains two glucagon-like sequences, Proc. Nat!. Acad. Sci. U.S.A., 81, 5007,
1984.
71. Holst, J. J., Orskov, C., Olsen, J., Buhl, T., Schjoldager, B., and Kofod, H.,
Secretion and effects of the naturally occurring products of proglucagon, Biomed. Res.,
9 (Suppl. 3), 181, 1988.
Lund 115
72. Mojsov, S., Heinrich, G., Wilson, I. B., Ravazzola, M., Orci, L., and Habener,
J. F., Preproglucagon gene expression in pancreas and intestine diversifies at the level
of post-translational processing, J. Bioi. Chern., 261, 11880, 1986.
73. Orskov, C., Holst, J., Knuhtsen, S., Baldissera, F. G. A., Poulsen, S. S., and Nielsen,
0. V., Glucagon-like peptides GLP-1 and GLP-2, predicted products of the glucagon
gene are secreted separately from pig small intestine but not pancreas, Endocrinology,
119, 1467, 1986.
74. Unger, R. and Oric, L., Physiology and pathophysiology of glucagon, Physiol. Rev.,
56, 778, 1976.
75. Jarrousse, C., Audousset-Puech, M.-P., Dubrasquet, M., Niel, H., Martinez, J.,
and Bataille, D., Oxyntomodulin (glucagon-37) and its C-terminal octapeptide inhibit
gastric acid secretion,. FEES Lett., 188, 81, 1985.
76. Holst, J. J., Orskov, C., Vagn Hielsen, 0. V., and Schwartz, T. W., Truncated
glucagon-like peptide I, an insulin-releasing hormone from the distal gut, FEES Lett.,
211, 169, 1987.
77. Drucker, D. J., Philippe, J., Mojsov, S., Chick, W. L., and Habener, J. F., Glucagon-
like peptide I stimulates insulin gene expression and increases cyclic AMP levels in a rat
islet cell line, Proc. Nat/. Acad. Sci. U.S.A., 84, 3434, 1987.
78. Bloom, S. R. and Polak, J. M., The hormonal pattern of intestinal adaptation: a major
role for enteroglucagon, Scand. J. Gastroenterol., 74 (Suppl.), 93, 1982.
79. Holst, J. J., Christiansen, J., and Kohl, C., The enteroglucagon response to intrajejunal
infusion of glucose, triglycerides, and sodium chloride, and its relation to jejunal inhibition
of gastric acid secretion in man, Scand. J. Gastroenterol., II, 297, 1976.
80. Chen, L., Komiya, I., Inman, L., O'Neill, J., Appel, M., Alam, T., and Unger,
R. H., Effects of hypoglycemia and prolonged fasting on insulin and glucagon gene
expression, J. Clin. Invest., 84, 711, 1989.
81. Chen, L., Komiya, I., Inman, L., McCorkle, K., Alam, T., and Unger, R. H.,
Molecular and cellular responses of islets during perturbations of glucose homeostasis
determined by in situ hybridization histochemistry, Proc. Nat/. Acad. Sci. U.S.A., 86,
1367, 1989.
82. Rountree, D. B., Ulshen, M. H., Selub, S., Fuller, C. R., Bloom, S. R., Ghatei,
M. A., and Lund, P. K., Rapid, nutrient independent increases in proglucagon and
ODC mRNAs after jejuno-ileal resection, Gastroenterology, in press.
83. Taylor, R. G., Verity, K., and Fuller, P. J., Ileal glucagon gene expression: ontogeny
and response to massive small bowel resection, Gastroenterology, 99, 724, 1990.
84. Buchan, A.M. J., Griffiths, C. J., Morris, J. F., and Polak, J. M., Enteroglucagon
cell hyperfunction in rat small intestine after gut resection, Gastroenterology, 88, 8, 1985.
85. Sagor, G. R., Al-Mukntar, M. Y. T., Ghatei, M. A., Wright, N. A., and Bloom,
S. R., The effect of altered luminal nutrition on cellular proliferation and plasma con-
centrations of enteroglucagon and gastrin after small bowel resection in rat, Br. J. Surg.,
69, 14, 1982.
86. Grodsky, G. M. and Fanska, R. E., The in vitro perfused pancreas, Methods Enzyrnol.,
39, 364, 1975.
87. Philippe, J., Drucker, D. J., Chick, W. L., and Habener, J. F., Transcriptional
regulation of genes encoding insulin, glucagon, and angiotensinogen by sodium butyrate
in a rat islet cell line, Mol. Cell Bioi., 7, 560, 1987.
88. Philippe, J., Drucker, D. J., and Habener, J. F., Glucagon gene transcription in an
islet cell line is regulated via a protein kinase C-activated pathway, J. Bioi. Chern., 262,
1823, 1987.
89. Philippe, J., Drucker, D. J., Knepel, W., Jepeal, L., Misulovin, Z., and Habener,
J. F., Alpha-cell-specific expression of the glucagon gene is conferred to the glucagon
promoter element by the interactions of DNA-binding proteins, Mol. Cell. Bioi., 8,
4877, 1988.
116 Nutrition and Gene Expression
90. Efrat, S., Teitelman, G., Anwar, M., Ruggiero, D., and Hanahan, D., Glucagon
gene regulatory region directs oncoprotein expression to neurons and pancreatic A cells,
Neuron, I, 605, 1988.
91. Drucker, D. J. and Brubaker, P. J., Proglucagon gene expression is regulated by a
cyclic AMP-dependent pathway in rat intestine, Proc. Nat!. Acad. Sci. U.S.A., 86, 3953,
1989.
92. Rindi, G., Grant, S. G. N., Yiangou, Y., Ghatei, M. A., Bloom, S. R., Bautch,
V. L., Solcia, E, and Polak, J, M., Development of neuroendocrine tumors in the
gastrointestinal tract of transgenic mice, Am. J. Pathol., 136, 1349, 1990.
Chapter 5
TABLE OF CONTENTS
0·8493-6961-4/93/$0 ()()+$.50
© 1993 by CRC Pre», Inc. 117
118 Nutrition and Gene Expression
I. OVERVIEW
in ER content and structure existing from one cell type to another. While
both the rough and smooth ER possess considerable overlap in their protein
content, a number of proteins concerned with ribosomal docking and protein
processing are enriched in the roughER. 3.4 Prominent among lumenal resident
proteins, or reticuloplasmins, are endoplasmin or GRP94, GRP78 or BiP, and
protein disulfide isomerase. An oxidizing environment is thought to exist
within the lumen supporting protein disulfide bond formation and protein
hydroxylation. Functions commonly associated with the ER include early
protein processing, phospholipid biosynthesis, oxidative metabolism of hy-
drophobic molecules including many drugs, and storage of Ca2 + releasable
in response to extracellular stimuli. It is apparent, therefore, that the ER is
a highly complex organelle that infiltrates the cytoplasmic space of the cell
and interfaces with and produces much of the structural material comprising
other organelles. Overall, the ER possesses the structural and functional prop-
t:rties that would be expected for an organelle functioning in the integration
and coordination of major cellular processes.
A wealth of literature supports the central role of Ca2 + in intracellular
signaling related to stimulus-response coupling. 4 - 12 Prominent processes reg-
ulated by the cation include secretion, membrane transport and permeability,
glycogen metabolism, and muscle contraction. As intracellular-free Ca 2 + in
response to a stimulus rises severalfold from resting values near 0.1 f.LM, the
cation binds to Ca2 + receptor proteins, such as calmodulin and troponin C,
which then become capable of activating various enzymatic processes_ Con-
tributions of Ca 2 + to the free pool are derived from the extracellular fluid
and/or from intracellular sites of storage; release of cation from either source
is driven by concentration gradients on the order of 104 • Ca 2 + entry across
the plasmalemma involves voltage and/or ligand gated Ca2 + channels; 6 •8 Ca 2 +
efflux is generaHy achieved by a Na+ /Ca 2 + anti port and an active Ca2 + pump.
Despite much investigative effort, controversy continues regarding the relative
contribution of intracellular sequestered Ca 2 + from one cell type to another
in Ca2 + signaling, the localization and characteristics of Ca2 + stores, and
their function in maintaining Ca2 + homeostasis. s- 11 At least two stores of Ca2 +
are frequently proposed to exist, one of which is sensitive to release by inositol
I ,4,5-trisphosphate (IP 3 ) generated in response to hormonal action and another
which may support spontaneous oscillations in intracellular free Ca2 +. 7 - 12
With the discrediting of the mitochondrion as a prominent storage site for
Ca2 + in normal cells5 and the increasing appreciation of IP3 as a second
messenger triggering release of intracellular sequestered Ca2 + from the mi-
crosomal fraction, 7 the ER gained acceptance as a Ca2 + storing organelle.
Unfortunately the ER is sufficiently heterogeneous in terms of its biochemical,
functional, and morphological properties, that subsequent characterization of
its role in Ca2 + homeostasis proved to be elusive and controversial. 9 - 11 Cellular
contents of ER and IP3 -releasable Ca2 + varied widely from one cell type to
120 Nutrition and Gene Expression
TABLE 1
The Functional Status of the Endoplasmic Reticulum and Regulation
of Translational Initiation
to accumulate poly somes in response to Ca2 +. Ca2 + did not influence either
the recovery of amino acid incorporation or the induction of the heat shock
proteins (hsp) that occurred during the next several hours. Restoration of the
Ca 2 + requirement occurred by 24 h. More modest degrees of thermal stress
(41 °) resulted in the induction of hsp 68 without loss of Ca2 +-stimulated
initiation. Under this condition hsp 68 was synthesized in a Ca 2 +-dependent
manner.
Viral infections interrupt host cell protein synthesis by various mecha-
nisms while reprogramming the translational apparatus to synthesize viral
coded proteins. HEp-2 cells infected with HSV-1 were employed to examine
the Ca 2 + requirement for translation of viral mRNAs. 56 Early in viral infec-
tion, the synthesis of nearly all proteins, including viral (a) proteins, was
sensitive to inhibition by Ca2 + ionophore or EGTA. By 4 to 6 h after
infection, overall polypeptide synthesis in infected cells had become resistant
to depletion of Ca2 + stores. Specific viral mRNAs were readily detected in
polysomes, and the synthesis of viral polypeptides of early (13) and late (-y)
kinetic classes was found to be insensitive to the effects of Ca2 + depletion.
Production of mature forms of viral glycoproteins, however, was reduced by
treatment with EGTA and ionophore. Thus, productive infection of cells by
HS V-1, like heat shock or chemical stress, results in a modification that
overcomes the requirement of translational initiation for stored Ca2 +. Pro-
duction of most viral proteins may then proceed in a Ca 2 +-independent
fashion. A second, Ca2 +-sensitive step affecting the maturation (glycosyla-
tion and/or transport) of viral glycoproteins was also apparent which was not
bypassed during HSV -1 infection.
peptidyl-tRNA is bound in the A site and the peptidyl end is located in the
P site. Although details of the mechanism have not been elucidated, it is
likely that binding of the eEF-2·GTP complex promotes movement of the
mRNA and the anticodon portion of the peptidyl-tRNA into the ribosomal P
site accompanied by the hydrolysis of the nucleotide to GDP.
Cytoplasmic extracts of mammalian cells are routinely found to incor-
porate phosphate from ATP into a major 100-kDa species. 57 In 1983 Palfrey5 8
reported that such incorporation was enhanced by Ca 2 + and the ubiquitous
Ca2 + mediator protein calmodulin. Independently, Palfrey and Nairn 58 .s9 and
Guroff and colleagues60 •61 isolated the I 00-kDa substrate and employed it in
procedures for partial purification of the protein kinase responsible for the in
vitro phosphorylation. The kinase, which possessed a molecular mass of 130
to 140 kDa, was identified in a broad variety of tissues, required Ca2 + and
calmodulin for optimal activity, and was distinct in substrate specificity from
previously characterized forms of Ca 2 +/calmodulin-dependent protein ki-
nase. 59 •62 The enzyme, named Ca 2 +/calmodulin-dependent protein kinase III
by Nairn eta!. , 59 was unable to phosphorylate a variety of substrates utilized
by other protein kinases. Its 100-kDa substrate was not phosphorylated by
other protein kinases.
Identification of the 100-kDa substrate as eEF-2 was reported in early
1987 by Ryazanov 63 and later by Nairn and Palfrey. 64 Since the factor con-
stituted the only known substrate for the enzyme, Ryazanov proposed the
enzyme be named eEF-2 kinase. 65 Partially purified eEF-2 kinase was ob-
served to phosphorylate three threonine residues of eEF-2 from reticulocytes 66
with threonine 56 advanced as the primary phosphorylation site. 67 The com-
parable factor from chick embryo exhibited additional phosphorylation sites
on tyrosine and serine residues. 68 The threonine phosphorylation sites of
reticulocyte eEF-2 were localized to a region near the N terminus that may
constitute the ribosome binding site. 66 Phosphorylation was found to stabilize
the factor against the action of trypsin. 69 Dephosphorylation of phosphory Ia ted
factor in cell-free systems could be accomplished by incubation with various
phosphatases, with type 2A phosphatase being the most effective. 70. 72 Recent
functional studies support the hypothesis that phosphorylated eEF-2 cannot
sustain elongation. Okadaic acid, a tumor promoter that potently inhibits
protein phosphatases, was found by Redpath and Proud 71 to increase the net
phosphorylation of eEF-2 in reticulocyte lysates while provoking inhibition
of peptide chain elongation. In contrast the stimulation of elongation rate
observed when reticulocyte Iysates are incubated with high concentrations of
cAMP correlated with dephosphorylation of eEF-2. 70 The dephosphorylated
form of eEF-2 was found to support poly(U)-directed polyphenylalanine syn-
thesis in a reconstituted elongation system when combined with eEF-1 , whereas
phosphorylated eEF-2 was ineffective. 64 •73 Phosphorylated eEF-2 is reported
to bind to 80S ribosomes but to be unable to catalyze translocation of peptidyl
tRNA from the A site to the P site for ribosomes carrying poly(U) and
phenylalanyl tRNA. 74
130 Nutrition and Gene Expression
only one nucleotide, 98 potentially exists in this region. The sequence from
-189 to -182 is 5'-TGACGTGA-3 and conforms to the CRE-consensus. 99
The human GRP78 gene has a similar sequence from -153 to -147. These
sequences correspond to protected domains and are footprinted on the non-
coding strand. 100 With development of promoter deletion mutants, it should
be possible to identify sequences that mediate regulation of GRP78 gene
expression by cAMP and phorbol ester. The second factor favoring GRP78
induction in stressed GH 3 preparations was the preferential translation of
GRP78 mRNA. This mRNA associated almost exclusively with polysomes,
whereas other mRNAs were subject to initiation block. 97 This efficient ini-
tiation of GRP78 may be explained by the recent observation 101 that the 5'
leader of GRP78 mRNA, unlike that of most cellular mRNAs, can confer
initiation by an internal ribosome binding mechanism.
On the basis of these and other findings, alterations in translational activity
following Ca 2 + deprivation or exposure to a reducing environment appeared
to emanate from perturbation of ER function. As noted earlier, the ER is
thought to function prominently in maintenance of Ca2 + homeostasis and
early protein processing reactions requiring an oxidizing environment. Agents
that deplete sequestered Ca2 + stores or that generate a hypoxic environment
acutely suppress translation at initiation. On chronic exposure to either class
of agent, transcriptionally dependent translational accommodation is seen to
develop that appears to depend on the rapid induction of GRP78/BiP, an ER
resident protein proposed to serve in protein translocation and folding. Major
open questions regarding GRP78 include: ( 1) How does the ER signal the
nucleus to transcribe new mRNA for the protein? (2) How does GRP78
promote the recovery of amino acid incorporation in Ca 2 + depleted cells?
Does GRP78 coordinate the relative rates of translational initiation and protein
processing? Evidence that both sequestered Ca 2 + and adequate oxidizing
eqivalents are needed to sustain early protein processing events is summarized
in the following section.
lesser extent other proteins, by HepG2 cells. 103 Following ionophore treatment
secretory glycoproteins with a high mannose configuration accumulated in
vesicles with the density of the rough ER. It was unclear from these studies,
however, that Ca2 + mobilization mediated the observed secretory arrest and
that resumption of normal glycoprotein processing and export was possible
upon removal of the stress.
HepG2 human hepatoma cells were employed as a model system to further
investigate relationships between early protein processing and Ca 2 + storage
by the ER. 104 Three lines of direct evidence were provided that Ca2 + per se
is required for processing and secretion. First, secretion of a 1 -antitrypsin and
to a lesser degree other newly synthesized polypeptides was reduced following
treatments in EGTA-containing medium. Second, inhibition of glycoprotein
processing by Cbz-Gly-Phe-NH 2 was found to depend on Ca2 +. Inhibitions
of a 1-antitrypsin processing and of complement factor 3 (C3) secretion by
dipeptide were largest in Ca2 + -depleted medium and were either markedly
reduced or prevented at elevated extracellular Ca2 +. Third, it was demon-
strated that arrest of a 1 -antitrypsin processing and export in the presence of
ionomycin was fully reversed by Ca2 + following extraction of the drug with
BSA. In addition to establishing a Ca 2 + involvement in the mechanism of
ionophore inhibition, accomplishment of the reversal illustrated (1) that io-
nomycin-treated preparations retained the normal precursor form of a 1-anti-
trypsin rather than a malprocessed intermediate destined for degradation, and
(2) that both vesicular processing and protein transport were not permanently
disrupted by ionophore treatment. In contrast to agents depleting Ca 2 + stores,
exposure to DTT reduced albumin export while affecting a 1-antitrypsin export
minimally. Although the inhibition of albumin export was not accompanied
by significant intracellular accumulation of native polypeptide, considerable
amounts of several cell-associated, immunoreactive polypeptides of differing
molecular mass were invariably seen to develop in DTT-treated cells. These
peptides appeared to be aggregative/degradative forms of albumin collectively
more than doubling cell associated retention of the protein.
Ca2 + has been proposed to be required for the transport of secretory
proteins from ER to Golgi. 105 That albumin secretion is affected minimally
by ionomycin and not affected by EGTA argues against a prominent role for
Ca 2 + in transport of all proteins from the ER. Alternatively, Ca 2 + stored in
the ER may be required for accurate folding which, in turn, is necessary for
transport of secretory glycoproteins to other vesicular compartments. This
mechanism assumes more stringent folding requirements for transport of gly-
coproteins, such as a 1-antitrypsin and C3, than for nonglycosylated secretory
species such as albumin. An additional possibility consistent with available
data is that depletion of sequestered Ca2 + specifically restricts the enzymatic
trimming of high mannose forms of glycoproteins, resulting in the accumu-
lation of species not recognized by transport systems. By contrast, these data
Brostrom and Brostrom 135
VI. SUMMARY
Inhibition of mRNA translation consequent to interruption of ER protein
processing appears to be of general significance in that suppression of amino
acid incorporation into total cellular proteins of HepG2 cells accompanied
inhibitions of protein processing by agents depleting sequestered Ca2 + stores
or by DTT. 104 Tunicamycin, which interferes with transfer of the core oli-
gosaccharide structure to newly synthesized polypeptides and retards secretion
of the nonglycosylated species, was found to inhibit translational initiation
in GH 3 cells. 95 It is most appealing, therefore, to propose that the ER effec-
tively coordinates rates of mRNA translation with protein processing. While
translation obviously provides the substrates for the processing apparatus of
the ER and Golgi, there is no evidence of any other influence of translation
on processing. 107 In contrast it is quite clear that some inhibitors of protein
processing interdict translational initiation, such that the synthesis of almost
all proteins, including those not subject to processing, is curtailed. It will be
of interest to ascertain whether cells expressing translational tolerance sub-
sequent to induction of GRP78 are capable of efficient protein processing
when challenged by stresses that perturb ER function. Other questions of
interest include: Where is the Ca2 + requirement in glycoprotein processing
exerted? Does sequestered Ca 2 + regulate the rate of glycoprotein processing?
REFERENCES
I. Brostrom, C. 0. and Brostrom, M. A., Calcium dependent regulation of protein
synthesis in intact mammalian cells, Annu. Rev. Physiol., 52, 577, 1990.
2. Lee, C. and Chen, L. B., Dynamic behavior of endoplasmic reticulum in living cells,
Cell, 54, 37, 1988.
3. Green, M. and Mazzarella, R. A., Biosynthesis and sorting of proteins of the endo-
plasmic reticulum, in Protein Transfer and Organelle Biogenesis, Das, R. C. and Robbins,
P. W., Eds., Academic Press, Orlando, 1988, 243.
4. Koch, G. L. E., The endoplasmic reticulum and calcium storage, Bioessays, 12, 527,
1990.
5. Carafoli, E., Intracellular calcium homeostasis, Annu. Rev. Biochem., 56, 395, 1987.
6. Meldolsi, J. and Pozzan, T., Pathways of Ca2 + entry at the plasma membrane: voltage-,
receptor-, and second messenger-operated channels, Exp. Cell Res., 171, 271, 1987.
136 Nutrition and Gene Expression
7. Berridge, M. J., Inositol phosphates and cell signalling, Nature (London), 341, 197,
1989.
8. Tsien, R. Y., Calcium channels, stores, and oscillations, Annu. Rev. Cell Bioi., 6, 715,
1990.
9. Walz, B. and Baumann, B., Calcium-sequestering cell organelles: in situ localization,
morphological and functional characterization, Prog. H istochem. Cytochem. , 20, I , 1989.
10. Meldolesi, J., Madeddu, L., and Pozzan, T., Intracellular Ca 2 + storage organelles in
non-muscle cells: heterogeneity and functional assignment, Biochim. Biophys. Acta, I 055,
130, 1990.
II. Rossier, M. F. and Putney, J. W., Jr., The identity of the calcium-storing, inositol
1,4,5-trisphosphate-sensitive organelle in non-muscle cells: calciosome, endoplasmic re-
ticulum ... or both?, Trends Neurosci., 14, 310, 1991.
12. Berridge, M. J., Cytoplasmic calcium oscillations: a two pool model, Cell Calcium,
12, 63, 1991.
13. Volpe, P., Krause, K.-H., Hashimoto, S., Zorzato, F., Pozzan, T., Meldolesi, J.,
and Lew, D. P., "Calciosome," a cytoplasmic organelle: The inositol I ,4,5-trisphos-
phate-sensitive Ca'+ store of nonmuscle cells?, Proc. Nat!. Acad. Sci. U.S.A., 85, 1091,
1989.
14. Baumann, 0., Walz, B., Somlyo, A. V., and Somlyo, A. P., Electron probe microan-
alysis of calcium release and magnesium uptake by endoplasmic reticulum in bee pho-
toreceptors, Proc. Natl. Acad. Sci. U.S.A., 88, 741, 1991.
15. Bian, J., Ghosh, T. K., Wang, J.-C., and Gill, D. L., Identification of intracellular
calcium pools. Selective modification by thapsigargin, J. Bioi. Chern., 266, 8801, 1991.
16. Satoh, T., Ross, C. A., Villa, A., Supattapone, S., Pozzan, T., Snyder, S. H., and
Meldolesi, J., The inositol! ,4,5-trisphosphate receptor in cerebellar purkinje cells: quan-
titative immunogold labeling reveals concentration in an ER subcompartment, J. Cell
Bioi., Ill, 615, 1990.
17. Wileman, T., Kane, L. P., Carson, G. R., and Terhorst, C., Depletion of cellular
calcium accelerates protein degradation in the endoplasmic reticulum, J. Bioi. Chern.,
266, 4500, 1991.
18. Pelham, H. R. B., Control of protein exit from the endoplasmic reticulum, Annu. Rev.
Cell Bioi., 5, I, 1989.
19. Milner, R. E., Baksh, S., Shemanko, C., Carpenter, M. R., Smillie, L., Vance,
J. E., Opas, M., and Michalak, M., Calreticulin, and not calsequestrin, is the major
calcium binding protein of smooth muscle sarcoplasmic reticulum and liver endoplasmic
reticulum, J. Bioi. Chern., 26, 7155, 1991.
20. Sonenberg, N., Cap-binding proteins of eukaryotic messenger RNA: functions in initi-
ation and control of translation, Prog. Nucleic Acid Res., 35, 173, 1988.
21. Hershey, J. W. B., Protein phosphorylation controls translation rates. J. Bioi. Chern.,
264, 20823, 1989.
22. Hershey, J. W. B., Translational control in mammalian cells, Annu. Rev. Biochem., 60,
717, 1991.
23. Albert, P. R. and Tashjian, A. H., Jr., Relationship of thyrotropin-releasing hormone-
induced spike and plateau phases in cytosolic free Ca 2 + concentrations to hormone se-
cretion, J. Bioi. Chern., 259, 15350, 1984.
24. Brostrom, C. 0., Bocckino, S. B., Brostrom, M.A., and Galuska, E. M., Regulation
of protein synthesis in isolated hepatocytes by calcium-mobilizing hormones, Mol. Phar-
macal., 29, 104, 1986.
25. Brostrom, C. 0., Bocckino, S. B., and Brostrom, M. A., Identification of a Ca2 +
requirement for protein synthesis in eukaryotic cells, J. Bioi. Chern., 258, 14390, 1983.
26. Brostrom, M. A., Brostrom, C. 0., Bocckino, S. B., and Green, S. S., Ca2 + and
hormones interact synergistically to stimulate rapidly both prolactin production and overall
protein synthesis in pituitary tumor cells, J. Cell. Physio/., 121, 291, 1984.
Brostrom and Brostrom 137
27. Wolfe, S. E., Brostrom, C. 0., and Brostrom, M. A., Mechanisms of action of
inhibitors of prolactin secretion in GH 3 pituitary cells. I. Ca 2 +-dependent inhibition of
amino acid incorporation, Mol. Pharmacal., 29, 411, 1986.
28. Wolfe, S. E. and Brostrom, M. A., Mechanisms of action of inhibitors of prolactin
secretion in GH 3 pituitary cells. II. Blockade of voltage dependent Ca 2 + channels, Mol.
Pharmacal., 29, 420, 1986.
29. Brostrom, M.A., Chin, K.-V., Cade, C., Gmitter, D., and Brostrom, C. 0., Stim-
ulation of protein synthesis in pituitary cells by phorbol esters and cyclic AMP, J. Bioi.
Chern., 262, 16515, 1987.
30. Chin, K.-V., Cade, C., Brostrom, M. A., Galuska, E. M., and Brostrom, M. A.,
Calcium-dependent regulation of protein synthesis at translational initiation in eukaryotic
cells, J. Bioi. Chern., 262, 16509, 1987.
31. Abdel-Latif, A. A., Calcium-mobilizing receptors, polyphosphoinositides, and the gen-
eration of second messengers, Pharmac. Rev., 38, 227, 1986.
32. Chin, K.-V., Cade, C., Brostrom, M.A., and Brostrom, C. 0., Regulation of protein
synthesis in intact rat liver by calcium mobilizing agents, Int. J. Biochem., 20, 1313,
1988.
33. Gmitter-Yellen, D., Brostrom, C. 0., Kuznetsov, G., and Brostrom, M. A., un-
published data, 1991.
34. Brostrom, C. 0., Chin, K.-V., Wong, W. L., Cade, C., and Brostrom, M. A.,
Inhibition of translational initiation in eukaryotic cells by calcium ionophore, J. Bioi.
Chern., 264, 1644, 1989.
35. Wolf, B. A., Turk, J., Sherman, W. R., and McDaniel, M. L., Intracellular Ca 2 +
mobilization by arachidonic acid, J. Bioi. Chern., 261, 3501, 1986.
36. Yasuda, H., Kishiro, K., Izumi, N., and Nakanishi, M., Biphasic liberation of ar-
achidonic and stearic acids during cerebral ischemia, J. Neurochem., 45, 168, 1985.
37. Kolesnick, R.N., Musacchio, I., Thaw, C., and Gershengorn, M. C., Arachidonic
acid mobilizes calcium and stimulates prolactin secretion from GH 3 cells, Am. J. Physiol.,
246, E458, 1984.
38. Chan, K.-M. and Turk, J., Mechanism of arachidonic acid-induced Ca 2 + mobilization
from rat liver microsomes, Biochim. Biophys. Acta, 928, 186, 1987.
39. Rotman, E. I., Brostrom, M.A., and Brostrom, C. 0., Inhibition of protein synthesis
in intact mammalian cells by arachidonic acid, Biochem. J., 282, 487, 1992.
40. Strittmatter, W. J., Couch, C. B., and Mundy, D. I., Role of metalloendoprotease
in the fusion of biological membranes, in Cell Fusion, Sowers, A. E., Ed., Plenum
Press, New York, 1987, 99.
41. Strous, G. J., van Kerkhof, P., Dekker, J., and Schwartz, A. L., Metalloendoprotease
inhibitors block protein synthesis, intracellular transport, and endocytosis in hepatoma
cells, J. Bioi. Chern., 263, 18197, 1988.
42. Hammerschlag, R., Bolen, F. A., and Stone, G. C., Metalloendoprotease inhibitors
block fast axonal transport, J. Neurochem. 52, 268, 1989.
43. Lelkes, P. I. and Pollard, H. B., Oligopeptide inhibitors of metalloendoprotease inhibit
catecholamine secretion from bovine adrenal chromaffin cells by modulating intracellular
calcium homeostasis, J. Bioi. Chern., 262, 15496, 1987.
44. Brostrom, M.A., Prostko, C. R., Gmitter-Yellen, D., Grandison, L. J., Kuznetsov,
G., Wong, W. L., and Brostrom, C. 0., Inhibition of translational initiation by me-
talloendoprotease antagonists, J. Bioi. Chern., 266, 7037, 1991.
45. Kelly, R. B., Tracking an elusive receptor, Nature (London), 345, 480, 1990.
46. Wong, W. L., Brostrom, M.A., and Brostrom, C. 0., unpublished data, 1991.
47. Kumar, R. V., Wolfman, A., Panniers, R., and Henshaw, E. C., Mechanism of
inhibition of polypeptide chain initiation in calcium-depleted Erhlich ascites tumor cells,
J. Cell Bioi., 108, 2107, 1989.
138 Nutrition and Gene Expression
48. Wong, W. L., Brostrom, M.A., and Brostrom, C. 0., Effects of Ca'* and ionophore
A23187 on protein synthesis in intact rabbit reticulocytes, Int. J. Biochem., 23, 605,
1991.
49. Kumar, R. V., Panniers, R., Wolfman, A., and Henshaw, E. C., Inhibition of protein
synthesis by antagonists of calmodulin in Ehrlich ascites tumor cells, Eur. J. Biochem.,
195, 313, 1991.
50. Asano, M. and Hidaka, H., Biopharmacological properties of naphthalenesul fonamides
as potent calmodulin antagonists, in Calcium and Cell Function, Vol. 5, Cheung, W. Y.,
Ed., Academic Press, Orlando, 1984, 123.
51. West, J. B., Physiological Basis of Medical Practice, lith ed., Williams and Wilkins,
Baltimore, 1985, 346.
52. Kimball, S. R. and Jefferson, L. S., Mechanism of inhibition of protein synthesis by
vasopressin in rat liver, J. Bioi. Chern., 265, 16794, 1990.
53. Fawell, E. H., Boyer, I. J., Brostrom, M.A., and Brostrom, C. 0., A novel calcium-
dependent phosphorylation of a ribosome-associated protein, J. Bioi. Chern., 264, 1650,
1989.
54. Traugh, J. A. and Pendergast, A. M., Regulation of protein synthesis by phosphor-
ylation of ribosomal protein S6 and aminoacyl tRNA synthetases, Pro I?,. Nuclei Acid Res.
Mol. Bioi., 33, 195, 1986.
55. Brostrom, M. A., Lin, X., Cade, C., Gmitter, D., and Brostrom, C. 0., Loss of a
calcium requirement for protein synthesis in pituitary cells following thermal or chemical
stress, J. Bioi. Chern., 264, 1638, 1989.
56. Pancake, B., Prostko, C. R., Brostrom, M. A., and Brostrom, C. 0., unpublished
data, 1989.
57. Ryazanov, A. G. and Spirin, A. S., Phosphorylation of elongation factor 2: a key
mechanism in regulating gene expression in vertebrates, The New Biologist, 2, 843, 1990.
58. Palfrey, H. C., Presence in many mammalian tissues of an identical major substrate (M,
100,000) for calmodulin-dependent protein kinase. FEBS Lett.. 157, 183. 1983.
59. Nairn, A. C., Bhagat, B., and Palfrey, H. C., Identification of calmodulin-dependent
protein kinase III and its major M, 100.000 substrate in mammalian tissues, Proc. Nat/.
Acad. Sci. U.S.A .. 82, 7939, 1985.
60. End, D., Tolson, N., Hashimoto, S., and Guroff, G., Nerve growth factor-induced
decrease in the cell free phosphorylation of a soluble protein in PC12 cells, J. Bioi.
Chern .. 258, 6549, 1983.
61. Hama, T. and Guroff, G., Distribution of NsplOO and NsplOO kinase, a nerve growth
factor-sensitive phosphorylation system, in rat tissues, J. Neurochem .. 45. 1279. 1985.
62. Togari, A. and Guroff, G., Partial purification and characterization of a nerve growth
factor-sensitive kinase and its substrate from PCI2 cells. J. Bioi. Chern., 260. 3804,
1985.
63. Ryazanov, A. G., CaZ+ /calmodulin-dependent phosphorylation of elongation factor 2,
FEBS Lett., 214.331, 1987.
64. Nairn, A. C. and Palfrey, H. C., Identification of the major M, 100.000 substrate for
calmodulin-dependent protein kinase III in mammalian cells as elongation factor 2, J.
Bioi. Chem., 262, 17299. 1987.
65. Ryazanov, A. G., Natapov, P. G., Shestakova, E. A., Severin, F. F., and Spirin,
A. S., Phosphorylation of the elongation factor 2: the fifth Ca'+ /calmodulin-dependent
system of protein phosphorylation, Biochimie, 70, 619. 1988.
66. Ovchinnikov, L. P., Motuz, L. P., Natapov, P. G., Averbuch, L. J., Wettenhall,
R. E. H., Szyzka, R., Kramer, G., and Hardesty, B., Three phosphorylation sites in
elongation factor 2. FEBS Lett., 275. 209, 1990.
67. Price, N. T., Redpath, N. T., Severinov, K. V., Campbell, D.G., Russell, J. M.,
and Proud, C. G., Identification of the phosphorylation sites in elongation factor-2 from
rabbit reticulocytes, FEBS Lett.. 282. 253, 1991.
Brostrom and Brostrom 139
68. Kim, Y. W., Kim, C. W., Kang, K. R., Byun, S.M., and Kang, Y.-S., Elongation
factor-2 in chick embryo is phosphorylated on tyrosine residues as well as serine and
threonine, Biochern. Biophys. Res. Cornrnun., 175,400, 1991.
69. Nilsson, L. and Nygard, 0., Altered sensitivity of eukaryotic elongation factor 2 for
trypsin after phosphorylation and ribosomal binding, J. Bioi. Chern., 266, 10578, 1991.
70. Sitikov, A. S., Simonenko, P. N., Shestakova, E. A., Ryazanov, A. G., and
Ovchinnikov, L. P., cAMP-dependent activation of protein synthesis correlates with
dephosphorylation of elongation factor 2, FEES Lett., 228, 327, 1988.
71. Redpath, N. T. and Proud, C. G., The tumor promoter okadaic acid inhibits reticulocyte-
lysate protein synthesis by increasing the net phosphorylation of elongation factor 2,
Biochern. J., 262, 69, 1989.
72. Gschwendt, M., Kittstein, W., Mieskes, G., and Marks, F., A type 2A protein
phosphatase dephosphorylates the elongation factor 2 and is stimulated by the phorbol
ester TPA in mouse epidermis in vivo, FEES Lett., 257, 357, 1989.
73. Ryazanov, A. G., Shestakova, E. A., and Natapov, P. G., Phosphorylation of elon-
gation factor 2 by EF-2 kinase affects rate of translation, Nature (London), 334, 170,
1988.
74. Ryazanov, A. G. and Davydova, E. K., Mechanism of elongation factor 2 (EF-2)
inactivation upon phosphorylation, FEES Lett., 251, 187, 1989.
75. Palfrey, H. C., Nairn, A. C., Muldoon, L. L., and Villereal, M. L., Rapid activation
of calmodulin-dependent protein kinase III in mitogen-stimulated human fibroblasts, J.
Bioi. Chern., 262, 9875, 1987.
76. Mackie, K. P., Nairn, A. C., Hampel, G., Lam, G., and Jaffe, E. A., Thrombin
and histamine stimulate the phosphorylation of elongation factor 2 in human umbilical
vein endothelial cells, J. Bioi. Chern., 264, 1748, 1989.
77. Nairn, A. C., Nichols, R. A., Brady, M. J., and Palfrey, H. C., Nerve growth factor
treatment or cAMP elevation reduces Ca2 + /calmodulin-dependent protein kinase III ac-
tivity in PC12 cells, J. Bioi. Chern., 262, 14265, 1987.
78. Brady, M. J., Nairn, A. C., Wagner, J. A., and Palfrey, H. C., Nerve growth factor-
induced down-regulation of calmodulin-dependent protein kinase-III in PC-12 cells in-
volves cyclic AMP-dependent protein kinase, J. Neurochern., 54, 1034, 1990.
79. Nygard, 0., Nilsson, A., Carlberg, U., Nilsson, L., and Amons, R., Phosphorylation
regulates the activity of the eEF-2-specific Ca2 +- and calmodulin-specific protein kinase
III, J. Bioi. Chern., 266. 16425, 1991.
80. Celis, J. E., Madsen, P., and Ryazanov, A. G., Increased phosphorylation of elongation
factor 2 during mitosis in transformed human amnion cells correlates with a decreased
rate of protein synthesis, Proc. Nat/. Acad. Sci. U.S.A., 87, 4231, 1990.
81. Severinov, K. V., Melnikova, E. G., and Ryazanov, A. G., Downregulation of the
translation elongation factor 2 kinase in Xenopus laevis occytes at the final stages of
oogenesis, The New Biologist, 2, 887, 1990.
82. Reed, J. C., Alpers, J. D., Nowell, P. C., and Hoover, R. G., Sequential expression
of protooncogenes during lectin-stimulated mitogenesis of normal human lymphocytes,
Proc. Nat/. Acad. Sci. U.S.A., 83, 3982, 1986.
83. Greenberg, M. E., Hermanowski, A. L., and Ziff, E. B., Effect of protein synthesis
inhibitors on growth factor activation of c-fos, c-myc, and actin gene transcription, Mol.
Cell. Bioi., 6, 1050, 1986.
84. Lee, A. S., Coordinated regulation of a set of genes by glucose and calcium-ionophore
in mammalian cells, Trends Biochern. Sci., 12, 20, 1987.
85. Kim, Y. K., Kim, K. S., and Lee, A. S., Regulation of the glucose-regulated genes
by [3-mercaptoethanol requires de novo protein synthesis and correlates with inhibition
of protein glycosylation, J. Cell. Physioi., 133, 553, 1987.
86. Watowich, S. S. and Morimoto, R. 1., Complex regulation of heat shock- and glucose-
responsive genes in human cells, Mol. Cell. Bioi., 8, 393, 1988.
140 Nutrition and Gene Expression
105. Balch, W. E., Biochemistry of interorganelle transport, J. Bioi. Chem., 264. 16965,
1989.
106. White, A., Handler, P., Smith, E. L., Hill, R. L., and Lehman, I. R., Principles of
Biochemistry, McGraw-Hill, New York, 1978, 136.
107. Wieland, F. T., Gleason, M. L., Serafini, T. A., and Rothman, J. E., The rate of
bulk flow from the endoplasmic reticulum to the cell surface, Cell, 50, 289, 1987.
Chapter 6
TABLE OF CONTENTS
I. INTRODUCTION
TABLE 1
Structural and Functional Features of Mammalian Hexokinases
Major sites
Hexokinase type Mass (kDa) Km for glucose G-6-P inhibition of expression
Note: The mass, Km for glucose, inhibition by glucose-6-P, and major tissue sites of expression
are shown for hexokinase types I through IV.
~
"';::i
~
"'(3•
"'::s
Magnuson and Jetton 149
~ : - :::J
X
GK.Z9 :::J:: :::J ..r::. ..r::. ~
>UJ (ij > > a. a. Cl Ci) ;:s
(XJ a.. a.. en (XJ (XJ
(lnsulinoma) a.. a.. \l en
2216 bp
FIGURE 2. Comparison of glucokinase cDNAs from rat liver and insulinoma tissues. The structural differences of glucokinase cDNAs isolated from different
tissue sources is illustrated. The GK.Z2 eDNA clone was isolated from a rat liver eDNA library and the GK.Z9 eDNA clone was isolated from a rat insulinoma
eDNA library. Differences in the cDNAs at the 5' end and within the coding sequence of the enzyme are indicated. The differences coincide with the location
of splice junctions indicating that alternate RNA splicing is involved in generating glucokinase isoform diversity.
....
....Ul
152 Nutrition and Gene Expression
MLDDRARMEATKKEKVEQILAEFQLQEEDLKKVMSRMQKEMDRGLRLETH
EEASVKMLPTYVRSTPEGSEVGDFLSLDLGGTNFRVMLVKVGEGEAGQWS
VKTKHQMYSIPEDAMTGTAEMLFDYISECISDLLDKHQMKHKKLPLGFTF
SFPVRHEDLDKGILLNWTKGFKASGAEGNNIVGLLRDAIKRRGDFEMDVV
AMVNDTVATMISCYYEDRQCEVGMIVGTGCNACYMEEMQNVELVEGDEGR
MCVNTEWGAFGDSGELDEFLLEYDRMVDESSANPGQQLYEKIIGGKYMGE
LVRLVLLKLVDENLLFHGEASEQLRTRGAFETRFVSQVESDSGDRKQIHN
ILSTLGLRPSVTDCDIVRRACESVSTRAAHMCSAGLAGVINRMRESRSED
VMRITVGVDGSVYKLHPSFKERFHASVRRLTPNCEITFIESEEGSGRGAA
LVSAVACKKACMLAQ
MAMDTTRCGAOLLTL ...
FIGURE 3. Cell type-specific sequences of the islet and liver isoforrns of glucokinase. The
different amino acids sequences of the islet (top) and hepatic (bottom) glucokinase isoforrns are
shown (underlined).
rat genomic DNA that showed there was only a single gene for glucokinase
in the rat. 35 .4 1 However, surprisingly, when the liver-derived eDNA was used
as a probe in RNA blot experiments of islet RNA, both Magnuson and
Shelton41 and Iynedjian et a!. 35 found that the pancreatic islet form of rat
glucokinase mRNA was longer by 200 to 400 bases than that in liver. More-
over, Iynedjian et a!. 35 found that the effects of a fasting/refeeding regimen
on hepatic and islet glucokinase mRNA and protein levels were also different.
While the amount of hepatic mRNA and immunoreactive protein varied with
fasting and refeeding, islet glucokinase mRNA and immunodetectable protein
did not change under these conditions. 35 To determine the structural basis for
the different sizes of the mRNAs, and to explore the basis for the differential
regulation, cDNAs for islet glucokinase mRNA were cloned by both groups.
Initial studies identifying structural differences in glucokinase isoforms
yielded intriguing results. The glucokinase cDNAs cloned by Magnuson and
Shelton41 were obtained from a rat insulinoma eDNA library, whereas the
cDNAs obtained by Hughes eta!. 42 were cloned from a rat islet eDNA library.
Both groups determined that glucokinase transcripts from islet tissue and
hepatocytes were not equivalent and that this difference was located at the
Magnuson and Jetton 153
5 1 end of the mRNAs. The nature of these differences suggested that additional
mechanisms, such as alternate RNA splicing and/or the utilization of distinct
promoter regions, were involved in producing tissue-specific glucokinase
mRNAs. Indeed, when the insulinoma and hepatic cDNAs for glucokinase
were compared by Magnuson and Shelton, 41 two differences were found that
were due to both the use of alternate promoter regions and to alternate RNA
processing, as shown in Figure 2. The sequences at the 5 1 ends of the cDNAs
were distinct and the site at which this difference occurred coincided with
the splice junction between the first and second exons, thus indicating that
the hepatic and islet glucokinase mRNAs utilized different first exon se-
quences in the liver and islet, respectively. This finding implicated the use
of different promoter regions in the production of glucokinase mRNAs in the
different cell types. Moreover, since the translation initiation codons for liver
and islet glucokinase were located in different first exons, the amino acid
sequence at the amino terminus of the enzyme from liver and pancreatic islets
was affected, as previously shown in Figure 3. In addition to sequence dif-
ferences related to the use of alternate promoter regions, there were also
sequence differences within the reading frame of the enzyme in the GK.Z9
eDNA clone, thus implicating alternate RNA processing in second region.
Together, the differences in the hepatic and islet glucokinase cDNAs indicated
that variant glucokinase isoforms were being produced as a result of alternate
RNA splicing of the glucokinase gene product.
The transcription unit that produces the hepatic glucokinase mRNA was
characterized by Magnuson et al. 43 and found to contain 10 exons that span
about 15 kb. The location of the f3 cell first exon, however, was not present
in the genomic DNA fragments characterized in the cloning of the hepatic
gene despite having 7.5 kb of 5 1 flanking DNA sequence in the lambda phage
clone studied (A.GK5). To determine the location of the genomic DNA se-
quences that gave rise to the 5 1 noncoding sequence in the islet glucokinase
mRNA, another genomic DNA fragment was isolated. 41 This new DNA
fragment (A.GK7) did not overlap with the 5 1 end of A.GK5, the clone con-
taining the liver-specific first exon and promoter region, thus indicating that
the upstream promoter region had to be at least 12 kb upstream from the
downstream promoter region. Although the exact distance separating the dif-
ferent promoter regions has not yet been determined, an upper limit for this
distance has been suggested by the cloning of cosmid DNAs for the gene. 44
Both promoter regions are contained in several cosmid clones that have an
insert size of approximately 40 kb, thus placing an upper limit on the distance.
However, since these clones have not been fully mapped, the actual distance
154 Nutrition and Gene Expression
t 1 kb
~
18
(>12kb)
:I'
•
1L
Ill I
2a 2 3
t
4
II
56
~
I I I.
789 10
• = Liver-specific
FIGURE 4. Glucokinase gene structure. The intronlexon structure of the glucokinase gene is
diagramatically illustrated. The gene has 2 different promoter regions that are separated by at
least 12 kb and which are linked to cell type-specific first exon sequences. The upstream promoter
region is active in the pancreatic [3 cell and pituitary, whereas the downstream promoter region
is active in the liver. In addition, the location of a cassette exon (2A) is shown which is utilized
in the production of about 5% of the glucokinase mRNA present in liver 59 (From Magnuson,
M. A., J. Cell. Biochem., 48, 115, 1992. With permission.)
is not known and may, in fact, be considerably less. A map of the glucokinase
gene, as currently understood, is shown in Figure 4. The gene spans over 27
kb and contains 12 different exons that are used in a tissue-specific manner.
Among the glucokinase isoforrns that are functional, only 10 exons are utilized
with the complete sequence of exons 2 through 10 being necessary for catalytic
activity.
The tissue-specific expression of the upstream and downstream promoter
regions has been tested by RNA-PCR amplification experiments. In these
studies, the glucokinase mRNAs containing exon 1~ sequences have been
detected only in the islet and pituitary, while sequences containing the 1H
sequences have only been identified in the liver. 41 •45
TABLE 2
Differential Regulation of Hepatic and
13 Cell Glucokinase
Stimulatory effect Inhibitory effect
expressed in the pituitary, whereas the insulin gene is not, a different set of
regulatory factors must be utilized to direct this different pattern of tissue
expression. With the exception of the pituitary, where the upstream promoter
region is utilized for mRNA expression, no other tissue has been found to
express this gene.
Fusion gene experiments have revealed that a small fragment of the
upstream promoter region is sufficient for expression in cultured ~ cell lines.
Indeed, a 294-bp fragment of DNA containing this region appears to be all
that is necessary for specific expression in~ cells. 52 Within this DNA fragment
are several novel elements that appear to contribute to transcription in ~ cells. 52
Moreover, in the islets of euglycemic rats, glucokinase appears to be expressed
specifically in the ~ cell as studied by immunohistochemical analysis. 53 In-
terestingly, glucokinase immunoreactivity among different pancreatic ~cells
is not uniform. Some ~ cells display striking amounts of glucokinase im-
munoreactivity while others have much less. 53 Evidence is accumulating in
support of the functional heterogeneity of pancreatic ~ cells although the basis
for this heterogeneity is not known. 54 •55 However, the observation of variable
glucokinase immunoreactivity itself is important since different amounts of
glucokinase activity in different ~ cells may cause the glucose sensitivity of
individual cells to be different.
Hormonal effects on the activity of the upstream glucokinase promoter
region have not been observed; however, this has been more difficult to study
given the lower abundance of islet glucokinase and the general difficulties
encountered in isolating pancreatic islets. However, there is evidence that
exercise training has a pronounced effect on islet glucokinase mRNA levels.
Koranyi et al. 56 determined that rats which had undergone an exercise training
regimen have decreased expression of both proinsulin and glucokinase mRNAs.
Whether the change in the amount of glucokinase mRNA in response to
exercise is due to a change in the rate of gene transcription or due to an
alteration in the stability of the mRNA has not been determined.
Regulation of glucokinase expression in the islet may not necessarily be
occurring at the level of gene transcription. For instance, the different 5'
noncoding sequences in the hepatic and islet glucokinase mRNAs might allow
for regulation at the translational level. The length of the leader sequence is
different in the liver and islet, which might affect the translational efficiencies
of the mRNAs. Moreover, the islet mRNA has two ATGs upstream of the
actual initiator ATG. The upstream ATGs have poor Kozac consensus se-
quences but may act to diminish translational efficiency of the RNA. 41
:
Iso form 17a.a.
Location Km Vmax
~
DNA!h
L2 j.c Liver ND ND
81
82
1------1
100 a.a.
: •
~
Liver-specific
sequences II
Islet-specific
sequences
~c
j.c
o
Islet
Islet
Common
sequences
15.5
ND
1111
Sequences
10.5
ND
deleted
by atternate RNA
splicing
FIGURE 5. Kinetics analysis of four different rat glucokinase isoforms produced by alternate
RNA splicing. The coding nucleotide sequences for each of four different glucokinase isoforms
were cloned into the eukaryotic expression vector pCMV4 and transfected into NIH-3T3 cells.
The Km and Vmu for each isoform was determined measuring glucose phosphorylation at different
glucose concentrations. 45 An endogenous low Km hexokinase activity was present in the NIH-
3T3 cells but did not affect the activity determinations for glucokinase. ND = not detected.
were then determined. 45 The results obtained (shown with the corresponding
isoform in Figure 5) indicated that both the L1 and B 1 isoforms were en-
zymatically active while the L2 and B2 isoforms were inactive. The Kms of
the enzymatically active L1 and B 1 isoforms were comparable, but a more
substantial difference was observed in the vmaxs of the enzyme expressed in
this manner. However, the differences in the Vmax of the L1 and B 1 isoforms
could be due to the expression of different amounts of protein since the
mRNAs encoding these isoforms have different sequences surrounding their
translation initiation codons. The differences in the Vmax and Km of the L1
and B 1 isoforms were small compared to the effect of the alternate splice
acceptor site in the 4th exon of the gene. Selection of this splice site resulted
in a glucokinase isoform without apparent catalytic activity. This was an
important findng since the generation of aberrant glucokinase isoforms that
were not functional could diminish glucose phosphorylation and glycolysis
in the 13 cell and liver, thereby affecting glucose sensing and glucose usage,
respectively.
TABLE 3
Alternate RNA Splicing Events Involved in the Production
of Glucokinase mRNA that Have a Deleterious
Effect on Enzyme Activity
1
or 2
indicates that these two alternative splice sites were utilized in a single mRNA.
From Magnuson, M.A., J. Cell. Biochem., 48, 115, 1992. With permission.
2/3 3/4
llllnnHnmiiHNHIIINIIIIIHIIIIIIIIIIJII••·
~ Putative ATP-binding domain
FIGURE 6. Pituitary glucokinase eDNA: alternate RNA splicing disrupts the reading frame
for glucokinase. Glucokinase cDNAs from rat pituitary tissue were obtained by RNA-PCR as
described previously. 45 Two different alternate splicing events were identified as illustrated.
Either alternate splicing event, by itself, would be sufficient to prevent the production of an
active glucokinase isoform.
However, the situation with the radiation-induced AtT-20 cells is more con-
fusing but may simply reflect the phenotypic divergence of this cell line into
different sublines.
VII. CONCLUSIONS
Biochemical studies over at least the last 25 years have established the
importance of glucokinase in the metabolism of glucose by the liver and 13
cell. Studies within the last 5 years that have examined the molecular control
points of this system have shown that glucokinase gene expression is under
multiple levels of regulation, as is shown diagramatically in Figure 7. Most
striking in this regard is the finding of alternate promoter regions in a single
glucokinase gene that impart differential cell type-specific regulation to the
enzyme. An upstream promoter region in the gene drives expression of glu-
cokinase mRNA in both the pancreatic 13 cell and the pituitary while the
downstream promoter region in the gene controls expression of glucokinase
mRNA in the liver. This gene organization allows a different set of regulatory
factors to be involved in both tissues, presumably allowing the tissue-specific
control of glucokinase gene expression.
In addition to the use of alternate promoter regions in the glucokinase
gene, alternate RNA processing contributes in an important way to the reg-
ulation of glucokinase activity. While glucokinase mRNAs are produced in
Magnuson and Jetton 163
GLUCOKINASE GENE
~
Beta cell PROMOTER SELECTION Hepatocyte
and pituitary
Insulin
~
Exercise? TRANSCRIPTION RATE Biotin
T3
cAMP
~
Glucose? TRANSLATION RATE
Exercise?
GLUCOKINASE
FIGURE 7. Multiple control points affecting glucokinase gene expression. A diagram showing
the flow of information from the glucokinase gene to protein. The sites at which regulation of
glucokinase gene expression has been identified are illustrated.
ACKNOWLEDGMENTS
These studies were supported by grants from the National Institutes of
Health and the Juvenile Diabetes Foundation. We thank our colleagues and
collaborators for their contributions to the studies which are summarized here.
REFERENCES
I. Granner, D. and Pilkis, S., The genes of hepatic glucose metabolism, J. Bioi. Chern.,
265, 10173, 1990.
2. Iynedjian, P. B. and Girard, J., Nomenclature for mammalian glucokinase, Biochem.
J., 275, 821, 1990.
3. Ureta, T., The comparative isozymology of vertebrate hexokinases, Camp. Bichem.
Physiol., 718, 549, 1982.
4. Cornish-Bowden, A. and Cardenas, M. L., Hexokinase and glucokinase in liver me-
tabolism, Trends Biochem. Sci., 16, 281, 1991.
5. Weinhouse, S., Regulation of glucokinase in liver, in Current Topics in Cellular Reg-
ulation, Vol. II, Horecker, B. L. and Stadtman, E. R., Eds., Academic Press, New
York, 1976, I.
6. Lawrence, G. M. and Trayer, I. P., Hexokinase isoenzymes: Antigenic cross-reactivities
and amino acid copositional relatedness, Camp. Biochem. Physiol., 798, 233, 1984.
7. Andreone, T. L., Printz, R. L., Pilkis, S. J,, Magnuson, M.A., and Granner, D. K.,
The amino acid sequence of rat liver glucokinase deduced from cloned eDNA, J. Bioi.
Chem., 264,363,1989.
8. Kopetzki, E., Entian, K.-D., and Mecke, D., Complete nucleotide sequence of the
hexokinase PI gene (HXKl) of Saccharomyces cervisiae, Gene, 39, 95, 1985.
9. Stachelek, C., Stachelek, J,, Swan, J,, Botstein, D., and Konigsberg, W., Identifi-
cation. cloning, and sequence determination of the genes specifying hexokinase A and
B from yeast, Nucleic Acids Res., 14, 1986.
10. Nishi, S., Seino, S., and Bell, G. I., Human hexokinase: sequences of amino- and
carboxyl-terminal halves are homologous, Biochem. Biophys. Res. Commun., 157, 937,
1988.
11. Schwab, D. A. and Wilson, J. E., Complete amino acid sequence of rat brain hexokinase,
deduced from the cloned eDNA, and proposed structure of a mammalian hexokinase,
Proc. Nat/. Acad. Sci. U.S.A., 86, 2563, 1989.
12. Arora, K. K., Fanciulli, M., and Pedersen, P. L., Glucose phosphorylation in tumor
cells, J. Bioi. Chem., 265, 6481, 1990.
13. Bennett, W. S. and Steitz, T. A., Structure of a complex between yeast hexokinase A
and glucose, J. Mol. Bioi., 140,211, 1980.
14. Lange, A. L., Xu, L. Z., Poelwijk, F. V., Lin, K., Granner, D. K., and Pilkis,
S. J,, Expression and site-directed mutagenesis of hepatic glucokinase, Biochem. J.,
277, 159, 1991.
15. Iynedjian, P. B., Mobius, G., Seitz, H. J,, Wollheim, C. B., and Renold, A. E.,
Tissue-specific expression of glucokinase: Identification of the gene product in liver and
pancreatic islets, Proc. Nat!. Acad. Sci. U.S.A., 83, 1998, 1986.
16. Thorens, B., Sarkar, H. K., Kaback, H. R., and Lodish, H. F., Cloning and functional
expression in bacteria of a novel glucose transporter present in liver, intestine, kidney,
and beta-pancreatic islet cells, Cell, 55, 281, 1988.
Magnuson and Jetton 165
17, Mueckler, M., Family of glucose-transporter genes. Implications for glucose homeostasis
and diabetes, Diabetes, 39, 6, 1990.
18. James, D. E., Brown, B., Navarro, J., and Pilch, P. F., Insulin-regulatable tissues
express a unique insulin-sensitive glucose transport protein, Nature (London), 330, 183,
1988.
19. Vilaro, S., Palacio, M., Pilch, P. F., Testar, X., and Zorzano, A., Expression of an
insulin regulatable glucose carrier in muscle and fat endothelial cells, Nature (London),
342, 792, 1989.
20. Matschinsky, F. M. and Ellerman, J. E., Metabolism of glucose in the islets of
Langerhans, J. Bioi. Chern., 243, 2730, 1968.
2L Malaisse, W. J., Sener, A., Herchuelz, A., and Hutton, J. C., Insulin release: the
fuel hypothesis, Metabolism, 28, 373, 1979.
22. Malaisse, W. J., Insulin release: the fuel concept, Diabete Metab., 9, 313, 1983.
23. Orci, L., Thorens, B., Ravazzola, M., and Lodish, H. F., Localization of the pancreatic
beta cell glucose transporter to specific plasma membrane domains, Science, 245, 295,
1989.
24. Chen, L., Alam, T., Johnson, J. H., Hughes, S., Newgard, C. B., and Unger, R. H.,
Regulation of 13-cell glucose transporter gene expression, Proc. Nat!. Acad. Sci. U.S.A.,
87, 4088, 1990.
25. Giroix, M.-H., Sener, A., Pipeleers, D. G., and Malaisse, W. J., Hexose metabolism
in pancreatic islets, Biochem. J., 223, 447, 1984.
26. Sener, A., Malaisse-Lagae, F., Giroix, M.-H., and Malaisse, W. J., Hexose metab-
olism in pancreatic islets: Compartmentation of hexokinase in islet cells, Arch. Biochem.
Biophys., 265,61,1986.
27. Shimizu, T., Knowles, B. B., and Matschinsky, F. M., Control of glucose phosphor-
ylation and glucose usage in clonal insulinoma cells, Diabetes, 37, 563, 1988.
28. Shimizu, T., Parker, J. C., Najafi, H., and Matschinsky, F. M., Control of glucose
metabolism in pancreatic 13-cells by glucokinase, hexokinase, and phosphofructokinase,
Diabetes, 37, 1524, 1988.
29. Liang, Y., Najafi, H., and Matschinsky, F. M., Glucose regulates glucokinase activity
in cultured islets from rat pancreas,], Bioi. Chern., 265, 16863, 1990.
30. Meglasson, M. D. and Matschinsky, F. M., New perspectives on pancreatic islet
glucokinase, Am. I Physiol., 13, E1, 1984.
3L Unger, R. H., Diabetic hyperglycemia: link to impaired glucose transport in pancreatic
13 cells, Science, 251, 1200, 199L
32. Ashcroft, F. M., Harrison, D. E., and Ashcroft, S. J. H., Glucose induces closure
of single potassium channels in isolated rat pancreatic 13-cells, Nature (London), 312,
446, 1984.
33. Cook, D. L., Satin, L. S., Ashford, M. L. J., and Hales, C. N., ATP-sensitive K+
channels in pancreatic 13-cells, Diabetes, 37, 495, 1988.
34. Boyd, A. E., Sulfonylurea receptors, ion channels, and fruit flies, Diabetes, 37, 847,
1988.
35. Iynedjian, P. B., Pilot, P.R., Nouspikel, T., Milburn, J. L., Quaade, C., Hughes,
S., Ucla, C., and Newgard, C. B., Differential expression and regulation of the glu-
cokinase gene in liver and islets ofLangerhans, Proc. Nat( Acad. Sci. U.S.A., 86, 7838,
1989.
36. Bedoya, F. J., Matschinsky, F. M., Shimizu, T., ONeil, J. J., and Appel, M. C.,
Differential regulation of glucokinase activity in pancreatic islets and liver of the rat, J.
Bioi. Chern., 261, 10760, 1986.
37, Magnuson, M. A., Glucokinase gene structure: functional implications of molecular
genetic studies, Diabetes, 39, 523, 1990.
38. Matschinsky, F. M., Glucokinase as glucose sensor and metabolic signal generator in
pancreatic beta-cells and hepatocytes, Diabetes, 39, 647, 1990.
166 Nutrition and Gene Expression
39. lynedjian, P. B., Ucla, C., and Mach, B., Molecular cloning of glucokinase eDNA,
J. Bioi. Chern., 262, 6032, 1987.
40. Chien, C.-T., Tauter, A., Lange, A. J., Chan, K., Printz, R. L., El-Maghrabi,
M. R., Granner, D. K., and Pilkis, S. J., Expression of rat hepatic glucokinase in
Escherichia coli, Biochem. Biophys. Res. Commun., 165, 817, 1989.
41. Magnuson, M. A. and Shelton, K. D., An alternate promoter in the glucokinase gene
is active in the pancreatic beta cell, J. Bioi. Chern., 264, 15936, 1989.
42. Hughes, S. D., Quaade, C., Milburn, J. L., Cassidy, L., and Newgard, C. B.,
Expression of normal and novel glucokinase mRNAs in anterior pituitary and islet cells,
J. Bioi. Chern., 266, 4521, 1991.
43. Magnuson, M.A., Andreone, T. L., Printz, R. L., Koch, S., and Granner, D. K.,
Rat glucokinase gene: Structure and regulation by insulin, Proc. Nat!. Acad. Sci. U.S.A.,
86, 4838, 1989.
44. Niswinder, K. and Magnuson, M. A., unpublished data.
45. Liang, Y., Jetton, T. L., Zimmerman, E., Najafi, H., Matschinsky, F. M., and
Magnuson, M. A., Effects of alternate RNA splicing on glucokinase isoform activities
in the pancreatic islet, liver, and pituitary, J. Bioi. Chern., 266, 6999, 1991.
46. Noguchi, T., Takenaka, M., Yamada, K., Matsuda, T., Hashimoto, M., and Tanaka,
T., Characterization of the 5' flanking region of rat glucokinase gene, Biochem. Biophys.
Res. Commun., 164, 1247, 1989.
47. lynedjian, P., Gjinovci, A., and Renold, A. E., Stimulation by insulin of glucokinase
gene transcription in liver of diabetic rats, J. Bioi. Chern., 263, 740, 1988.
48. Hoppner, W. and Seitz, H.-J., Effect of thyroid hormones on glucokinase gene tran-
scription in rat liver, J. Bioi. Chern., 264, 20643, 1989.
49. Narkewicz, M. R., lynedjian, P. B., Ferre, P., and Girard, J., Insulin and tri-
iodothyronine induce glucokinase mRNA in primary culture of neonatal rat hepatocytes,
Biochem. J., 271, 1990.
50. Chauhan, J. and Dakshinamurti, K., Transcriptional regulation of the glucokinase gene
by biotin in starved rats. J. Bioi. Chern., 266, 10035, 1991.
51. lynedjian, P. B., Jotterand, D., Nouspikel, T., Asfari, M., and Pilot, P.-R., Tran-
scriptional induction of glucokinase gene by insulin in cultured liver cells and its repression
by the glucagon-cAMP system, J. Bioi. Chern., 264, 21824, 1989.
52. Shelton, K. D., Franklin, A. J., Khoor, A., Beechem, J., and Magnuson, M. A.,
Multiple elements in the upstream glukokinase promoter contribute to transcription in
insulinoma cells. Mol. Cell. Bioi., in press.
53. Jetton, T. L. and Magnuson, M. A., Heterogeneous expression of glucokinase among
different pancreatic [3-cells, Proc. Nat/. Acad. Sci. U.S.A., 89, 2619, 1992.
54. Schuit, F. C., lntVeld, P. A., and Pipeleers, D. G., Glucose stimulates proinsulin
biosynthesis by a dose-dependent recruitment of pancreatic beta cell, Proc. Nat!. Acad.
Sci. U.S.A., 85, 3865, 1988.
55. Solomon, D. and Meda, P., Heterogeneity and contact-dependent regulation of hormone
secretion by individual beta cells, Exp. Cell Res., 162, 507, 1986.
56. Koranyi, L. I., Bourey, R. E., Slentz, C. A., Holloszy, J, 0., and Permutt, M.A.,
Coordinate reduction of rat pancreatic islet glucokinase and proinsulin mRNA by exercise
training, Diabetes, 40,401, 1991.
57. Newgard, C. B., Quaade, C., Hughes, S.D., and Milburn, J. L., Glucokinase and
glucose transporter expression in liver and islets: implications for control of glucose
homeostasis, Biochem. Soc. Trans., 18, 851, 1990.
58. Nakashima, R. A., Mangan, P. S., Columbini, M., and Pedersen, P. L., Hexokinase
receptor complex in hepatoma mitochondria: Evidence from N ,N' -dicyclohexylcarbodi-
imide-labeling studies for the involvement of the pore-forming protein VDAC, Biochem-
istry, 25, 1986.
59. Hayzer, D. J, and Iynedjian, P. B., Alternative splicing of glucokinase mRNA in rat
liver, Biochem. J., 270, 261, 1990.
Magnuson and Jetton 167
60. Quaade, C., Hughes, S. D., Coats, W. S., Sestak, A. L., lynedjian, P. B., and
Newgard, C. B., Analysis of the protein products encoded by variant glucokinase tran-
scripts via expression in bacteria, FEBS Lett., 280, 47, 1991.
61. Tanizawa, Y., Koranyi, L. 1., Welling, C. M., and Permutt, M. A., Human liver
glucokinase gene: cloning and sequence determination of two alternately spliced cDNAs,
Proc. Nat/. Acad. Sci. U.S.A., 88, 7294, 1991.
62. Jetton, T. L. and Magnuson, M. A., unpublished data.
63. Magnuson, M. A., Tissue-specific regulation of glucokinase gene expression, J. Cell.
Biochem., 48,115,1992.
Chapter 7
TABLE OF CONTENTS
I. Overview ........................................................ I 70
V. Summary ........................................................ I 83
0-8493-6961-4i93t$0.00 +$.50
© !993 by CRC Press, Inc. 169
170 Nutrition and Gene Expression
I. OVERVIEW
LPK
•
GK
p-Actin
0 2 4 6 10 16 0 2 4 6 10 16
Treatment time (h)
FIGURE 1. Time courses of induction by insulin or dietary fructose of L-type pyruvate kinase
and glucokinase mRNAs in diabetic rat liver. LPK, L-type pyruvate kinase; GK, glucokinase.
TABLE 1
Effects of Various Carbohydrates on the Level
of L-Type Pyruvate Kinase mRNA in Liver,
Kidney, and Small Intestine
tissues. 19 The time courses of these inductions were very similar to that
observed in diabetic liver. Glycerol feeding also increased the mRNA level
in the kidney after 6 h, but did not affect the mRNA level in the small
intestine. Glucose induced a slight but significant (1.6-fold, p < 0.001)
increase in the level of the L-type PK mRNA in the small intestine, but not
in the kidney. No other carbohydrates, including galactose, sorbitol, xylose,
and xylitol, were effective inducers of the L-type PK mRNA in the kidney
or small intestine or in the liver of diabetic rats. Thus, the tissues in which
fructose and glycerol induce the L-type PK mRNA correspond to the tissues
in which these compounds are metabolized, supporting the metabolite hy-
pothesis mentioned above. The candidate molecule may not accumulate sig-
nificantly during metabolism of any other carbohydrate except glucose.
We examined the mechanism of induction of the L-type PK mRNA by
insulin and dietary carbohydrates in diabetic rat liver using a nuclear tran-
scription assay. No significant change in gene transcription was observed 3
h after insulin treatment. 13 As shown in Figure 2, however, insulin enhanced
the rate of transcription of this gene about 28-fold after 12 h. 13 • 18 This stim-
ulation was inhibited by cycloheximide, suggesting that insulin may stimulate
transcription of the L-type PK gene by stimulating the synthesis of some
unknown protein. On the other hand, only four- to sixfold stimulation of the
gene transcription was observed 4 h after fructose or glycerol feeding. 18 • 19 On
the contrary, Munnich et al. reported that dietary fructose stimulated tran-
scription of the L-type PK gene in the liver of normal rats. 16 We tested the
Noguchi and Tanaka 173
••
M2PK
LPK
pUC18
FIGURE 2. Effects of insulin, fructose, and glycerol on transcription of pyruvate kinase genes.
LPK, L-type pyruvate kinase; M2PK, M2-type pyruvate kinase.
(A)
Transcription rate
(Fold chan ge)
r- Normal ---, . - - - - - Diabetic
~
~
~
~ ..g4' ~
()
§ .5'
(j ~ . §
(j
• •
M PK
2
LPK
pUC18
FIGURE 3. Effects of fructose on transcription of pyruvate kinase genes in normal and diabetic
liver. Data are normalized to the value for control rats. LPK, L-type pyruvate kinase; M2 PK,
M2-type pyruvate kinase.
possibility that this difference was due to a difference in the condition of the
animals used, that is, diabetic or normal. 19 Feeding fructose for 4 h resulted
in only 2.4-fold stimulation of the transcription of the L-type PK gene in
diabetic rats, but about 27-fold increase in its transcription in normal rats
(Figure 3). In contrast, the extent of increase in the L-type PK mRNA levels
was similar in these two conditions. To determine whether this difference in
174 Nutrition and Gene Expression
(A)
Transc ription rate
(Fold c hange)
r Normal ---, Diabetic
()-
.$
.,"' ()- .,"' "'
.$"' ~
()-
0<:-
~
.:§'
~
0<:- 8
..._;:; .§' ,k
(j l<.' (j () l<.' (;'
M PK
1
LPK
pUC18
(B)
mRNA level
(Fold change)
LPK 1. 0 5. 5 1.0 0.8 4. 4 12 . 1
± 0. 1 ± 0. 4 ± 0. 1 ± 0. 1 ± 0. 4 ~ 1. 1
FIGURE4. Effects of fructose and glycerol on transcription of pyruvate kinase genes in normal
and diabetic kidney. Data are normalized to the control values. LPK, L-type pyruvate kinase;
M,PK, M 2-type pyruvate kinase.
so
Ql 40
01
r::
nl
.c. 30
u
"C
0
u. 20
10
0
Time after insulin addition (h)
FIGURE 5. Time courses of induction by insulin of L-type pyruvate kinase mRNA in the
presence of various carbohydrates. Hepatocytes were cultured with 0.1 fLM insulin in the presence
of 28 mM glucose (e), 5.6 mM fructose (O), or 5.4 mM glycerol (.") for the indicated times.
Data are expressed relative to the value at zero time.
176 Nutrition and Gene Expression
Pyruvate
0
5
4
3
2
.
Ql
01
c: 5
.!:
u 4
"'0
3
0
u. 2
0
40
30
20
10
0
Concentration (mM)
LPK GK p-Actin
• • Control
• Glucose
• • Glucose + insulin
• • 2-Deoxyglucose
• 2- Deoxyglucose + insulin
liver as described above. The reason why the effects of fructose in normal
and diabetic liver were not reproduced in cultured hepatocytes is not known .
Quantitative and/or qualitative reproduction of the in vivo metabolism of
fructose and glycerol in a culture system may be difficult because the effect
of fructose in vivo is attributable to a common metabolite of fructose and
glycerol. In any case, these results suggest that the metabolism of glucose is
required for the induction of the L-type PK mRNA by insulin . If so, the effect
of insulin should not be observed in the presence of 2-deoxyglucose, a non-
metabolizable glucose analog. As shown in Figure 7, thi s was, in fact, the
case: the effect of insulin was not observed in hepatocytes cultured in a
medium containing 2-deoxyglucose. This was not due to a nonspecific effect
of 2-deoxyglucose because the induction of GK mRNA by insulin was not
impaired by this glucose analog. In fact, the effect of insulin on GK mRNA
did not require the presence of glucose itself. Thus , we suggest that some
metabolite accumulating during the metabolism of glucose is necessary for
the induction of the L-type PK mRNA by insulin and that this metabolite can
be produced from fructose and glycerol to some extent, but not from pyruvate,
in cultured hepatocytes. However, the role of insulin in the induction remains
to be determined. Insulin may simply increase the concentration of this me-
tabolite by stimulating glucose metabolism, or, in addition to this effect,
another signal from insulin may also be necessary.
Consistent with in vivo studies, cycloheximide inhibited the induction of
the L-type PK mRNA by insulin , s uggesting that ongoing protein synthesis
is required for the effect of insulin on the L-type PK. The addition of H-7,
an inhibitor of protein kinase C, caused dose-dependent inhibition of the
induction of L-type PK mRNA by insulin . However, phorbol 12-myristate
13-acetate did not increase the level of the mRNA . Thus, the effect of H-7
may not involve protein kinase C, although the possibility cannot be ruled
178 Nutrition and Gene Expression
TABLE 2
Effects of Various Hormones on the Level
of L-Type Pyruvate Kinase mRNA in
Cultured Hepatocytes
Amount of mRNA
(fold change)
Treatment Pyruvate kinase 13-Actin
out that kinase C may be involved in the action of insulin but not sufficiently
so to induce the L-type PK mRNA by itself.
As mentioned above, another group reported that glucocorticoids and
thyroid hormones are involved in dietary induction of the L-type PK mRNA
by acting at a posttranscriptional level. 21 If this is the case in cultured he-
patocytes, the additions of these hormones alone to cultured hepatocytes
should cause some increase in the L-type PK mRNA level. As shown in Table
2, dexamethasone alone slightly decreased the mRNA level. This is consistent
with the in vivo findings that adrenalectomy resulted in a slight elevation of
the L-type PK mRNA level in the liver. 13 Interestingly, however, dexameth-
asone greatly enhanced the induction of the mRNA by insulin when added
with insulin. As glucocorticoids are known to be necessary to maintain the
differentiated function of hepatocytes in vitro, 23 this hormone may act to
maintain the levels of hepatocyte-specific transcription factors in vitro. Con-
sistent with in vivo findings, T 3 had no effect on the L-type PK mRNA level
either alone or with dexamethasone or on the induction of the mRNA by
insulin/dexamethasone. However, this hormone had a permissive effect on
the induction of GK mRNA by insulin. 8-CPT-cAMP, a stable analog of
cAMP, inhibited the induction of the mRNA by insulin, confirming the in
vivo results.
Noguchi and Tanaka 179
1. Liver
2. Kidney
3. Small intestine
4. Brain
5. Muscle
6. Spleen
7. Testis
8. Erythrocytes
The in vivo and in vitro studies described above suggest that the mech-
anism of transcriptional stimulation of the L-type PK gene by insulin or dietary
fructose is very complicated. In both cases, metabolism of carbohydrates is
involved, and so a common metabolite could be responsible for the induction
of the L-type PK. If this is the case, the same cis- and trans-acting factors
are involved in both cases. To test this possibility, we tried to identify a cis-
acting element responsible for transcriptional stimulation of the L-type PK
gene by insulin or dietary fructose. First, we produced transgenic mice car-
rying the 5' flanking region of the L-type PK gene from about nucleotide
-3000 to + 37 linked to the chloramphenicol acetyltransferase (CAT) struc-
tural gene and determined the CAT activity in various tissues. 24 This activity
was detected in the liver, kidney, and small intestine, but not in other tissues
(Figure 8). Thus the tissues expressing CAT activity were the same as those
expressing the endogenous L-type PK. Dietary glucose or insulin induced
similar increases in the levels of CAT and L-type PK mRNAs in the liver of
transgenic mice (Figure 9). Dietary fructose also caused a 15-fold increase
in the level of CAT mRNA, but this increase extent was significantly less
than that of the L-type PK mRNA (41-fold increase), suggesting that stabi-
lization of the transcripts of the L-type PK gene plays a significant role in
the induction by fructose even in normal liver. Thus, the sequence of about
3 kb upstream of the L-type PK gene contains all the cis-acting elements
responsible for tissue-specific expression of this enzyme and its transcriptional
stimulation by dietary carbohydrates and insulin. Dietary fructose, but not
glucose, also induced a marked increase in the L-type PK mRNA level in
the kidney, but induced only a marginal increase in the level of CAT mRNA.
These results are consistent with our finding mentioned above that posttran-
180 Nutrition and Gene Expression
Normal
CAT
LPK
p - Actin
FIGURE 9. Northern blot analysis of liver RNA of transgenic mice after administration of
glucose or fructose diet or insulin for 16 h. LPK, L-type pyruvate kinase.
Transcription
CGCCACGGGGCACTCCCGTGGTTCCTGGACTCTGGCCCCCAGTGT
(l-IIIF) (L-IIF)
FIGURE 10. Schematic representation of the regulatory regions of the rat pyruvate kinase
gene.
-e2 Relative
L-111 L-11 L-1 ~ activity(%)
-
_.,.. 3. 3 ± 0. 7
-- -
2. 7 ± 0.2
_.,.. 0. 11 ± 0. 01
0. 10 ± 0. 10
0. 15 ± 0.05
0.60 ± 0.26
2.0 ± 0.8
-- -
1.3 ± 0.6
_.,.. 21. 2 ± 6. 7
--- -- -
5.0 ± 1.2
_.,.. _.,.. 2. 8 ± 1.2
_.,.. 4.1 ± 1.6
-- -
66.3 ± 26.8
_.,..
---
25. 7 ± 12.8
7. 5 ± 3. 2
27. 1 ± 17.6
plcat62 0.24 ± 0.23
plcat189 100
FIGURE ll. Interaction of three cis-acting elements of the L·type pyruvate kinase gene. A
single copy of each double-stranded oligonucleotide was ligated to pLcat62 in the orientation
shown by arrows on the left, which is the 5' to 3' direction. The relative activities of the
constructs are expressed as percentages of that of pLeat 189, which includes the region up to
-189.
182 Nutrition and Gene Expression
Relative
-==:J.
~
()
activity (%)
A)
-63 -185 ~- 100
II 1700
8)
-63 -185
IT=riill. 100
~- II 2400
•1-
r
C) I IC A Tl <0.5
-63 -185
s I 100
FIGURE 12. Interaction of the regulatory regions with heterologous promoter. A fragment
containing the three elements was inserted upstream (A) or downstream (B) of the SV40 early
promoter, or upstream of the pyruvate kinase M gene promoter (C) in the orientation shown by
the arrows, which are the 5' to 3' direction.
V. SUMMARY
in the body. In turn, the genes for these isozymes are different in their
responsiveness to the kind of dietary carbohydrate and the hormones which
influence its expression. The identification of the cis-acting regulatory region
of the liver type pyruvate kinase gene has yet to be resolved.
REFERENCES
I. Imamura, K., Noguchi, T., and Tanaka, T., Regulation of isozyme patterns of pyruvate
kinase in normal and neoplastic tissues, in Markers of Human Neuroectodermnl Tumors,
Staal, G. E. J. and van Veelen, C. W. M., Eds., CRC Press, Boca Raton, FL, 1986,
chap. 13.
2. Noguchi, T., Yamada, K., Inoue, H., Matsuda, T., and Tanaka, T., The L-type and
R-type isozymes of rat pyruvate kinase are produced from a single gene by use of different
promoters, J. Bioi. Chern., 262, 14366, 1987.
3. Noguchi, T., Inoue, H., and Tanaka, T., TheM,- and M,-type isozymes of rat pyruvate
kinase are produced from the same gene by alternative RNA splicing, J. Bioi. Chern.,
261, 13807, 1986.
4. Takenaka, M., Noguchi, T., Inoue, H., Yamada, K., Matsuda, T., and Tanaka, T.,
Rat pyruvate kinase M gene. Its complete structure and characterization of the 5 '-flanking
region, J. Bioi. Chern., 264, 2363, 1989.
5. Goodridge, A. G., Dietary regulation of gene expression: enzymes involved in carbo-
hydrate and lipid metabolism, Annu. Rev. Nutr., 7, 157, 1987.
6. Van Den Berghe, G., Metabolic effects of fructose in the liver, in Current Topics in
Cellular Regulation, Vol. 13, Horecker, B. L. and Stadtman, E. R., Eds., Academic
Press, New York, 1978, 97.
7. Gunn, J, M. and Taylor, C. B., Relationships between concentration of hepatic inter-
mediary metabolites and induction of the key glycolytic enzymes in vivo, Biochem. J.,
136, 455, 1973.
8. Takeda, Y., Inoue, H., Honjo, K., Tanioka, H., and Daikuhara, Y., Dietary response
of various key enzymes related to glucose metabolism in normal and diabetic rat liver,
Biochim. Biophys. Acta, 136, 214, 1967.
9. Fukuda, H., Iritani, N., and Tanaka, T., Effects of high-fructose diet on lipogenic
enzymes and their substrate and effector levels in diabetic rats, J. Nutr. Sci. Vitaminol.,
29, 691, 1983.
10. Adelman, R. C., Spotter, P. D., and Weinhouse, S., Dietary and hormonal regulation
of enzymes of fructose metabolism in rat liver, J. Bioi. Chern., 241, 5467, 1966.
11. Noguchi, T., Inoue, H., and Tanaka, T., Regulation of rat liver L-type pyruvate kinase
mRNA by insulin and by fructose, Eur. J. Biochem., 128, 583, 1982.
12. Noguchi, T., Inoue, H., Chen, H., Matsubara, K., and Tanaka, T., Molecular cloning
of DNA complementary to rat L-type pyruvate kinase mRNA. Nutritional and hormonal
regulation of L-type pyruvate kinase mRNA concentration, J. Bioi. Chern., 258, 15220,
1983.
13. Noguchi, T., Inoue, H., and Tanaka, T., Transcriptional and post-transcriptional reg-
ulation of L-type pyruvate kinase in diabetic rat liver by insulin and dietary fructose, J.
Bioi. Chern., 260, 14393, 1985.
14. Iynedjian, P. B., Gjinovci, A., and Renold, A. E., Stimulation by insulin of glucokinase
gene transcription in liver of diabetic rats, J. Bioi. Chern., 263, 740, 1988.
Noguchi and Tanaka 185
15. Iynedjian, P. B., Ucla, C., and Mach, B., Molecular cloning of glucokinase eDNA.
Developmental and dietary regulation of glucokinase mRNA in rat liver, J. Bioi. Chern.,
262, 6032, 1987.
16. Munnich, A., Lyonnet, S., Chauvet, D., Van Schaftingen, E., and Kahn, A., Dif-
ferential effects of glucose and fructose on liver L-type pyruvate kinase gene expression
in vivo, J. Bioi. Chern., 262, 17065, 1987.
17. Inoue, H., Noguchi, T., and Tanaka, T., Rapid regulation of L-type pyruvate kinase
mRNA by fructose in diabetic rat liver, J. Biochern. (Tokyo), 96, 1457, 1984.
18. Noguchi, T. and Tanaka, T., unpublished data, 1989.
19. Matsuda, T., Noguchi, T., Takenaka, M., Yamada, K., and Tanaka, T., Regulation
of L-type pyruvate kinase gene expression by dietary fructose in normal and diabetic rats,
J. Biochern. (Tokyo), 197, 655, 1990.
20. Vernon, R. G. and Walker, D. G., Glycerol metabolism in the neonatal rat, Biochern.
J., 118,531, 1970.
21. Vaulont, S., Munnich, A., Decaux, J.-F., and Kahan, A., Transcriptional and post-
transcriptional regulation of L-type pyruvate kinase gene expression in rat liver, J. Bioi.
Chern., 261, 7621, 1986.
22. Matsuda, T., Noguchi, T., Yamada, K., Takenaka, M., and Tanaka, T., Regulation
of the gene expression of glucokinase and L-type pyruvate kinase in primary cultures of
rat hepatocytes by hormones and carbohydrates, J. Biochern. (Tokyo), 108, 778, 1990.
23. Feliu, J. E., Coloma, J., Gomez-Lechon, M.-J., Garcia, M.D., and Baguena, J.,
Effect of dexamethasone on the isozyme pattern of adult rat liver parenchymal cells in
primary cultures, Mol. Cell. Biochern., 45, 73, 1982.
24. Yamada, K., Noguchi, J., Miyazaki, J., Matsuda, T., Takenaka, M., Yamamura,
K., and Tanaka, T., Tissue-specific expression of rat pyruvate kinase Llchloramphenicol
acetyltransferase fusion gene in transgenic mice and its regulation by diet and insulin,
Biochern. Biophys. Res. Cornrnun., 171, 243, 1990.
25. Yamada, K., Noguchi, T., Matsuda, T., Takenaka, M., Monaci, P., Nicosia, A.,
and Tanaka, T., Identification and characterization of hepatocyte-specific regulatory
regions of the rat pyruvate kinase L gene. The synergistic effects of multiple elements,
J. Bioi. Chern., 265, 19885, 1990.
26. Noguchi, T., Yamada, K., and Tanaka, T., unpublished data, 1989.
27. Frain, M., Swart, G., Monaci, P., Nicosia, A., Stampfli, S., Frank, R., and Cortese,
R., The liver-specific transcription factor LF-Bl contains a highly diverged homeobox
DNA binding protein, Cell, 59, 145, 1989.
28. Chouard, T., Blumenfeld, M., Bach, 1., Vandekerhove, J., Cereghini, S., and Yaniv,
M., A distal dimerization domain is essential for DNA binding by the atypical HNFl
homeodomain, Nucleic Acids Res., 18, 5853, 1990.
29. Mendel, D. B., Hansen, L. P., Graves, M. K., Conley, P. B., and Crabtree, G. R.,
HNF-lo: and HNFl J3 (vHNF-1) share dimerization and homeodomains, but not activation
domains, and form heterodimers in vitro, Genes Dev., 5, 1042, 1991.
30. Hard on, E. M., Frain, M., Paonessa, G., and Cortese, R., Two distinct factors interact
with the promoter regions of several liver-specific genes, EMBO J., 7, 1711, 1988.
31. Chodosh, L. A., Carthew, R. W., Morgan, j. G., Crabtree, G. R., and Sharp,
P. A., The adenovirus major late transcription factor activates the rat -y-fibrinogen pro-
moter, Science, 238, 684, 1987.
32. De Simone, V., De Magistris, L., Lazzaro, D., Gerstner, J., Monaci, P., Nicosia,
A., and Cortes, R., LFB3, a heterodimer-forming homeoprotein of the LFBl family,
is expressed in specialized epithelia, EMBO J., 10, 1435, 1991.
33. Rey-Campos, J., Chouard, T., Yaniv, M., and Cereghini, S., vHNFl is a borneo-
protein that activates transcription and forms heterodimers with HNFl, EMBO J., 10,
1445, 1991.
186 Nutrition and Gene Expression
34. Noguchi, T., Miyazaki, J., Yamamura, K., and Tanaka, T., unpublished data, 1991.
35. Thomson, K. S. and Towle, H. C., Localization of the carbohydrate response element
of the rat L-type pyruvate kinase gene, J. Bioi. Chern., 266, 8679, 1991.
36. Cognet, M., Bergot, M.-0., and Kahn, A., cis-Acting elements regulating expression
of the liver pyruvate kinase gene in hepatocytes and hepatoma cells. Evidence for tissue-
specific activators and extinguisher, J. Bioi. Chern., 266, 7368, 1991.
Chapter 8
TABLE OF CONTENTS
0·8493·6961·4193/$0.00+$.50
© 1993 by CRC Pre». Inc. 187
188 Nutrition and Gene Expression
I. INTRODUCTION
TABLE 1
Influence of Diet Composition on the Percent Increment
in Hepatic G6PD Activity Due to Starvation-Refeeding
broad range in values for some of the diets. In part, this is due to the variation
within strains and in part to the variation in response between strains. Some
rats, notably those of the Wistar strain, are relatively unresponsive to star-
vation-refeeding, whereas Sprague-Dawley rats are more responsive to this
feeding paradigm. BHE rats are more responsive than Sprague-Dawley rats. 15
Not only are there differences due to the strain in enzyme activity, there are
also differences attributable to the diet fed the rat prior to its use in a starvation-
refeeding experiment, and, too, its sex and its age. If rats were fed a high
fat diet prior to use, the increment in enzyme activity was twice as high as
in rats fed a standard stock diet prior to use. 17 Middle-aged rats ( -12 months
of age) were similar to young rats 18 • 19 in their response to starvation-refeeding,
while aged rats (22 months of age) were less responsive to this treatment than
their younger (2 months old) cohorts. 2° Female rats are less responsive to
starvation-refeeding than are male rats. 21 This gender difference in response
is probably due to the fact that female rats have a higher G6PD activity in
the basal ad libitum fed state. In part, this is due to the cyclical presence of
estrogen which imposes a cyclical pattern of reductions and increases in food
intake. Thus, postpubescent female rats experience a 5-day food intake de-
crease/increase cycle which, in effect, is a modified starve-refeed cycle.
Prepubescent female rats do not evidence an estrus cycle and also do not have
cyclical variations in food intake. Estrogen has also been shown to be essential
to the induction of G6PD activity in the uterus. 22
Variation in food supply, as well, can affect the G6PD response to star-
vation-refeeding. Rats that are deprived of food during shipping have a larger
190 Nutrition and Gene Expression
50
" 0 Intact
~ ADX
. 40 Intact + 8AZ
....
.<:
"'
·;; ADX + 8 AZ
5:
>
-o 30
0
CD
(!)
0
~ 20
.~
c:
=>
10
FIGURE 1. Hepatic G6PD activity in ad libitum fed and starved-refed Sprague-Dawley rats.
Rats were fed or refed a 65% glucose diet. The starvation period and the refeeding period were
48 h each; 8 azaguanine was administered daily. Abbreviations: GC, glucocorticoid; 8 AZ, 8-
azaguanine. Data are from References 33 and 34.
The studies carried out with intact animals have demonstrated that a
variety of nutrients and hormones are either directly or indirectly involved
with the regulation of hepatic G6PD. However, it is often difficult to distin-
guish direct vs. indirect involvement of hormones or nutrients on gene expres-
sion in intact animals. For example, in attempting to determine whether insulin
or glucose is a primary signal for induction of G6PD, experimentation in
animals is compromised since blood glucose is maintained at 5 mM, and
elevating it by feeding glucose causes insulin release. Eliminating insulin by
making the animal diabetic results in high levels of glucagon which cause an
elevation of hepatic cAMP and gluconeogenesis, conditions that might repress
G6PD. The well-known induction of G6PD following injection of insulin into
the animal could be the result of a positive influence of insulin on the G6PD
gene or it could be result of derepression of the influence of glucagon. Thus,
direct assignment of the responsibility for control of G6PD expression requires
the use of less complex systems than the intact animal.
192 Nutrition and Gene Expression
In order to sort out the various factors involved in the regulation of hepatic
G6PD, we have employed primary cultures of liver parenchymal cells main-
tained in chemically defined, serum-free medium. In this system, cells retain
the terminally differentiated phenotype for a discrete period of time (up to
several weeks or months, depending on culture conditions), 37 - 39 in contrast
to hepatoma cell lines which seldom display the full realm of differentiated
functions expected of a hepatocyte. Thus, metabolic responses of the primary
cultures to hormones or nutrients can be expected to mimic that observed in
vivo. With this system it is then possible to add hormones or nutrients,
individually or in combination, to define precisely the involvement of these
factors in the control of G6PD expression. Several groups have employed the
hepatocyte primary culture system in studies on G6PD regulation. 40 -45 While
culture conditions were not identical, some common themes have emerged
and some areas of disagreement are apparent. This review will attempt to
analyze and summarize results on nutrient and hormonal control of G6PD
from the six laboratories 40-45 employing this culture system.
An additional development that has greatly aided analysis of G6PD expres-
sion is the availability of molecular probes for the mRNA encoding rat liver
G6PD. 46 The complete sequence of the protein 47 and mRNA 48 has been es-
tablished and genomic fragments have been isolated. 49 These sequences along
with high titer antibodies against G6PD provide all of the necessary tools to
analyze expression in either animals or in cells in culture. Thus, it is now
possible to dissect in a molecular fashion the effects of the hormones or
nutrients on the control of expression. This is particularly important in those
situations where a hormone or nutrient might increase transcription of the
G6PD gene, but a different hormone might be required for the optimum rate
of translation of that mRNA species. If one simply measures enzyme activity
or enzyme protein by immunochemical techniques, this sort of interaction
would not be observed. Therefore, combining these molecular tools with the
primary culture model should provide precise information on the control of
G6PD expression.
Insulin has long been considered the primary signal for induction of G6PD
in addition to the other lipogenic enzymes in animals. Kurts and Wells 45 and
Nakamura et al. 41 were the first to demonstrate that insulin could induce
G6PD in primary cultures of hepatocytes. Subsequently, others extended this
observation to show that insulin increased the relative rate of G6PD synthesis 5°
and mRNA encoding G6PD. 50 ·51 The ability of insulin to induce G6PD is not
rapid in onset, 50 taking 36 to 48 h to reach its maximum effect. However,
induction of G6PD in vivo following feeding a high carbohydrate diet is also
a relatively slow process, taking 48 to 72 h to reach maximal levels of
Kletzien and Berdanier 193
activity. 49 ·52 Thus, although the absolute change in enzyme activity observed
in tissue culture following insulin treatment is not as great as that observed
in the intact animal following carbohydrate refeeding, the time course is quite
similar. This lends strength to the argument that insulin is the primary signal
involved but suggests that other factors may be involved in amplification of
the insulin response. The six laboratories 40 -45 that have utilized primary cul-
tures of hepatocytes are in agreement with the primary role of insulin in
control of G6PD expression.
The precise level at which insulin regulates G6PD expression has not
been established. The fact that insulin elevates G6PD mRNA in cultured
hepatocytes suggests that transcriptional regulation of the G6PD gene may
be a critical site of regulation. In addition, two studies employing transcription
"run on" assays from rat liver nuclei isolated from fasted-refed animals have
demonstrated that there is a transient activation of G6PD gene transcrip-
tion.49·53 However, it is also possible that insulin stabilizes the G6PD mRNA
and/or causes a selective translation of it. Changes in G6PD mRNA stability
following refeeding a high carbohydrate diet have been observed in rats. 49
The study of how insulin accomplishes regulation of G6PD and other insulin
regulated genes remains as a major area of interest for molecular endocri-
nologists.
The glucocorticoids have a well established role in lipogenesis (for review
see Reference 54), and, thus, the interaction of insulin and the glucocorticoids
in regulation of G6PD has been explored in the hepatocyte primary culture
model. The results from these studies have shown that some differences are
apparent which are perhaps the result of culture conditions. Kurtz and Wells 45
first demonstrated that a synthetic glucocorticoid, dexamethasone (Dex), in-
duced G6PD activity by itself and its effect was additive with insulin. In
addition, the study 45 suggested that the glucocorticoid played a critical ''per-
missive" role for insulin induction of G6PD since insulin failed to increase
G6PD in hepatocytes from adrenalectomized rats unless Dex was added to
the cultures. Kletzien and co-workers50 ·5l.5 5 concluded that while the gluco-
corticoids enhanced the insulin induction of G6PD activity, it exerted its main
effect on mRNA levels encoding G6PD. Using molecular probes and a high
titer, monospecific antibody against G6PD, they demonstrated that Dex ap-
pears to increase the mRNA encoding G6PD but translation of the active
protein does not occur unless insulin is also present. 50 ·51 Holten and co-workers
have also demonstrated that maximum induction of G6PD in the primary
culture model requires the presence of a glucocorticoid. 42 Thus, it would seem
that this class of hormones is involved in at least a permissive way, in reg-
ulating G6PD expression.
In contrast to the above results, two groups 41 .43 have reported that the
glucocorticoids are not involved in regulation of G6PD. However, Ichihara
and colleagues 41 maintained their cultures in the continuous presence of glu-
194 Nutrition and Gene Expression
The consensus sequence for rat G6PD derived from eDNA and genomic
clones is presented in Figure 2. We have previously shown that fragments of
this sequence will hybrid select mRNA encoding G6PD46 • Recently, partial
protein sequence for rat liver G6PD has been published47 allowing direct
comparison with our nucleic acid sequence. For the 71 amino acid amino
terminal sequence published by Jeffery et a!. ,47 there exists 100% identity
with our deduced amino acid sequence from the 5' coding region, unequiv-
ocally establishing this consensus sequence for rat G6PD. In addition, com-
parison of our consensus sequence with one of the published sequences for
198 Nutrition and Gene Expression
1 2 3
9.4-
6.7-
4.4-
2. 3-
2 . 0-
0.56-
FIGURE 2. Southern blot of rat liver chromosomal DNA. Rat liver chromosomal DNA was
digested with Pstl, Eco Rl, and Bam HI (Lanes I, 2, and 3). The blot was probed with a
nicknamed eDNA from pGDY-38 46 which is a 3' untranslated region probe. The 9.4 and 9 kb
(lanes 2 and 3) fragment is the functional gene and the 6.7 and 6.2 kb (lanes 2 and 3) fragment
is the pseudogene.
TABLE 2
Comparison of Rat/Human Exon/Intron Boundaries
Note: The rat sequence is shown on the top line, and in those positions where the human sequence
is different, the base present in the human sequence is on the bottom line.
signal, and in two other areas within this region are highly conserved between
rat and human.
While the human genome apparently does not contain a G6PD pseudo-
gene,71 a processed pseudogene for G6PD is present (Figure 2, 6.7 kb) in
the rat genome and is more highly represented than the single copy of the
functional gene (Figure 2, 9.4 kb). A genomic clone was isolated which
contained a 6.7 Eco Rl fragment within which was a 3.2 kb sequence having
the characteristics of a processed pseudo gene for G6PD. Sequence analysis
(Figure 3) revealed the absence of introns and the presence of a poly A tail,
indicative of a pressed pseudogene. As in the case of other processed pseu-
dogenes, a short direct repeat (GAAAAGACAA) which probably represents
the site of insertion was located flanking the sequence. It occurs immediately
3' of the poly A tail and 65 base pairs 5' of the "translation start" site.
Numerous base substitutions were found throughout the coding, 5' and 3'
untranslated regions. The overall sequence identity between the pseudogene
and consensus sequence is 95.8%.
200 Nutrition and Gene Expression
•I•
COliS 6T&MI:6T6nT6&U61:&&tAACTAAAmASAMAUTIIA6AIIU56TS&tTTT6A6CC66ACCCA6&T6T6T6&6ATC 41
I I I
P6 ~C666CTAT6CAA!ttA&AltliT6AACiliTTT66CA6CA&tAACTAAATTtASAAAACAT A6A6tA&ATi&CTTTiA&tTS&AtCCA&iT616T666ATC
I I
CONS CT~&IM6AITTiTACtA666T&AT6tCmUCW&CT&ATACAUC.ITATTTATtATI:ATi&ST6CATmiT6ACt166ttAA6AA&AA&ATTTATmAmTCT66T6SCT6 161
I I II II
Pi CTM6&1M6Ain&TACCAT&ST&ATSCCmCioCAAS&n&ATACACA--TATTTATCATCAT666T6tATCS66T&ACCT66tCAAiAASAA&ATTTATCCTACCATCTS6T66CTS
TTI:t&&&AT66ttTTCIACctiAASoiCACtTTI:ATl6TA66tTAT6ttC6CTCAC&ICTCACA6166AI6ACATCC6CAAACA6A6~6AitcrTTCTTTAAAI61CACTCCA6AASAAASA 1111
I I I I I
Pi TTm&&AT&6CtTTIIACUAAA&ACACC1TtATTSTA66CTATim&nCAT&ACTCACA6T&SA16ACATCC6CAACCAiA616A&CCCTTCTTTAAAiltACTCCASAASAAA6A
COliS A6Att16CA6A6CTCCAATCAACT6Tt6AACCACATtltt!CTtT6TTIC616A66ACCA6ATtTACC6CATT&ACCAC1AtCT&66CAAA6ASATSSTCCAiAACCTCATSSTSCTGAS Ml
I 111111111111
Pi A6ACCmA6A6CTCCAATCAACT&n6AoiCUCATCTmmT&TTTC&T6A66ACCA6ATtTACC6CATTSACCACTAttT66------------TCCA6AAmW&STSCTSA6
I
CONS ATTTSCI:AACA66ATCTT!&SACCCATCTi&AATC~ACAACATT6tTTST-----iliATCCTIACATTIAAA6A6CmTT6iTACISASSSTCSTSSS66tTATTTT&AT&AA 161
I 11111111 I I
P6 ATTI6tCAACA66ACCTTT&SACCCATCI&&AATC6A6ACAACATTSCTTSTmmTT&T&AmTTACATTTAAAiAStcCTT!iATACI&Ai66TTST&S&S&nATTTT6ATSAA
I I
l:llNS TTT666ATI:ATCA666AT6TI:AT&CA6AACCAUTCCTSCA6AT6TT6TmTA&T&SCCAT66AAAA6CCTSCmTACA6ATTI:A&ATiATSTCC6TSAT6A6AA661CAAA616TTA 1181
I II I I I I
Pi TTTA66ATCATA66&SAT6TTATACA&MI:CACmcmA&AT&TT6TiTCTA6166ttAT66AAAA6CCT6tCTtTACA6ATTCAAAT6ATiTCCA16Al&A6AAi61CAAAST6TT A
TTWC&AT6T&itA6&T&ACATCTTCI:ACI:A&CA616tAASCiTAAC&A&CT66TCATCC6TiTii:A6tcrAAT6A6&t:i6TATACACI:AA6AT&AT6ACCAA&AASI:tT6&CAT&TTC 1141
I I I I IIIII I I
P& TTCTIC&AT&T&itA6&T&ACATCTTCCACCA&CA6T61:AAST6TAoiCAA6CT66TCATCTiT6T6CA--AT&A66C6&TATACACCAA&AT&AT&ACCAA&AACCCT66tCT6TTC
I
TTCAACttT6A66A&Tti&A61:T&iAmAACCTAT66CAAI:A&ATACAA&AAT6T&AA&CTI:ttT6ATSCCTAT6AAC6CCTCATCCT6&AT6TCTTCT61666A6tCAAAT&CACTTT 1365
11111111111111111111111111111111111111111111111111111111111111111111111
P& TTCAAI:ttT6A66A&TCT&A6CT&ilmAACCTAT&iCAACA6A-----------------------------------------------------CTTT
em TTSACAna:rccrcACCA&CT&iAA&Ao\CTCTCATmmrmAATATTTT&&&&imTASATArcrmAA!tAAnmrAsrccArA&TCA&ccrcArccAACCCAT&&&eA& 1101
II I I
P& TT6ACATTCCTCCICACCA61:166AA&AAACCTCAT6tT6CCTA&C6ATATTTT66666tCATA6ATATCTCCTAAACAATTCCATA&TCCATA6TCAAttTCATCCAACCCAT666CA6
The coding region and intron/exon boundaries were sequences from the
genomic fragments containing the functional G6PD gene from rat liver (Table
2 and Figure 3). The structure of the rat liver gene was found to be remarkably
similar to that of the human gene as reported by Martini et a!. 71 The number
of exons (13), the position of the introns in the coding sequence, and the size
of the corresponding exons are identical in both genes. The intron boundaries
(indicated by arrows in Figure 3) are highly conserved although intron size
varies considerably (Table 2). IVS I does not intersect the coding region, and
the exon/intron junction at this site in the 5' untranslated region is not con-
served. Exon 13 contains all of the 3' untranslated region, the termination
codon, and 88 base pairs of coding sequence.
Comparison of the rat consensus G6PD sequence with that of the human
demonstrates a high degree of homology except at the sequences encoding
the amino terminal end of the protein. However, two different sequences
have been reported for the human amino terminal sequence of G6PD. One
was determined by sequence analysis of the protein from red blood cells 67
and the other deduced from cDNAs produced from a variety of human cell
types and transformed celllines. 69 •70 The sequence reported by Jeffery et al. 47
for the rat liver protein corresponds to that deduced from the human cDNAs.
In order to determine the location of this difference in the sequence infor-
mation, we compared eDNA and protein sequence data from rat and human
with that of the G6PD gene. As shown in Figure 4, the point of divergence
is localized to amino acid position #35 in the G6PD protein sequence. At
the nucleic acid level, the site of sequence divergence is immediately 5' of
the IVS 2 region of the gene. IVS 2 is extremely large (> 11 kb in the human
gene) and is the first intron to intersect the coding sequence. In the case of
the rat liver sequence, only 120 bases that encode protein are upstream of
this intron. That this structural feature has been preserved in evolution is
indicative of a possible important structural role in gene function. Either
alternative splicing or use of different promoters as in the case of R-type vs.
L-type pyruvate kinase could give rise to amino terminal heterogeneity for
G6PD.
FIGURE 3. Comparison of the consensus coding sequence for rat G6PDH with a processed
pseudogene. Nucleotide residues are numbered from the start of translation. The consensus
sequence for G6PDH (upper, CONS) is from complete, overlapping eDNA clones from rat liver
and brain libraries and from the coding region of the functional gene. Each clone was sequenced
twice in both directions to verify the authenticity of the sequence. The pseudogene sequence
(lower, PG) starts with the direct repeat (underlined) and ends with the same sequence immediately
3' of the poly A taiL Bases that are different between the two sequences are indicated with a
bar. The polyadenylation signal, the start, and termination of translation are denoted by the box.
The positions of the 12 introns are denoted by arrows.
202 Nutrition and Gene Expression
Human~7:~~~~~~~~~~~~~~~~~~~~~~~~~~~~~
eDNA
Rat
cDNA~~_Q~~~~~~~~~~~~~~~~~~~~~~~~~~~
protein
HUMAN
protein
IVS2
FIGURE 4. Amino terminal protein and 5' coding sequence relationships between rat and
human G6PDH. The rat protein sequence (I) and the human protein (19) and eDNA sequence
(17) are from published reports. Identities in the sequences are posed. The position of the second
intron of the G6PDH gene is noted by the arrow. The rat amino acid positions from 29 to 44
are shown and for the human, positions 46 to 61.
XI. CONCLUSIONS
REFERENCES
1. Tepperman, H. M. and Tepperman, J., The hexose monophosphate shunt and adaptive
hyperlipogenesis, Diabetes, 7, 478, 1958.
2. Glock, G. E. and McLean, P., A preliminary investigation of the hormonal control of
the hexose monophosphate oxidative pathway, Biochem. J., 61, 390, 1955.
3. Fitch, W. M. and Chaikoff, J. L., Extent and patterns of adaptation of enzyme activities
in liver of normal rats fed diet high in glucose and fructose, J. Bioi. Chern., 235, 554,
1960.
4. Rudack, D., Chisholm, E. M., and Holten, D., Rat liver glucose 6 phosphate dehy-
drogenase. Regulation by carbohydrate diet and insulin, J. Bioi. Chern., 246, 1249, 1971.
5. Potter, V. R. and Ono, T., Enzyme patterns in rat liver and Morris hepatoma 5123
during metabolic transitions, Cold Spring Harbor Symp. Quant. Bioi., 26, 355, 1961.
6. Tepperman, J. and Tepperman, H. M., Effects of antecedent food intake pattern on
hepatic lipogenesis, Am. J. Physiol., 193, 55, 1958.
7. Tepperman, H. M. and Tepperman, J., On the response of hepatic glucose 6 phosphate
dehydrogenase activity to changes in diet composition and food intake pattern, Adv.
Enzyme Reg., !, 121, 1963.
8. Sassoon, H. F., Dror, Y., Watson, J. J., and Johnson, B. C., Dietary regulation of
liver glucose 6 phosphate dehydrogenase in the rat: starvation and dietary carbohydrate
induction, J. Nutr., 103, 321, 1973.
9. Szepesi, B. and f'reedland, R. A., Differential requirement for de novo RNA synthesis
in the starved refed rat; inhibition of the overshoot by 8-azaquanine after refeeding, J.
Nutr., 99, 449, 1969.
10. Szepesi, B. and Berdanier, C. D., Time course of the starve-refeed response in rats;
the possible role of insulin, J. Nutr., 101, 1563, 1971.
11. Szepesi, B. and Michaelis, 0. E., Comparison of the fructose effect and the starve-
refeed response of rat liver enzymes, Life Sci., II, 113, 1972.
12. Michaelis, 0. E. and Szepesi, B., Effect of various sugars on hepatic glucose 6 phosphate
dehydrogenase, malic enzyme and total lipid and the rat, J. Nutr., 103, 697, 1973.
13. Williams, B. H. and Berdanier, C. D., Effects of diet composition and adrenalectomy
on the lipogenic responses of rats to starvation-refeeding, J. Nutr., 112, 534, 1982.
14. Baltzell, J. K. and Berdanier, C. D., Effect of the interaction of dietary carbohydrate
and fat on the responses of rats to starvation-refeeding, J. Nutr., 115, 104, 1985.
15. Berdanier, C. D., Johnson, B., and Buchanan, M., Interacting effects of menhaden
oil and sucrose on the lipogenic response to starvation-refeeding, Nutr. Res., 9, 1167,
1989.
16. Johnson, B. J. and Berdanier, C. D., Effect of menhaden oil on the responses of rats
to starvation-refeeding, Nutr. Rep. Int., 36, 809, 1987.
17. Bue, J. and Berdanier, C. D., Effect of antecedent diet on the responses of BHE rats
to starvation-refeeding, Nutr. Rep. Int., 35, 791, 1987.
18. Szepesi, B., Berdanier, C. D., and Michaelis, 0. E., Effect of age on the responses
to starvation-refeeding in the rat, Proc. Soc. Exp. Bioi. Med., 147, 195, 1974.
19. Kim, M.-J., Pan, J.-S., and Berdanier, C. D., Age and thyroid hormone as factors
in the responses of BHE rats to starvation-refeeding, Proc. Soc. Exp. Bioi. Med., 183,
273, 1986.
20. Bois-Joyeux, B., Chanez, M., Aranda-Haro, F., and Peret, J., Age dependent hepatic
lipogenic enzyme activities in starved-refed rats, Diabetes Metab., 16, 290, 1990.
21. Berdanier, C. D., Effects of estrogen on the responses of male and female rats to
starvation refeeding, J. Nutr., 111, 1425, 1981.
22. Smith, E. R. and Barker, K. L., Effects of estradiol and NADP on the rate of synthesis
of uterine glucose 6 phosphate dehydrogenase, J. Bioi. Chern., 249, 6541, 1974.
204 Nutrition and Gene Expression
23. Szepesi, B. and Michaelis, 0. E., Problems in the demonstration of the disaccharide
effect due to stress during shipment of rats, Nutr. Rep. Int., 12, 299, 1975.
24. Michaelis, 0. E., Scholfield, D. J., Nace, C. S., and Reiser, S., Demonstration of
the disaccharide effect in nutritionally stressed rats, J. Nutr., 108, 919, 1978.
25. Szepesi, B., Metabolic memory: effect of antecedent dietary manipulations on subsequent
diet induced responses of rats. I. Effects on body weight, food intakes, glucose 6 phosphate
dehydrogenase and malic enzyme, Can. J. Biochem., 51, 1604, 1973.
26. Szepesi, B., Effect of previous caloric restriction on two NADP-linked dehydrogenases
of rat liver, Nutr. Rep. Int., 10, 127, 1974.
27. Szepesi, B., Berdanier, C. D., Diachenko, S. K., and Moser, P. B., Effect of length
of starvation, refeeding and 8-azaquanine on serum insulin and NADP-linked dehydro-
genases of rat liver, J. Nutr., 101, 1147, 1971.
28. Homan, H. D. and Berdanier, C. D., Effect of adrenalectomy on the enzyme responses
of starved-refed rats, Nutr. Rep. Int., 22, 213, 1980.
29. Gimenez, R. S. and Johnson, B. C., Pair feeding in the dietary control of G6PD, J.
Nutr., Ill, 260, 1981.
30. Freedland, R. A., Cunliffe, T. L., and Zinkl, J, G., Effect of insulin on enzyme
adaptations to diets and hormones, J. Bioi. Chem., 241, 5448, 1966.
31. Szepesi, B., Vegors, R., and DeMosey, J, M., On the possible role of insulin in the
starve-refeed response, Nutr. Rep. Int., 5, 281, 1972.
32. Berdanier, C. D., Wurdeman, R., and Tobin, R. B., Further studies on the role of
the adrenal hormones in the responses ofrats to meal feeding, J. Nutr., 106, 1791, 1976.
33. Wurdeman, R., Berdanier, C. D., and Tobin, R. B., Enzyme overshoot in starved-
refed rats: role of adrenal glucocorticoid, J. Nutr., 108, 1457, 1978.
34. Bouillon, D. J, and Berdanier, C. D., Role of glucocorticoid in adaptive hyperlipo-
genesis in the rat, J. Nutr., 110, 286, 1980.
35. Berdanier, C. D. and Shubeck, D., Interaction of glucocorticoid and insulin in the
responses of rats to starvation-refeeding, J. Nutr., 109, 1766, 1979.
36. Nessmith, S., Baltzell, J., and Berdanier, C. D., Interaction of glucocorticoid and
thyroxine in the responses of rats to starvation-refeeding, J. Nutr., 113, 2260, 1983.
37. Kletzien, R. F., Pasiza, M. W., Becker, J, E., Potter, V. R., and Butcher, F. R.,
Induction of amino acid transport in primary cultures of adult rat liver parenchymal cells
by insulin, J. Bioi. Chem., 251, 3014, 1976.
38. Inoue, C., Yamamoto, H., Nakamura, T., Ichihara, A., and Okamoto, H., Nico-
tinamide prolongs survival of primary cultured hepatocytes without involving loss of
hepatocyte-specific functions, J. Bioi. Chem., 264, 4747, 1989.
39. Dunn, J, C. Y., Yarmush, M. L., Koebe, H. G., and Tompkins, R. G., Hepatocyte
function and extracellar matrix geometry: long-term culture in a sandwich configuration,
Fed. Proc., 3, 174, 1989.
40. Kelley, D. S. and Kletzien, R. F., Ethanol modulation of the hormonal and nutritional
regulation of glucose-6-phosphate dehydrogenase activity in primary cultures of rat he-
patocytes, Biochem. J., 217, 543, 1984.
41. Nakamura, T., Yoshimoto, K., Aoyama, K., and Ichihara, A., Hormonal regulations
of glucose-6-phosphate dehydrogenase and lipogenesis in primary cultures of rat hepa-
tocytes, J. Biochem., 91, 681, 1982.
42. Manos, P., Nakayama, R., and Holten, D., Regulation of glucose-6-phosphate de-
hydrogenase synthesis and mRNA abundance in cultured rat hepatocytes, Biochem. J.,
276, 245, 1991.
43. Salati, L. M., Adkins-Finke, B., and Clarke, S. D., Free fatty acid inhibition of the
insulin induction of glucose-6-phosphate dehydrogenase in rat hepatocyte monolayers,
Lipids, 23, 36, 1988.
44. Kelley, D. S., Nelson, G. J., and Hunt, J. E., Effect of prior nutritional status on the
activity of lipogenic enzymes in primary monolayer cultures of rat hepatocytes, Biochem.
J., 235, 87, 1986.
Kletzien and Berdanier 205
TABLE OF CONTENTS
0-8493-6961-41931$0.00 +$.50
© 1993 by CRC Pre>s. Inc. 207
208 Nutrition and Gene Expression
I. INTRODUCTION
FIGURE 1. Relative levels of liver (B-type) phosphofructokinase, fatty acid synthase, and
p90 rnRNAs during fasting and refeeding of a high carbohydrate, fat-free diet. Poly (A+) RNA
{4 fLg per lane) prepared from livers of mice starved 48 h or previously starved mice refed for
16 h were subjected to 0. 7% agarose gel electrophoresis, transferred to nitrocellulose, and probed
with 32 P-labeled eDNA inserts.
cipitation method was employed for enrichment of its message before prep-
aration of a eDNA library in A. gtll and screening with liver phosphofruc-
tokinase isozyme-specific antibodies. 15
Northern blot analysis using these eDNA sequences showed that the fatty
acid synthase and p90 mRNAs were present in high concentration in adipose
tissue and liver. 11 ·14 Liver phosphofructokinase mRNA, on the other hand
was detected in all tissues examined although liver showed the highest con-
centration.15 Fatty acid synthase and p90 mRNAs were not detectable in fasted
mouse liver and increased by two orders of magnitude when mice were refed
a high carbohydrate, fat-free diet. 11 In contrast, the liver phosphofructokinase
mRNA was easily detectable in fasted mice liver and increased fivefold during
refeeding (Figure l ). Dunaway and co-workers previously reported a decrease
Sul et al. 211
70
~
w 10
~~
< 50
z
a:
E ~
~
w
en JO
<
w
u
a: 20
~
0 10
~
~
0
0 4
• 10 12 14 11
TIME !hl
- 8 .2 kb
- 6.8 kb
. ..
F RF F RF RF + Bt ~ cAMP
DiabetiC Normal
FIGURE 2. Effects of streptozotocin-induced diabetes and cAMP on fatty acid synthase and
p90 mRNA induction by fasting/refeeding. Poly (A + ) RNA was prepared from livers of diabetic
and normal mice fasted (F) or refed (RF) for 8 h. Dibutyryl cAMP and theophyll ine were injected
at the time of refeeding in normal, fasted mice. Northern analyses were carried out as in Figure
I. 2.1 kb mRNA represents a-actin .
Sul et al. 213
6.8 kb-
8 .2 kb -
0 2 4 6 8 8C
TIME (h )
20
1-
z
w
1-
5(.)
15
<(
z
c:
E 10
~
w
C)
~
I
(.)
a
c5u.
TIME(h)
FIGURE 3. Effect of insulin on mRNA levels for p90 and fatty acid synthase in streptozotocin-
diabetic mice. Poly (A+) RNA was prepared from livers of streptozotocin-diabetic mice killed
at the indicated times after administration of combined dose of regular insulin (3 U/100 g)
intraperitoneally and Lente Insulin (30 U/100 g) subcutaneously. Saline-injected mice were also
used at 8 h as a control (8C). Northern analyses were done as in Figures I and 2.
214 Nutrition and Gene Expression
for fatty acid synthase and p90 increased four- and twofold 1 h after intra-
peritoneal insulin injection, respectively, and achieved maximal increases of
about 20-fold after 6 h. However, it is known that glucagon levels are de-
creased when insulin is administered to diabetic animals and this may be a
factor. In cultured avian embryo hepatocytes, Goodridge and co-workers
reported that insulin only amplified the effect of T3 on fatty acid synthase
gene expression. 19 ·20 It would be surprising if there is a difference in regulation
of avian and murine fatty acid synthase by insulin.
NORMAL DIABETIC
--
pBR322
p13
pFAS-1
pAM 91
.. --
0 6 69 16 0 6
Bt 2 cAMP
Q)
n;
a:
c: 30
·a0
"5
U)
c:
~ 20
I-
.E
Ql
Ol
~ to
.r.
(.)
"0
&
5 tO 15
pBR322 • •
p13
pFAS-1
pAM91 •
0 0.5 1.0 2.0 6.0
TIME (h)
Q)
~
a:
c:
0
~
a.
·;;::
(.)
(/)
c:
~
1-
c:
Q)
0>
c:
cu
.s::
()
"0
0
lL
Time (h)
FIGURE 5. Effect of insulin on transcriptional rates of p90 and fatty acid synthase genes in
streptozotocin-diabetic mice liver. Nuclei were isolated from diabetic mice treated with insulin
as in Figure 3 and run-on assays were carried out. (•) pl3; (eJ) pFAS-1; (+l pAM91.
V. DIFFERENTIATION-DEPENDENT EXPRESSION
IN 3T3-Ll CELLS
pBR322
FAS pBiuescript
mANA
pFAS1
pSCD1
dO d5 dO d5
FIGURE 6. Comparisons of mRNA levels and transcription rates for fatty acid synthase gene
in 3T3-Ll preadipocytes and adipocytes. RNA (5 JLg per lane) prepared from confluent 3T3-Ll
cells (dO) and from cells differentiated by treatment of methylisobutylxanthine and dexamethasone
(d5) were used for Northern blot. Nuclei isolated from preadipocytes and adipocytes were used
for run-on transcription and hybridized with 10 JLg each of fatty acid synthase (pFAS-1), stearoyl
CoA desaturase (pSCDl), and control plasmids fixed on nitrocellulose.
preadipocytes adipocytes
, 234 4c 12344 c
Cl)
>
.!!
<
z
a:
E
0 10 20
time (hour)
200
...c
0
-
(_)
0
# 100
FIGURE 8. Effect of insulin and cAMP on mRNA levels for fatty acid synthase p90 in 3T3-
Ll adipocytes. 3T3-Ll cells were differentiated, maintained in serum-free medium for 24 h,
and then treated either with I J.Lg/ml of insulin or 1 mM dibutyryl cAMP plus 1 mM theophylline
for 16 h. Poly (A+) RNA prepared were used in Northern blot analyses for fatty acid synthase
(D) and p90 (~).
(A)
FAS mANA-
0 6 12 24
Time (Hours)
(B)
p13
pFAS1
pBR322
0 6 12 24
Time (Hours)
FIGURE 9. Effect of T3 on mRNA level and transcription rate for fatty acid synthase. Dif-
ferentiated 3T3-Ll adipocytes were treated with T3 for indicated times. RNA and nuclei were
prepared and used for Northern blot (A) and transcription run-on assays (B), respectively.
VII. CONCLUSIONS
(a) (b)
0
2
15 30
3 2 3
0 15 30 45 0 0 15 30
0 15 30 45 0 0 15 30 45 0 0 15 30 45 0
- 530
r -10
A r
l
E - 510
L
-3 0
- 490
r
8
- - 50
l- -7 0 0
r
I L
c
L ...• •450
- 90
FIGURE 10. Binding of nuclear proteins to the mouse liver phosphofructokinase promoter
region. DNA fragments containing the mouse liver phosphofructokinase promoter regions. + 87
to -169 (a) and -351 to -699 (b), were labeled, incubated with indicated amounts (j.Lg) of
liver nuclear extracts, and digested with DNase I. The regions of protection are demarcated: (a)
I, no competing oligonucleotides; 2, 60-fold excess of oligonucleotides (- 65 to - 97); 3, 60-
fold excess of AP-I consensus sequence and (b) I, no competing oligonucleotides; 2, excess of
oligonucleotides (-445 to -475); 3, excess of oligonucleotides (-485 to -522).
ACKNOWLEDGMENTS
REFERENCES
I. Gibson, D. M., Lyons, R. T., Scott, D. F., and Muto, Y., Synthesis and degradation
of the lipogenic enzymes of rat liver, Adv. Enzyme Regul., 10, 187, 1972.
2. Volpe, J. J. and Vagelos, P. R., Mechanisms and regulation of biosynthesis of saturated
fatty acids, Physiol. Rev., 56, 339, 1976.
3. Lakshmanan, M. R., Nepokroeff, C. M., and Porter, J, W., Control of the synthesis
of fatty acid synthetase in rat liver by insulin, glucagon, and adenosine 3':5'cyclic
monophosphate, Proc. Nat!. Acad. Sci. U.S.A., 69, 3516, 1972.
4. Goodridge, A. G., Dietary regulation of gene expression: Enzymes involved in carbo-
hydrate and lipid metabolism, Annu. Rev. Nutr., 7, 157, 1987.
5. Brent, G. A., Moore, D. D., and Larsen, P. R., Thyroid hormone regulation of gene
expression, Annu. Rev. Physiol., 53, 17, 1991.
6. Gonzalez, G. A., Yamamoto, K. K., Fischer, W. H., Karr, D., Menzel, P., Biggs,
W., Vale, W. W., and Montiminy, M. R., A cluster of phosphorylation sites on the
cAMP-regulated nuclear factor CREB predicted by its sequence, Nature (London), 337,
749, 1989.
7. Bates, E. J, and Saggerson, E. D., A study of the glycerol phosphate acyltransferase
and dihydroxyacetone phosphate acyltransferase activities in rat liver mitochondrial and
microsomal fractions, Biochem. J., 182, 751, 1979.
8. Saggerson, E. D. and Carpenter, C. A., Effects of streptozotocin-diabetes and insulin
administration in vivo or in vitro on the activities of five enzymes in the adipose-tissue
triacylglycerol-synthesis pathway, Biochem. J., 243, 289, 1987.
9. Dunaway, G. A. and Weber, G., Effects of hormonal and nutritional changes on rates
of synthesis and degradation of hepatic phosphofructokinase isozymes, Arch. Biochem.
Biophys., 162, 629, 1974.
10. Dunaway, G. A., Leung, G. L.-Y., Thrasher, J, R., and Cooper, M.D., Turnover
of hepatic phosphofructokinase in normal and diabetic rats. Role of insulin and peptide
stabilizing factor, J. Bioi. Chern., 253, 7460, 1978.
I I. Paulauskis, J. D. and Sui, H. S., Cloning and expression of mouse fatty acid synthase
and other specific mRNAs. Development and hormonal regulation in 3T3-LI cells, J.
Bioi. Chern .. 263, 7049, 1988.
12. Yan, C., Wood, E. A., and Porter, J, W., Characterization of fatty acid synthetase
eDNA clone and its mRNA, Biochem. Biophys. Res. Commun., 126, 1235, 1985.
13. Back, D. W., Goldman, M. J,, Fisch, J. E., Ochs, R. S., and Goodridge, A. G.,
The fatty acid synthase gene in avian liver. Two mRNAs are expressed and regulated in
parallel by feeding, primarily at the level of transcription, J. Bioi. Chern., 261, 15179,
1986.
14. Shin, D. H., Paulauskis, J, D., Moustaid, N., and Sui, H. S., Transcriptional regulation
of p90 with sequence homology to E. coli glycerol-3-phosphate acyltransferase, J. Bioi.
Chern., 266, 23 834, 1991.
15. Gehnrich, S. C., Gekakis, N., and Sui, H. S., Liver (B-type) phosphofructokinase
mRNA. Cloning, structure, and expression, J. Bioi. Chern., 263, 11755, 1988.
16. Raymondjean, M., Vaulont, S., Cognet, M., Decaux, J. F., Puzenat, N., Bergot,
M. 0., and Kahn, A., Positive and negative regulation of gene expression by insulin
and glucagon: the model of L-type pyruvate kinase gene, Biochimie, 73, 41, 1991.
17. Sui, H. S., Wise, L. S., Brown, M. L., and Rubin, C. S., Cloning of eDNA sequences
for murine ATP-citrate lyase. Construction of recombinant plasmids using an immuno-
purified mRNA template and evidence for the nutritional regulation of ATP-citrate lyase
mRNA content in mouse liver, J. Bioi. Chern., 259, 1201, 1984.
18. Sui, H. S., Wise, L. S., Brown, M. L., and Rubin, C. S., Cloning of eDNA sequences
for murine malic enzyme and the identification of aberrantly large malic enzyme mRNA
in MOD-I null mice, J. Bioi. Chern., 259, 555, 1984.
Sul et al. 225
19. Wilson, S. B., Back, D. W., Morris, S.M., Jr., Swierczynski, J., and Goodridge,
A. G., Hormonal regulation of lipogenic enzymes in chick embryo hepatocytes in culture.
Expression of the fatty acid synthase gene is regulated at both translational and pretran-
slational steps, J. Bioi. Chern., 261, 15179, 1986.
20. Stapleton, S. R., Mitchell, D. A., Salati, L. M., and Goodridge, A. G., Triiodothy-
ronine stimulates transcription of the fatty acid synthase gene in chick embryo hepatocytes
in culture. Insulin and insulin-like growth factor amplify that effect, J. Bioi. Chern., 265,
18442, 1990.
21. Paulauskis, J. D. and Sui, H. S., Hormonal regulation of mouse fatty acid synthase
gene transcription in liver, J. Bioi. Chern., 264, 574, 1989.
22. Green, H. and Kehinde, 0., Sublines of mouse 3T3 cells that accumulate lipid, Cell,
I, 113, 1974.
23. Rubin, C. S., Hirsch, A., Fung, C., and Rosen, 0. M., Development of hormone
receptors and hormonal responsiveness in vitro, J. Bioi. Chern., 253, 7570, 1978.
24. Sui, H. S., Adipocyte differentiation and gene expression, Curr. Opinion Cell Bioi., 1,
1116, 1989.
25. Weiss, G. H., Rosen, 0. M., and Rubin, C. S., Regulation of fatty acid synthetase
concentration and activity during adipocyte differentiation. Studies on 3T3-LJ cells, J.
Bioi. Chern., 255, 4751, 1980.
26. Student, A. K., Hsu, R. Y., and Lane, M. D., Induction of fatty acid synthetase
synthesis in differentiating 3T3-LJ preadipocytes, J. Bioi. Chern., 255, 4745, 1980.
27. Wise, L. S., Sui, H. S., and Rubin, C. S., Coordinate regulation of the biosynthesis
of ATP-citrate lyase and malic enzyme during adipocyte differentiation. Studies on 3T3-
Ll cells, J. Bioi. Chern., 259, 4827, 1984.
28. Moustaid, N. and Sui, H. S., Regulation of expression of the fatty acid synthase gene
in 3T3-Ll cells by differentiation and triiodothyronine, J. Bioi. Chern., 266, 18550,
1991.
29. Bernlohr, D. A., BoJanowski, M. A., Kelly, T. J., and Lane, M. D., Evidence for
an increase in transcription of specific mRNAs during differentiation of 3T3-Ll prea-
dipocytes, J. Bioi. Chern., 260, 5563, 1985.
30. Cook, D. S., Hunt, C. R., and Spiegelman, B. M., Developmentally regulated mRNAs
in 3T3-adipocytes: analysis of transcriptional control, J. Cell. Bioi., 100, 514, 1985.
31. Djian, P., Phillips, M., and Green, H., The activation of specific gene transcription
in the adipose conversion of 3T3 cells, J. Cell. Physioi., 124, 554, 1985.
32. Mackall, J. C. and Lane, M.D., Role of pyruvate carboxylase in fatty acid synthesis:
alterations during preadipocyte differentiation, Biochern. Biophys. Res. Cornrnun., 79,
720, 1977.
33. Spiegelman, B. M. and Green, H., Control of specific protein biosynthesis during the
adipose conversion of 3T3 cells, J. Bioi. Chern., 255, 8811, 1980.
34. Gekakis, N., Gehnrich, S.C., and Sui, H. S., Phosphofructokinase isozyme expression
during myoblast differentiation, J. Bioi. Chern., 264, 3658, 1989.
35. Volpe, J. J. and Kishimoto, Y., Fatty acid synthetase of brain: development, influence
of nutritional and hormonal factors and comparison with liver enzyme, J. Neurochern.,
19, 737, 1972.
36. Mariash, C. N., Kaiser, F. E., and Oppenheimer, J. H., Comparison of the response
characteristics of four lipogenic enzymes to 3,5,3' -triiodothyronine administration: evi-
dence for variable degrees of amplification of the nuclear 3,5,3' -triiodothyronine signal,
Endocrinology, 106, 22, 1980.
37. Diamant, S., Gorin, E., and Shafrir, E., Enzyme activities related to fatty acid synthesis
in liver and adipose tissue of rats treated with triiodothyronine, Eur. J. Biochern., 26,
553, 1972.
38. Mariash, C. N., Kaiser, F. E., Schwartz, H. L., and Towle, H. C., Synergism of
thyroid hormone and high carbohydrate diet in the induction of lipogenic enzymes in the
rat. Mechanisms and implications, J. Clin. Invest., 65, 1126, 1980.
226 Nutrition and Gene Expression
39. Spence, J, T. and Pilot, H. C., Induction of lipogenic enzymes in primary cultures of
rat hepatocytes. Relationship between lipogenesis and carbohydrate metabolism, Eur. J.
Biochem., 36, 15, 1982.
40. Rongnoparut, P., Verdon, C. P., Gehnrich, S. C., and Sui, H. S., Isolation and
characterization of the transcriptionally regulated mouse liver (B-type) phosphofructo-
kinase gene and its promoter, J. Bioi. Chern., 266, 8086, 1991.
41. O'Brien, R. M., Lucas, P. C., Forest, C. D., Magnuson, M. A., and Granner,
D. K., Identification of a sequence in the PEPCK gene that mediates a negative effect
of insulin on transcription, Science, 249, 533, 1990.
42. Nasrin, N., Ercolani, L., Denaro, M., Kong, X. F., Kang, I., and Alexander, M.,
An insulin response element in the glyceraldehyde-3-phosphate dehydrogenase gene binds
a nuclear protein induced by insulin in cultured cells and by nutritional manipulations in
vivo, Proc. Nat/. Acad. Sci. U.S.A., 87, 5273, 1990.
Chapter 10
TABLE OF CONTENTS
0-8493-6961-4/93/$0.00 +$.50
© 1993 by CRC Press, Inc. 227
228 Nutrition and Gene Expression
I. INTRODUCTION
The classic studies of Chaikoff and his colleagues 14 had indicated that fat
ingestion would suppress acetate incorporation into hepatic lipids. It was
C Iarke and Jump 229
TABLE 1
Dietary Fatty Acid Effects on Rat Liver Proteins
proposed that fatty acids or their CoA esters functioned as negative allosteric
modifiers of key glycolytic and lipogenic enzymes, e.g., phosphofructokinase
and acetyl-CoA carboxylase. 14 • 15 In our early studies, the putative fatty acid
allosteric binding sites of hepatic phosphofructokinase and/or acetyl-CoA
carboxylase were hypothesized to possess a higher affinity for polyunsaturated
fatty acyl-CoAs than for the saturated fatty acid CoA esters. Thus, polyun-
saturated fatty acids could selectively inhibit glucose flux to fatty acids by
selectively binding to regulatory enzymes and inhibiting their catalytic effi-
230 Nutrition and Gene Expression
125~-----------------------------------------,
100
(])
(/)
c:
0
0. 75
(/)
(])
a:
(])
>
·.;:::; 50
<tl
(])
a:
25
FIGURE 1. Structural requirements for a fatty acid to suppress hepatic lipogenesis. Mice
(n = 4) were fed for 5 d a high glucose diet containing 5% (w/w) of the respective fatty acids
cited in the figure. In vivo fatty acid synthesis was determined by tritiated water incorporation
into fatty acids. Relative response is defined relative to mice fed a high glucose fat-free diet
(SEM < 15%).
ciency. 16 ·17 Recently, we reported that polyunsaturated fats were in fact better
acute inhibitors of hepatic fatty acid synthesis and malonyl-CoA accumulation
than were saturated fats. 16 However, the acute suppression of fatty acid syn-
thesis by the ingestion of a single meal containing polyunsaturated fat was
modest when compared to the 60 to 90% reduction in fatty acid synthesis
and lipogenic enzymatic activities brought on by the prolonged (3 to 4 d)
ingestion of polyunsaturated fat (Figure 4). 1-12 The acute suppression of lipo-
genesis by safflower oil was not the consequence of reduced catalytic effi-
ciency or capacity of pyruvate kinase or phosphofructokinase, i.e. glycolytic
activity was unaffected. 5 In addition, the quantity of catalytically functional
acetyl-CoA carboxylase (as determined by increased resistance to avidin ac-
tivation)18·19 was not reduced by the ingestion of saturated or polyunsaturated
fats (Figure 5). Subsequent analysis of the time course of inhibition of fatty
acid synthesis vs. acetyl-CoA carboxylase and fatty acid synthase activities
revealed that the polyunsaturated fatty acid suppression of hepatic fatty acid
biosynthesis paralleled the decline in the amount of acetyl-CoA carboxylase
and fatty acid synthase. 5 Thus, polyunsaturated fatty acids appeared to exert
their greatest impact on the fatty acid biosynthetic capacity via a coordinate
Clarke and Jump 231
Inhibition of enzyme protein synthesis is the major reason for the reduction
in hepatic activity of lipogenic enzymes (e.g., fatty acid synthase) caused by
the ingestion of polyunsaturated fatty acids. 6 •20 Two mechanisms may explain
this inhibition of enzyme synthesis: (1) interference with translation rate of
the mRNA and/or (2) reduction in the amount of mRNA. To address this
issue we have focused our efforts on two genes: fatty acid synthase and the
putative lipogenic protein, S 14. 10- 12
When young growing rats were fed for 6 d a diet containing 10% safflower
oil, the abundance of fatty acid synthase and S14 mRNA was decreased to
levels that were 30 and 50%, respectively, of the values observed with a fat-
free high glucose diet. 11 Comparable amounts of dietary monounsaturated or
saturated fat (e.g., triolein) had no suppressive effect. 11 Polyunsaturated fatty
acids not only impaired fatty acid synthase and Sl4 expression in the growing
and adult rodents, but also blocked the 20- to 30-fold rise in mRNA levels
associated with weaning rats onto a high carbohydrate diet. 11 The weanling
animal appeared to be more sensitive to the polyunsaturated fat inhibition
than was the growing or adult rat, e.g., 3% 18:2 (n-6) suppressed fatty acid
synthase expression 80 to 90% in the 30-d-old rat, whereas 10% 18:2 (n-6)
suppressed fatty acids synthase mRNA only 65% in the 3-month-old rat. 11
The regulation of gene expression by dietary polyunsaturated fats is not unique
to fatty acid synthase and Sl4, but also extends to the suppression of acetyl
CoA carboxylase 21 and glucose-6-phosphate dehydrogenase. 22 Thus, dietary
polyunsaturated fats appear to coordinately suppress the level of transcripts
coding for lipogenic enzymes, and in this way decrease the synthesis of these
enzymes which in tum results in a reduced lipogenic capacity. 4 •5 • 11 • 12 However,
each gene possesses its own regulatory sequences, and expression of each
gene coding for a protein in the fatty acid biosynthetic pathway is a summation
of integrated hormonal and nutrient signals. Consequently, individual proteins
may vary in the degree of response to dietary polyunsaturated fatty acids,
e.g., S14 mRNA is less dramatically reduced by polyunsaturated fats than is
fatty acid synthase mRNA. 11 Nevertheless, polyunsaturated fatty acids clearly
possess a selective ability to reduce the amount of protein and mRNA for
several enzymes of fatty acid biosynthesis.
While dietary fat rich in 18:2 (n-6) suppressed the expression of enzymes
involved in fatty acid biosynthesis, the level of mRNA and protein for the
liver fatty acid binding protein (1-FABP) is induced by dietary fatsY A
232 Nutrition and Gene Expression
-- 150.--------------------------------------------,
+-'
Q)
0
+-'
If
0
z 100
'*'
Cl)
Cl)
Q)
~
+-'
c
>
Cl) 50
:'2
()
<0
>
+-'
+-'
If
Liver Gut Lung Brain Adipose
Tissue
selective induction of 1-FABP due to polyunsaturated fats has not been firmly
established, but studies with rat liver cells in primary culture indicate 18:2
(n-6) directly enhanced 1-FABP synthesis, and resulted in higher levels of 1-
FABP protein than did 16:0. 24 Clearly, polyunsaturated fatty acids uniquely
regulate hepatic gene expression, and this effect may extend to genes coding
for proteins other than enzymes in fatty acid biosynthesis.
The polyunsaturated fatty acid control of lipogenic proteins is predomi-
nately a liver-specific phenomenon (Figures 2 and 3). 25 Fatty acid biosynthesis
and the activities of lipogenic enzymes within rodent lung, brain, and small
intestine were unaffected by a diet that contained 20 to 30% kcal as hydro-
genated cottonseed oil, com oil, and menhaden oil (Figure 2). 9 ·25 Comparable
levels of dietary fat reduced de novo fatty acid biosynthesis in mammary and
epididymal fat pads, but saturated and polyunsaturated fats were equipotent
(Figure 2). Interestingly, fatty acid synthase mRNA levels were unaffected
by either dietary saturated or polyunsaturated fat (Figure 3), 9 which is in sharp
contrast to the hepatic responses to these dietary lipids. 11 •23 Since dietary fats
suppress adipose fatty acid synthase activity and rates of fatty acid biosyn-
thesis, 15 the data suggest fatty acid synthase synthesis in adipose tissue may
be regulated at the level of translation as well as at the point of transcription.
Clarke and Jump 233
-Cl)
150
-
0
CD
u..
0
z
*'
<(
100
z
a:
E
Cl)
-
fll
CD
..r:
c: 60
>
Vl
"'0
"(j
<(
-
>
CD
u.. 0
Liver Mam Fat Epid Fat
Tissue
120
-.....
Q)
i:5
100 ---- + - -~--t--
.....
'\,""'---J
'
co
LL. 80
z
0
~~~
-
'#.. 60 r -~~"
~
>
.-::
>
'_;j 40
--------
"i----
()
<(
en
20
·t-~---~-
~ ~--
0
0 3 6 10 15
Diet Fat Dose (% w/w)
FIGURE 4. Response of hepatic fatty acid synthase activity to varying doses and types of
dietary fat. Mice (n = 4) were fed a high glucose diet for 5 d containing the level of fat (w/w)
cited in the figure. Hydrogenated cottonseed oil, com oil, and menhaden fish oil are represented
by the open squares, open triangles, and crosses, respectively. Data are expressed as means ±
SEM for n = 4 mice per point.
quirement for fish oil to achieve 50% of maximum inhibition (Figure 4). 9 A
comparison of 18:3 (n-6) and 20:4 (n-6) with fish oil has shown that the key
difference in potency between fish oil and vegetable oil lies in the conversion
rate of 18:2 (n-6) to 18:3 (n-6), i.e., 20:4 (n-6) is equipotent to fish oil (Figure
6). Thus, the greater potency of fish oil as an inhibitor of fatty acid synthase
and S 14 gene expression is due primarily to its high proportion (> 30%) of
20- and 22-carbon polyenoic fatty acids.
V. HEPATIC-SPECIFIC INHIBITION
co 80
en
.5!
>
X
0 ~--:::-
..0
..... / :;..e-
co 60 /
uI b/
<(
0
u()
<( /
....c: 40
/
/
....enco /
I"
/> /
~
"(j)
co
a: 20
c:
"U
<(
>
"#. 0
0 10 20 30 120
Minutes into Meal
FIGURE 5. The effect of dietary saturated and polyunsaturated fats on the activation of acetyl
CoA carboxylase during meal ingestion. Rats were trained to eat their entire daily food allotment
during a 3-h meal. The fraction of catalytically active acetyl CoA carboxylase present in hepatic
tissue during the ingestion of a single meal of 10% tripalmitin (solid circles), 10% safflower oil
(open squares), or no fat (solid squares) was determined using the avidin inactivation proce-
dure.'•·'9
enoic (n-6) fatty acids to the media (Figure 7). 26 •27 As occurs in vivo, 20:4
(n-6) was more potent than 18:2 (n-6). Approximately 100 IJ.M 20:4 (n-6)
was required to suppress fatty acid synthase mRNA induction 50%. 26 Ad-
mittedly, circulating levels of 20:4 (n-6) above 50 ~J.M are unlikely. However,
hepatocytes in culture rapidly utilize 20:4 (n-6) for triglyceride and phospho-
lipid synthesis. 28 Thus, the media concentration of 20:4 (n-6) must be main-
tained at a high level in order to provide an effective concentration sufficient
to exert an inhibitory action. As would be expected from the in vivo responses,
as much as 500 1J.M 20: 1 (n-9) did not inhibit fatty acid synthase expression,
and in fact, increased synthase mRNA levels twofold (Figure 7). 26 One pos-
sible explanation for the enhancement of fatty acid synthase gene expression
by 20:1 (n-9) may be that 20: 1 (n-9) displaced the inhibitor 20:4 (n-6).
Interestingly, as the media content of 20:4 (n-6) was increased from 50
to 500 IJ.M, the level of 13-actin mRNA rose 70 to 275%, but 20:1 (n-9) had
no effect on 13-actin expression (Figure 7). 26 13-Actin expression is used as
an indicator of hepatocyte differentiation status, i.e., high levels of 13-actin
mRNA may be indicative of a loss of adult differentiation traits. 29 It is curious
236 Nutrition and Gene Expression
-CD
0
c
100
as
"C
c 75
::l
.c
as
,...
-
co
,...
50
0
-z
0~
<(
25
a:
E
CIJ
~ 0
FIGURE 6. Delta-6 desaturation of linoleate and o:-linolenate is required to suppress fatty acid
synthase gene expression. Mice were fed a high glucose diet for 5 days containing various fatty
acids with (solid bars) and without (slashed bars) the delta-6 desaturase inhibitor eicosatetraynoic
acid (ETYA). The level of fat supplementation to the diet was 5% for 18:2, 18:3, and 18:1, and
3% for 20:3 and 20:4. The 20:5/22:6 column represents 10% dietary menhaden fish oil which
provides 3% 20:5/22:6 (n-3).
501!M
0 20:1 20:4 20:1 20:4 20:1 20:4
FAS ·
l'l · Actin·
FIGURE 7. Northern analysis demonstrating suppression of fatty acid synthase gene expression
in primary cultures of rat liver cells by albumin-bound arachidonate. Rat liver cells were main-
tained in primary culture 38 and incubated for 36 h with the levels of albumin-bound 20:4 (n-6)
or 20: I (n-9) cited in the figure.
(9c, 12c, 15c-18:3) both reduce the abundance of fatty acid synthase mRNA
and the rate of lipogenesis. 11 •20 With respect to double bond conformation,
one of the two double bonds may be in the trans configuration, but 9t,12t-
18:2 does not lower the level of hepatic fatty acid synthase rnRNA (Figure
7).7
While dietary 18:2 (n-6) and 18:3 (n-3) markedly reduce the abundance
of hepatic fatty acid synthase and S14 mRNAs, 11 the actual intracellular
modulator of gene expression is not 18:2 (n-6) or 18:3 (n-3) per se. 9 In order
for these fatty acids to suppress the expression of genes coding for lipogenic
enzymes, they must first undergo desaturation by the delta-6 desaturase. When
delta-6 desaturation was blocked by the fatty acid analog, eicosatetraynoic
acid (ETYA), the suppression of fatty acid synthase mRNA level by dietary
safflower oil, com oil, or 18:2 (n-6) was completely prevented (Figure 6),
but the hepatic concentration of 18:2 (n-6) dramatically increased. 8 •9 Adding
the product of the delta-6 desaturase (e.g., 6c,9c,12c-18.3) completely rein-
stated the inhibition of gene expression (Figure 6). Under these conditions,
18:3 (n-6) and 20:3 (n-6) content of hepatic lipids increased, but conversion
of 20:3 (n-6) to 20:4 (n-6) continued to be impaired because ETYA also
inhibited the delta-5 desaturase. Thus, while 20:4 (n-6) and 20:5 (n-3) are
potent suppressors of fatty acid synthase gene expression, the formation of
these fatty acids is not an a priori requirement for the polyenoic fatty acid
regulation of gene expression to occur.
238 Nutrition and Gene Expression
250,------------------------------------------ ---.
Q)
c
0
E
0
2oo
I
-
0
z
150
0
'#.
c
0 100
CJ)
CJ)
CD
.....
a.
X
w 50
~
u
Hormonal Treatment
FIGURE 8. Stimulation of fatty acid synthase promoter activity by glucocorticoids. Rat liver
cells in primary culture 38 were transiently transfected by lipofectin with a pSV2 vector containing
- 2190 to + 65 of the fatty acid synthase gene, the chloramphenicol acetyltransferase reporter,
and the SV40 enhancer 5' to the promoter. The hormone concentrations were 0.1 f.LM and SEM
< 15%.
fatty acid synthase gene and its promoter, coupled with the extensive char-
acterization of the S14 gene, will permit us to identify potential cis-elements
and trans-acting nuclear proteins which may be under polyunsaturated fatty
acid control.
1 2 3 4 5 6 7 8
FAS-
28S-
tJ- Actin-
FIGURE 9. Hormonal induction of fatty acid synthase gene expression in primary cultures of
rat liver cells. Rat liver cells were maintained in primary culture" and treated with the hormones
cited in the figure. RNA was extracted and subjected to Northern analyses. The hormonal
concentrations were 0. I f.LM. Lane I, no hormone; Jane 2, insulin; Jane 3, insulin/dexamethasone;
lane 4, dexamethasone; lane 5, insulinldexamethasoneff3 ; Jane 6, T 3 ; Jane 7, T 3/insulin; and lane
8, T,!dexamethasone.
sion of fatty acid synthase and S 14 transcription. There is little question that
dietary polyunsaturated fats alter hormone binding and membrane signaling
mechanisms, 39 A 2 but the hormonal responsivity changes induced by polyun-
saturated fatty acids do not present a pattern consistent with suppression of
fatty acid synthase and S 14 gene expression. For example, although dietary
polyunsaturated fats increase glucagon-stimulated adenylate cylase activity
twofold, 39 they also significantly increase tissue sensitivity to insulin. 40 .4 2
Moreover, the dose of com oil required to elicit maximal increases in glu-
cagon sensitivity was only 2% of the diet, 39 while inhibition of fatty acid
synthase gene expression by com or safflower oil continues to be expressed
at doses up to 10% of the diet (Figure 4). 11 •37 The strongest argument against
the idea that fatty acid membrane compositional changes are responsible for
the control of gene expression lies in the kinetics of transcriptional inhibition
vs. membrane fatty acid changes. Removing fish oil from the diet for simply
one 3-h meal completely reverses the near 90% suppression of fatty acid
synthase gene expression. 11 Similarly, transcription of acetyl CoA carbox-
ylase was suppressed 50% within 6 h of adding com oil to the diet. 21 In our
opinion, 3 to 6 h is insufficient time for a dietary lipid to achieve significant
changes in membrane phospholipid fatty acid composition. Rather, the ki-
netics of transcriptional regulation are more consistent with direct control of
gene transcription by polyunsaturated fatty acids or their metabolites.
The final piece of evidence against a change in cAMP balance caused
by dietary polyunsaturated fats is derived from the effects of these fatty acids
on gene transcription. 12 If the polyunsaturated fatty acid suppression of fatty
acid synthase and Sl4 gene transcription were to involve a cAMP-dependent
process, then one would expect that the transcription rate of phosphoenol-
pyruvate carboxykinase (PEPCK) would be increased by dietary polyunsa-
turated fats. However, PEPCK transcription, which is exquisitely sensitive
to stimulation by cAMP and inhibition by insulin, 43 was not affected by dietary
polyunsaturated fat. 12 Thus, collectively the available data suggest that the
regulation of gene expression by polyunsaturated fats does not involve shifts
in the balance of hormones which modulate the cAMP second message system.
While enrichment of membrane lipids with polyunsaturated fats appears
not to be the mechanism by which polyunsaturated fatty acids regulate fatty
acid synthase and S 14 gene expression, it is feasible that long chain polyenoic
fatty acids interfere with nuclear events regulated by lipogenic hormones. For
example, fatty acids or nuclear fatty acid binding proteins might potentially
interact with T3 or glucocorticoid receptors in such a way as to interfere with
receptor/DNA interactions and thereby alter transcriptional events. While such
an idea is purely speculation, dietary polyunsaturated fats were found to
interfere with T3 induction of hepatic fatty acid synthase, malic enzyme, and
glucose-6-phosphate dehydrogenase. 44 The effect of a diet containing 10%
safflower oil was to shift the dose-response curve for T3 to the right, 44 but
not to reduce the maximum activity induced by high doses ofT 3 • Such a shift
Clarke and Jump 243
ACKNOWLEDGMENTS
The work presented within this review was supported in part by the Upjohn
Company, Kalamazoo, MI and the National Institutes of Health-DK 39302
(SOC) and GM 36851 (DBJ). The authors wish to thank Ormond MacDougal,
William Blake, Meera Kamdar, and Ana Mildner for the technical assistance
and thoughtful discussions. Dr. Clarke is the recipient of the Lillian Fountain
Smith Endowed Professorship of Human Nutrition at Colorado State Uni-
versity.
244 Nutrition and Gene Expression
REFERENCES
I. AHmann, D. W. and Gibson, D. W., Fatty acid synthesis during early linoleic acid
deficiency in the mouse, J. Lipid Res., 6, 51, 1969.
2. Sabine, J. R., McGrath, H., and Abraham, S., Dietary fat and the inhibition of hepatic
lipogenesis in the mouse, J. Nutr., 98, 312, 1969.
3. Musch, K., Ojakian, M. S., and Williams, M. A., Comparison of o:-linolenate and
oleate in lowering activity of lipogenic enzymes in rat liver: evidence for a greater effect
of dietary linolenate independent of food and carbohydrate intake, Biochirn. Biophys.
Acta, 337, 343, 1974.
4. Clarke, S. D., Romsos, D., and Leveille, G. A., Differential effects of dietary methyl
esters of long-chain saturated and polyunsaturated fatty acids on rat liver and adipose
tissue lipogenesis, J. Nutr., 107, 1170, 1977.
5. Toussant, M. J,, Wilson, M. D., and Clarke, S. D., Coordinate suppression of liver
acetyl-CoA carboxylase and fatty acid synthase by polyunsaturated fat, J. Nutr., Ill,
146, 1981.
6. Flick, P. K., Chen, J,, and Vagelos, P.R., Effect of dietary linoleate on synthesis and
degradation of fatty acid synthetase from rat liver, J. Bioi. Chern., 252, 4242, 1977.
7. Emken, E. A., Abraham, S., and Lin, C. Y., Metabolism of cis-12-octadecenoic acid
and trans-9, trans-12 octadecadienoic acid and their influence on lipogenic enzyme ac-
tivities in mouse liver, Biochirn. Biophys. Acta, 919, Ill, 1987.
8. Clarke, B. A. and Clarke, S. D., Suppression of rat liver fatty acid synthesis by eicosa-
5,8,11, 14-tetraynoic acid without a reduction in lipogenic enzymes, J. Nutr., 112, 1212,
1982.
9. Clarke, S. D. and Abraham, S., Inhibition of fatty acid synthase gene expression by
polyunsaturated fat: a requirement for delta-6 desaturation, submitted.
10. Clarke, S. D., Armstrong, M. K., and Jump, D. B., Nutritional control of rat liver
fatty acid synthase and Sl4 mRNA abundance. J. Nutr., 120, 218, 1990.
II. Clarke, S. D., Armstrong, M. K., and Jump, D. B., Dietary polyunsaturated fats
uniquely suppress rat liver fatty acid synthase and S14 mRNA content, J. Nutr., 120,
225, 1990.
12. Blake, W. L. and Clarke, S. D., Suppression of hepatic fatty acid synthase and S14
gene transcription by dietary polyunsaturated fat, J. Nutr., 120, 1727, 1990.
13. Hillard, B. L., Lundin, P., and Clarke, S.D., Essentiality of dietary carbohydrate for
maintenance of liver lipogenesis in the chick,}. Nutr., 110, 1533, 1980.
14. Bortz, W. M., Abraham, S., and Chaikoff, I. L., Localization of the block in lipo-
genesis resulting from feeding fat, J. Bioi. Chern., 238, 1266, 1963.
15. Romsos, D. R. and Leveille, G. A., Effects of diet on activity of enzymes involved in
fatty acid and cholesterol synthesis, Adv. Lipid Res., 12, 97. 1974.
16. Wilson, M. D., Blake, W. L., Salati, L. M., and Clarke, S. D., Potency of polyun-
saturated and saturated fats as short term inhibitors of hepatic lipogenesis in rats, J. Nutr.,
120. 544, 1990.
17. Clarke, S. D. and Hillard, B. L., Suppression of hepatocyte fatty acid synthesis by
albumin-bound linoleate involves depolymerization of acetyl-CoA carboxylase filaments,
Lipids, 15, 207, 1981.
18. Ashcraft, B. A., Fillers, W. S., Augustine, S. L., and Clarke, S.D., Polymer-protomer
transition of acetyi-CoA carboxylase occurs in vivo and varies with nutritional conditions,
J. Bioi. Chern., 255, 10033, 1980.
19. Clarke, B. A. and Clarke, S.D., Polymer-protomer transition of acetyl-CoA carboxylase
as a regulator of lipogenesis in rat liver, Arch. Biochern. Biophys., 218, 92, 1982.
20. Schwartz, R. S. and Abraham, S., Effect of dietary polyunsaturated fatty acids on the
activity and content of fatty acid synthetase in mouse liver, Biochirn. Biophys. Acta, 711,
316, 1982.
Clarke and Jump 245
21. Katsurada, A., Iritani, N., Fukuda, H., Matsumura, Y., Nishimoto, N., Noguchi,
T., and Tanaka, T., Effects of nutrients and hormones on transcriptional and post-
transcriptional regulation of acetyl-CoA carboxylase in rat liver, Eur. }. Biochem., 190,
435, 1990.
22. Tomlinson, J, E., Nakayama, R., and Holten, D., Repression of pentose phosphate
pathway dehydrogenase synthesis and mRNA by dietary fat in rats, J. Nutr., 118,408,
1988.
23. Dempsey, M. E., Hargis, P. S., McGuire, D. M., McMahon, A., Olson, C. D.,
Salati, L. M., Clarke, S. D., and Towle, H. C., Role of sterol carrier protein in
cholesterol metabolism, Chern. Phys. Lipids, 38, 223, 1985.
24. Salati, L. M., Hargis, P. S., Olson, C. D., Clarke, S. D., and Dempsey, M. E.,
Rapid regulation of sterol carrier protein synthesis, secretion, and turnover in rat hepa-
tocytes, Fed. Proc., 45 (Abstr.), 1581, 1986.
25. Clarke, S. D., Wilson, M. D., and lbnoughazala, T., Resistance of lung fatty acid
synthesis to inhibition by dietary fat in the meal-fed rat, J. Nutr., 114, 598, 1984.
26. Armstrong, M. K., Blake, W. L., and Clarke, S. D., Arachidonic acid suppression
of fatty acid synthase gene expression in cultured rat hepatocytes, Biochem. Biophys.
Res. Commun., 177. 1056, 1991.
27. Salati, L. M., Adkins-Finke, B. A., and Clarke, S.D., Fatty acid regulation of glucose-
6-phosphate dehydrogenase in rat liver cell monolayers, Lipids, 23, 36, 1988.
28. Odin, R. S., Adkins-Finke, B. A., Blake, W. L., Phinney, S.D., and Clarke, S. D.,
Membrane phospholipid enrichment of cultured rat hepatocytes with omega-3 and omega-
6 fatty acids is associated with decreased triglyceride production and secretion, Biochim.
Biophys. Acta, 921, 378, 1987.
29. Reid, L. M., Narita, M., Fujita, M., and Rosenberg, L., Isolated and Cultured
H epatocytes, Guill ouzo, A. and Gugen-Guillouzo, C., Eds., Hohn Libbey, London, 1986,
225.
30. Schwartz, R. S. and Abraham, S., Effect of dietary fat on the activity, content, rates
of synthesis, and degradation and translation of messenger RNA coding for malic enzyme,
Arch. Biochem. Biophys., 221, 206, 1983.
31. Szepesi, B., Kamara, A. K., and Clarke, S.D., Lack of specificity of polyunsaturated
fats in the inhibition of rat liver glucose-6-phosphate dehydrogenase, J. Nutr., 119, 161,
1989.
32. Nebert, D. W., Proposed role of drug-metabolizing enzymes: regulation of steady state
levels of the ligands that effect growth, homeostasis, differentiation, and neuroendocrine
functions, Mol. Endocrinol., 5, 1203, 1991.
33. Dozin, B., Rail, J., and Nikodem, V. M., Tissue-specific control of rat malic enzyme
activity and messenger RNA levels by a high carbohydrate diet, Proc. Nat/. Acad. Sci.
U.S.A., 83, 4705, 1986.
34. Katsurada, A., Iritani, N., Fukuda, H., Noguchi, T., and Tanaka, T., Influence of
diet on the transcriptional and post -transcriptional regulation of malic enzyme induction
in the rat liver, Eur. J. Biochem., 168, 487, 1987.
35. Amy, C. M., Williams-Ahlf, B., Naggert, J,, and Smith, S., Molecular cloning of
the mammalian fatty acid synthase gene and identification of the promoter region, Biochem.
J., 271, 675, 1990.
36. MacDougal, 0., Clarke, S.D., and Jump, D. B., Unpublished observations.
37. Jump, D. B., Bell, A., Lepar, G., and Hu, D., Insulin rapidly induces rat liver Sl4
gene transcription, Mol. Endocrinol., 4, 1655, 1990.
38. Salati, L. M. and Clarke, S. D., Fatty acid inhibition of the hormonal induction of
acetyl-CoA carboxylase in hepatocyte monolayers, Arch. Biochem. Biophys., 246, 82,
1986.
246 Nutrition and Gene Expression
39. Dax, E. M., Partilla, J. S., Pineyro, M.A., and Gregerman, R. 1., Altered glucagon-
and catecholamine hormone-sensitive adenylyl cyclase responsiveness in rat liver mem-
branes induced by manipulation of dietary fatty acid intake, Endocrinology, 127, 2236,
1990.
40. Field, C. J., Ryan, E. A., Thomson, A. B. R., and Clandinin, M. T., Diet fat
composition alters membrane phospholipid composition insulin binding, and glucose
metabolism in adipocytes from control and diabetic animals, J. Bioi. Chem., 265, 11143,
1990.
41. Blake, W. L. and Clarke, S. D., Type of dietary fat alters rat liver cell responsiveness
to vasopressin, J. Nutr. Biochem., 2, 87, 1991.
42. Clandinin, M. T., Cheema, S., Field, C. J., Garg, M. L., Vendatraman, J., and
Clandinin, T. R., Dietary fat: exogenous determination of membrane structure and cell
function, FASEB J., 5, 2761, 1991.
43. O'Brien, R. M., Lucas, P. C., Forest, C. D., Magnuson, M. A., and Granner,
D. K., Identification of a sequence in the PEPCK gene that mediates a negative effect
of insulin on transcription, Science, 249, 533, 1990.
44. Clarke, S. D. and Hembree, J., Dietary fat suppresses the triiodothyronine induction
of rat liver lipogenic enzymes, J. Nutr., 120, 625, 1990.
45. Dawson, P. A., Hofmann, S. L., Vander Westhuyzen, D. R., Sudhof, T. C., Brown,
M. S., and Goldstein, J. L., Sterol-dependent repression of low density lipoprotein
receptor promoter mediated by 16-base pair sequence adjacent to binding site for tran-
scription factor Spl, J. Bioi. Chem., 263, 3372, 1988.
46. Issemann, I. and Green, S., Activation of a member of the esteroid hormone receptor
superfamily by peroxisome proliferators, Nature (London), 347, 645, 1990.
47. Clarke, S. D., Romsos, D. R., and Leveille, G. A., Influence of dietary fatty acids
on liver and adipose tissue lipogenesis and on liver metabolites in meal-fed rats, J. Nutr ..
107, 1277, 1977.
48. Herzberg, G. R., The influence of dietary fatty acid composition on lipogenesis, Adv.
Nutr. Res., 5, 221, 1983.
49. Christiansen, E. N., Lund, J. S., Rortveit, T., and Rustan, A. C., Effect of dietary
n-3 and n-6 fatty acids on fatty acid desaturation, Biochim. Biophys. Acta. 1082, 57,
1991.
50. Baltzell, J. K., Wooten, J. T., and Otto, D. A., Lipoprotein lipase in rats fed fish oil:
apparent relationship to plasma insulin levels, Lipids, 26, 289.
51. Miller, J. C. E., Barth, R. K., Shaw, P. H., Elliot, R. W., and Hastie, N. D.,
Identification of a eDNA clone for mouse apoprotein A-1 (apo A 1) and its use in
characterization of apo A-1 mRNA expression in liver and small intestine, Proc. Nat/.
Acad. Sci. U.S.A., 80, 1511, 1983.
Chapter 11
Howard P. Glauert
TABLE OF CONTENTS
-------
0-8493-6961-4/93/$0.00+$50
C0 1993 by CRC Press, 1nc. 247
248 Nutrition and Gene Expression
I. INTRODUCTION
The amount and type of fat consumed in the diet may be important in
the development of human cancer. Several organizations have advocated
decreasing the fat content of the diet as a means of preventing the development
of cancer. For example, the American Cancer Society 1 advises people to "cut
down on total fat intake'', and the American Institute for Cancer Research 2
advises people to "reduce the intake of total dietary fat from the current
average of approximately 37% to a level of no more than 30% of total calories
and, in particular, reduce the intake of saturated fat."
In this chapter, the role of dietary fat on the development of human and
experimental cancer will be discussed. The role of gene expression as a
mechanism by which dietary fat may alter the development of cancer will be
examined. Cancer clearly is a disease of alterations both in genetic structure
and in genetic expression, both of which can be affected by dietary fat.
A. CHEMICAL CARCINOGENESIS
At present, epidemiologists believe that 60 to 90% of human cancers are
caused by environmental agents. 3 Worldwide, the incidence of cancer in
different tissues and organs varies widely from country to country. 4 When
people migrate from one country to another, their descendants tend to develop
the same types of cancers as the native population.
The main environmental agents which are thought to cause cancer are
viruses, radiation, and chemicals. 5 The induction of cancer by chemicals was
first documented in 1775 by Percival Pott, who found that men who worked
as chimney sweeps had a high incidence of scrotal cancer. 6 Many other human
carcinogens have since been identified, including azo dyes, benzene, vinyl
chloride, and cigarette smoke. 3 In animals, chemical carcinogenesis was first
observed in the early 1900s, when the application of coal tar to skin was
found to induce tumors. 7•8 Subsequently, chemicals isolated from coal tar,
the polycyclic aromatic hydrocarbons (PAH), were found to induce skin tu-
mors in mice. 9 • 10 Several other classes of chemicals have since been found
to induce tumors, including aromatic amines, dialkylnitrosamines, and al-
kylating agents. 3
In order to induce cancer, most chemical carcinogens have to be metab-
olized to some other form. The procarcinogens are generally metabolized by
mixed-function oxidases in the smooth endoplasmic reticulum to electrophilic
forms (the ultimate carcinogens). 3 These electrophiles then react with nu-
cleophilic sites in the cell, such as the nitrogen and oxygen atoms in DNA.
The binding of an ultimate carcinogen to DNA can lead to mispairing during
DNA replication and thus to the formation of mutations in DNA. Chemical
carcinogens or their reactive metabolites have been shown to be mutagenic
in bacterial and mammalian mutagenesis systems. 11 • 12
Glauert 249
B. MULTISTAGE CARCINOGENESIS
After the administration of a chemical carcinogen, there is a latency period
before the actual appearance of a neoplasm. The latency period may vary
depending on the carcinogen used, its dosage, and the tissue or organ being
investigated, but it may be considered to be a general feature of the patho-
genesis of neoplasia. 13 Concurrently with early investigations into the latency
phenomenon, Rous and Kidd 14 provided experimental evidence suggesting a
two-stage mechanism for carcinogenesis in skin. They found that coal tar
application could "initiate" tumor formation while wounding could "pro-
mote" this process. Subsequently, Berenblum and Shubik 15 clearly demon-
strated that skin carcinogenesis could be divided into an initiation stage fol-
lowed by a longer promotion stage. These authors and many others have
demonstrated that administering a low dose of an initiating agent (which may
be a chemical, UV light, or ionizing radiation) to mouse skin normally results
in few or no tumors being formed during the animal's lifetime. Subsequent
repeated application of a promoting agent (such as croton oil or its most potent
component, 12-0-tetradecanoylphorbol-13-acetate [TPA]) over several weeks
or months will, however, induce the formation of skin papillomas and car-
cinomas.
Subsequent studies have demonstrated that carcinogenesis in other tis-
sues-such as the liver, mammary gland, bladder, and pancreas-can also
be divided into the two stages of initiation and promotion. 5 Initiation and
promotion, regardless of the tissue in question, have certain common prop-
erties. In two-stage carcinogenesis, the administration of a single subcarcin-
ogenic dose of a carcinogen (i.e., initiation) is followed by the long-term
administration of a promoter. 16 Initiation is not reversible, whereas promotion
is. Neither the administration of an initiator alone, repetitive doses of the
promoter alone, nor the administration of the promoter before the initiator
will lead to tumor production: the application of the initiator must be followed
by repeated doses of the promoter. 16 Mechanistically, an initiator has been
defined as an agent which irreversibly alters the native molecular structure
of DNA (i.e., produces a mutation), whereas a promoter has been defined
as an agent that alters the expression of genetic information in the cell. 13
An additional model for multistage carcinogenesis has been proposed by
Moolgavkar and colleagues. 17 In this model, two mutations must occur in
order for a cell to become neoplastic. The rate at which a tumor appears can
be influenced by two factors: an increase in the mutation rate, which could
be brought about by a genotoxic carcinogen, or an increase in the rate of cell
proliferation, which would expand the population of normal and intermediate
cells (i.e., cells with one mutation), and increase the likelihood of a mutation
occurring. A tumor-promoting agent could bring about this second mecha-
msm.
250 Nutrition and Gene Expression
A. NUTRITIONAL CONSIDERATIONS
In experiments examining the effect of dietary fat, fat has to be substituted
for carbohydrate isocalorically in order to ensure that fat is the only experi-
mental variable. In some studies, fat was added to a nutritionally complete
diet, such as a ground unrefined diet. Since animals consume approximately
the same number of calories, animals on such a high fat diet would consume
less of all other nutrients in the diet. Therefore, any changes seen in the high
fat group could not strictly be attributed to a higher consumption of fat. In
other studies, fat was substituted for a carbohydrate (such as corn starch or
dextrose) on a gram-for-gram basis. Since fat contains 9 kcal per gram and
carbohydrate contains only 4 kcal per gram/ 7 such a protocol would also
result in lower consumption of other nutrients in the diet. In order to substitute
fat for carbohydrate on a metabolizable energy basis, 4 g of fat have to be
substituted for 9 g of carbohydrate.
Another issue in studies examining dietary fat and carcinogenesis is whether
the enhancing effect of dietary fat, when it is seen. is due to dietary fat per
se (for example, by an effect on bile acid metabolism in colon carcinogenesis)
or rather to a more efficient utilization of calories. Even when the ratio of
calories to nutrients remains the same when the dietary fat content of the diet
is raised, animals frequently gain more weight than animals on a low fat diet.
When fat is substituted for carbohydrate isocalorically, the substitution is done
on the basis of metabolizable energy rather than net energy. Donato and
Hegsted/ 8 however, have demonstrated that fat is used more efficiently than
carbohydrate, so that equating their metabolizable energy values may be
inaccurate.
252 Nutrition and Gene Expression
B. SKIN CARCINOGENESIS
Mouse skin is one of the oldest and most widely used systems for studying
chemical carcinogenesis, including multistage carcinogenesis. As discussed
earlier, two-stage carcinogenesis (initiation-promotion) was first observed in
mouse skin and involves initiation by a subcarcinogenic dose of radiation or
of a chemical, such as PAH, followed by the long-term administration of
croton oil or its active ingredient TPA.
Most studies examining dietary fat have studied complete carcinogenesis
by PAH or ultraviolet light. Early studies demonstrated that high fat diets
enhanced skin carcinogenesis induced by tar39 or PAH. 40 -46 In studies where
skin tumors were induced by ultraviolet light, Mathews-Roth and Krinsky47
found that high fat diets increased skin carcinogenesis, whereas Black et a!. 48
found that high fat diets did not increase skin carcinogenesis, but that feeding
a saturated fat inhibited tumorigenesis.
The effect of fatty acids on the initiation and promotion of skin carcin-
ogenesis has also been studied. Certain fatty acids-oleic acid and lauric
acid-have promoting activity when applied daily to mouse skin after a single
application of 7, 12-dimethylbenz(a)anthracene (DMBA); stearic acid and pal-
mitic acid do not have any effect. 49 When diets varying in their fat content
are fed during the initiation or promotion stages of DMBA-initiated, TPA-
promoted skin carcinogenesis, high fat diets enhance the promotion of skin
carcinogenesis and slightly inhibit initiation. 50 •51
C. HEPATOCARCINOGENESIS
Many early studies of dietary fat and cancer used the liver as the target
organ. In these studies, aromatic amines and azo dyes were frequently used
to induce hepatocellular carcinomas. In later studies, effects of dietary fat on
initiation and promotion in the liver were examined. In initiation-promotion
protocols, the administration of a single subcarcinogenic dose of a carcinogen
(such as diethylnitrosamine [DEN] or DMBA) along with a proliferative
stimulus (such as partial hepatectomy) followed by the long-term feeding of
chemicals such as phenobarbital, 2,3,7,8-tetrachlorodibenzo-p-dioxin, or po-
lyhalogenated biphenyls, leads to a high incidence of hepatocellular adenomas
and carcinomas. 13 •52 In addition, foci of putative preneoplastic hepatocytes
appear before the development of gross tumors. These foci, known as altered
hepatic foci or enzyme-altered foci, contain cells which exhibit qualitatively
altered enzyme activities or alterations in one or more cell functions. 53 The
enzymes most frequently studied include -y-glutamyltranspeptidase (GGT) and
placental glutathione-S-transferase, which are normally not present in adult
liver, but which are often present in foci, and ATPase and glucose-6-phos-
phatase, which are normally present but which are frequently missing from
foci. 54 •55 Altered hepatic foci can also be identified on hematoxylin and eosin-
stained tissue. 56 •57 The appearance of foci has been correlated with the later
development of malignant neoplasms. 58 •59
Glauert 253
The first studies examined the effect of dietary fat on the induction of
hepatocellular carcinomas by complete hepatocarcinogens. In the liver, in-
creasing the fat content of the diet enhances the development of 2-acetylam-
inofluorene (AAF)-, p-dimethylaminoazobenzene (DAB)-, and aflatoxin B 1
(AFB)-induced tumors and GGT-positive foci in rats. 60 ·63 Furthermore, he-
patocarcinogenesis by DAB is enhanced by feeding a diet which contains a
greater proportion of polyunsaturated fatty acids. 64 •65 In these studies, how-
ever, the diets were administered at the same time as the carcinogen injections,
so that the stage of carcinogenesis which was affected could not be determined.
More recent studies have examined whether this enhancement of hepa-
tocarcinogenesis is caused by an effect on the initiation of carcinogenesis,
the promotion of carcinogenesis, or both. Misslbeck et al. 66 found that in-
creasing the com oil content of the diet after the administration of I 0 doses
of aflatoxin increased the number and size of GGT -positive foci, but Baldwin
and Parker, 67 using a similar protocol, found no effect of dietary com oil.
Glauert and Pitot68 similarly found that increasing the safflower oil or palm
oil content of the diet did not promote DEN-induced GGT-positive foci or
greatly affect phenobarbital promotion of GGT-positive foci. The promotion
of GGT-positive foci by dietary tryptophan also is not affected by dietary
fat. 69 Newbeme et al. 70 found that increasing dietary com oil (but not beef
fat) during and after the administration of AFB increased the incidence of
hepatic tumors, but not when the diets were fed only after AFB administration.
Baldwin and Parke~ 7
also found that increasing the com oil content of the
diet before and during AFB administration increased the number and volume
of GGT-positive foci. Recently, it has been found that when rats are fed diets
high in polyunsaturated fatty acids (but not in saturated fatty acids) before
receiving the hepatocarinogen DEN, they develop more GGT-positive and
ATPase-negative foci than rats fed low fat diets. 71 The results of these studies
suggest that the enhancement of hepatocarcinogenesis by dietary fat is pri-
marily due to an effect on initiation, and that polyunsaturated fats have a
greater effect than do saturated fats.
D. COLON CARCINOGENESIS
Studies in experimental animals have produced differing results. Because
no animal models develop colon tumors spontaneously, a variety of chemi-
cals have been used to induce colon tumors, usually in rats or mice. These
include 1,2-dimethylhydrazine (DMH) and its metabolites azoxymethane
(AOM) and methylazoxymethanol (MAM); 3,2 '-dimethyl-4-aminobiphenyl
(DMAB); methylnitrosourea (MNU); and N-methyl-N' -nitro-N-nitrosoguan-
idine (MNNG). 72 "75 DMH and AOM have been used most frequently to study
nutritional effects. Both can induce colon tumors by single76. 80 or multi-
ple81.85 injections.
Animal studies examining the effect of dietary fat have used a variety of
protocols, and the results obtained often have been dependent on the inves-
254 Nutrition and Gene Expression
tigator's protocol. In several studies, fat was either added to a chow diet or
was substituted for carbohydrate on a weight basis, so that the ratio of calories
to essential nutrients was altered. 83 ·84 •8691 These studies all found that in-
creasing dietary fat enhanced colon carcinogenesis, but the effect could have
been due to a lower consumption of essential nutrients rather than to an effect
of fat. In other studies, rats or mice were subjected to multiple doses of a
colon carcinogen, with the dietary fat content being varied (isocalorically)
during, and frequently before or after, the carcinogen injections. Some of
these studies found an enhancement when the dietary fat content of the diet
was increased, but most saw no effect and one saw an inhibition of tumor
development. 73 ·75 •81 •82 •92 -95 Feeding unsaturated rather than saturated fat in the
diet also was found to increase colon carcinogenesis induced by multiple
injections of AOM. 96 More recent studies have examined the effect of the
amount and type of dietary fat on the initiation and promotion of colon
carcinogenesis induced by one, two, or three injections of AOM. Reddy and
colleagues found that increasing the unsaturated fat, but not the saturated fat,
content of the diet during the promotional phase enhanced colon carcinogen-
esis; the initiation of colon carcinogenesis by AOM, however, was not affected
by the level of unsaturated fat, but was slightly enhanced by increased levels
of saturated fat. 97 -99 Other investigators, however, did not see an enhancement
of the promotional phase by dietary polyunsaturated fat. 100 •101
E. PANCREATIC CARCINOGENESIS
Dietary fat has been studied extensively in animal models, primarily rats
and hamsters. In rats, azaserine is the chemical carcinogen usually used. 102
Single or multiple doses of this chemical induce carcinomas derived from
acinar cells. Putative preneoplastic lesions-basophilic and acidophilic foci-
have also been described in the rat pancreas. 102 The carcinogen N-nitrosobis(2-
oxopropyl)arnine (BOP) has been used primarily to induce pancreatic tumors
in hamsters, although it also has been shown to be effective in rats. 102 In
hamsters, tumors are derived from ductal cells, as are the majority of human
pancreatic tumors. The hamster model, therefore, may be more relevant to
human pancreatic cancer.
Dietary fat has been found to influence tumorigenesis in both animal
models. In rats, feeding high-fat diets after, or during and after, the injection
of azaserine enhances the development of pancreatic tumors and putative
preneoplastic lesions. 103 -' 11 Pancreatic carcinogenesis induced by N-nitroso(2-
hydroxypropyl) (2-oxopropyl)amine in rats is also enhanced by feeding high
fat diets. 112 In several of these studies, the effect cannot be attributed un-
equivocally to dietary fat because fat was substituted for carbohydrate on a
weight basis. In hamsters, BOP-induced pancreatic carcinogenesis is also
increased by feeding high-fat diets. 108- 110 •113- 116 Roebuck and colleagues 104 •105 •111
found that polyunsaturated fat, but not saturated fat, enhanced pancreatic
carcinogenesis, and that a certain level of essential fatty acids are required
Glauert 255
F. MAMMARY CARCINOGENESIS
The effect of dietary fat on mammary carcinogenesis in experimental
animals has been examined extensively: over 100 experiments have been
conducted. 119 Two animal models are primarily used: a rat model (usually the
Sprague-Dawley strain) in which mammary tumors are induced by DMBA
or MNU and a mouse model, in which tumors develop spontaneously. The
use of these models is advantageous because tumor latency, tumor size, and
tumor progression can easily be quantified by palpation of mammary tumors
as they appear. In both models, increasing the fat content of the diet clearly
enhances the development of mammary tumors. 119 In the rat model, a high
fat diet increases tumorigenesis both when it is fed during and after carcinogen
administration, and when it is fed only after carcinogen injection.
G. OTHER SITES
Dietary fat has also been studied for its effect on experimental carcino-
genesis in other organs. In the lung, dietary fat enhanced BP- or BOP-induced
carcinogenesis in hamsters, 114 • 120 whereas in mice, a high fat diet did not
affect spontaneous carcinogenesis in one study, but the feeding of egg extracts
enhanced it in another. 44 • 121 Spontaneous brain tumors in mice and prostate
tumors in rats were increased by the feeding of high fat diets, as was the
induction of BOP-induced renal tumors, but DMH- or MAM-induced renal
or ear duct tumors were not affected by dietary fat. 87• 114 • 122 • 123
tuting menhaden oil for com oil or coconut oil did not affect skin tumor
promotion by TPA.
A. MEMBRANE FLUIDITY
An important function of dietary fatty acids is their presence in membrane
lipids. Altering the fatty acid content of the diet alters the composition of
membrane lipids, particularly in certain tissues; feeding diets high in n-6 or
n-3 fatty acids increases the concentrations of these fatty acids in membrane
lipids. 133 The activities of membrane-bound enzymes are increased in mem-
branes that are more fluid, i.e., that have a higher content of polyunsaturated
fatty acids. 133 Higher amounts of unsaturated fatty acids in cell membranes
are also associated with a higher rate of cell proliferation. 134 The alteration
by dietary fatty acids of the catalytic abilities of membrane-bound enzymes,
such as cytochrome P-450, may play an important role in carcinogenesis.
B. TOXICITY
One possible mechanism by which dietary fat may enhance carcinogenesis
is by the toxicity of fatty acids or of metabolites that increase after the feeding
of high fat diets. Such toxicity would bring about a proliferative response in
the tissue to replace lost cells. Cellular genes involved in cell proliferation,
including cellular oncogenes, would likely be increased.
In the colon, toxicity may play a role in the enhancement of carcinogenesis
by dietary fat. One hypothesis for the effect of dietary fat is that dietary fat
increases the concentration of metabolites with carcinogenic or promoting
activity in the fecal stream. Bile acids, particularly secondary bile acids, have
promoting activity in the colon; 135 - 137 their concentration in the feces has been
found to be increased by dietary fat in some but not all studies. 83 • 138- 141 Bile
acids function as detergents; therefore, high concentrations may be toxic to
epithelial cells in the colon. Increasing the concentration of bile acids as well
as fatty acids in the colon has been demonstrated to increase colon epithelial
cell proliferation. 142 - 146 It has been further demonstrated that the increased
expression of protein kinase C may play a role in bile acid-induced cell
proliferation. 147
C. EICOSANOID METABOLISM
Another mechanism by which dietary fat may influence carcinogenesis
is by altering the synthesis of eicosanoids. Fatty acids that are consumed in
the diet can be metabolized to a variety of other compounds, including longer
and more unsaturated fatty acids, prostaglandins, leukotrienes, thromboxanes,
hydroperoxyeicosatetraenoic acids, and hydroxyeicosatetraenoic acids. 148 Al-
tering the type of fatty acid in the diet changes the composition of the ei-
Glauert 257
cosanoids that are produced by the body. 149 For example, increasing the n-3
content of the diet will alter the type of eicosanoids that are produced as well
as their function. 150 Specific eicosanoids bind to receptors and cause specific
alterations in gene expression and cellular function, 151 some of which may
be related to carcinogenesis. It has been found that inhibition of eicosanoid
synthesis inhibits DNA synthesis and tumor promotion in both the skin and
liver. 152-154
D. CALORIC EFFECTS
The issue of whether the enhancing effect of fat in carcinogenesis IS
due to higher consumption of calories or more efficient utilization of energy
has been examined in several tissues. The earliest study was conducted by
Boutwell et a!. 46 using the mouse skin carcinogenesis system; they attributed
most of the enhancing effect of dietary fat to an increased consumption of
calories. Birt eta!., 51 however, found that the promotion of skin carcinogenesis
was enhanced even though the high-fat diets were pair-fed. The greater caloric
density of fat has also been proposed to play a role in colon tumorigenesis.
Caloric restriction inhibits chemically induced colon carcinogenesis, even if
the percentage of dietary fat in the diet is greatly increased. 155 • 156 In the
pancreas, the enhancement by dietary fat appears to be an effect of dietary
fat rather than of an increased consumption of calories, as pair-feeding does
not inhibit the enhancing effect of dietary fat in hamsters. 115 Several studies
have suggested that the enhancement of mammary carcinogenesis by dietary
fat may be caused, at least in part, by an alteration in the efficiency of energy
utilization. 157 - 159 Recently, Freedman et a!., 119 using a combined statistical
analysis of over 100 animal experiments, concluded that the enhancing effect
of high-fat diets was caused in part by a higher caloric intake and in part by
some other effect of dietary fat. Finally, caloric restriction has been found to
inhibit tumorigenesis in many tissues in experimental animals. 160
Several mechanisms have been proposed to explain how caloric con-
sumption affects carcinogenesis. These include effects on growth factors,
immunity, neuroendocrine function, and metabolic regulation.' 60 • 161 Any of
these effects could be brought about by alterations in gene expression.
E. EFFECT ON INITIATION
Dietary fat may also affect the initiation stage of carcinogenesis. Since
initiation involves the mutation of DNA, its alteration (by dietary fat or other
agents) would mainly affect the structure of genes rather than their expression.
In the liver, dietary fat appears to enhance carcinogenesis primarily by an
effect on initiation. In other tissues, many protocols have varied the levels
of dietary fat during the time of carcinogen injections; therefore dietary fat
may be affecting some aspect of initiation in these studies. Higher levels of
dietary fat may enhance initiation of carcinogenesis by several mechanisms,
including alterations in absorption of the carcinogen from the gut, transport
258 Nutrition and Gene Expression
Clearly there is much variability in studies of dietary fat and cancer, both
in epidemiological and experimental studies. In epidemiological studies, a
relationship between dietary fat and colon, pancreatic, and mammary cancer
has been found in correlational and case-control studies. Few prospective
studies, however, have found an enhancement of these cancers by dietary fat,
and, for colon cancer, two studies have found that a high-fat diet is protective.
Glauert 259
REFERENCES
I. American Cancer Society, Cancer Facts and Figures 1991, American Cancer Society,
Atlanta, 1991.
2. American Institute for Cancer Research, Dietary Guidelines to Lower Cancer Risk, Amer-
ican Institute for Cancer Research, Washington, D.C., 1990.
3. Miller, E. C., Some current perspectives on chemical carcinogenesis in humans and
experimental animals, Cancer Res., 38, 1479, 1978.
4. Armstrong, B. and Doll, R., Environmental factors and cancer incidence and mortality
in different countries with special references to dietary practices, Br. J. Cancer, 15,
617, 1975.
5. Pitot, H. C., Fundamentals of Oncology, 3rd ed., Marcel Dekker, New York, 1986.
6. Pott, P., Chirurgical observations relative to cancer of the scrotum, Reprinted in Nat/.
Cancer lnst. Monogr., 10, 7, 1963.
7. Yamagiwa, K. and Ichikawa, K., Experimental study of the pathogenesis of carcinoma,
J. Cancer Res., 3, I, 1918.
8. Tsutsui, H., Ober das kunstlich erzeugte Cancroid bei der Maus, Gann, 12, 17, 1918.
9. Cook, J. W., Hewett, C. L., and Heiger, I., The isolation of cancer-producing hydro-
carbon from coal tar, J. Chem. Soc., 395.
10. Kennaway, E. L. and Hieger, I., Carcinogenic substances and their fluorescence spectra,
Br. Med. J., ii, 1044, 1930.
II. McCann, J., Choi, E., Yamasaki, E., and Ames, B. N., The detection of carcinogens
as mutagens in the Salmonella/microsome test: assay of 300 chemicals, Proc. Nat/. Acad.
Sci. U.S.A., 72, 5135, 1975.
260 Nutrition and Gene Expression
12. DeMarini, D. M., Brockman, H. E., de Serres, F. J., Evans, H. H., Stankowski,
C. F., and Hsie, A. W., Specific-locus mutations induced in eukaryotes (especially
mammalian cells) by radiation and chemicals: a perspective, Murat. Res., 220, I I, 1989.
13. Pitot, H. C. and Sirica, A. E., The stages of initiation and promotion in hepatocarcin-
ogenesis, Biochem. Biophys. Acta, 605, 191, 1980.
14. Rous, P. and Kidd, J, G., Conditional neoplasms and subthreshold neoplastic states,
J. Exp. Med., 73, 365, 1941.
15. Berenblum, I. and Shubik, P., The role of croton oil applications, associated with a
single painting of a carcinogen, in tumour induction of the mouse's skin, Br. J. Cancer,
I, 379, 1947.
16. Diamond, L., O'Brien, T. G., and Baird, W. M., Tumor promoters and the mechanism
of tumor promotion, Adv. Cancer Res., 32, I, 1980.
17. Moolgavkar, S. H. and Knudson, A. G., Mutation and cancer: a model for human
carcinogenesis, J. Nat/. Cancer lnst., 66, 1037, 1981.
18. Stowers, S. J., Maronpot, R. R., Reynolds, S. H., and Anderson, M. W., The role
of oncogenes in chemical carcinogenesis, Environ. Health Perspect., 75, 81, 1987.
19. Sager, R., Tumor suppressor genes: the puzzle and the promise, Science, 246, 1406,
1989.
20. Smith, M. L., Yeleswarapu, L., Locker, J., and Lombardi, B., p53 mutation(s) in
diethylnitrosamine-induced foci of putative preneoplastic hepatocytes in male Fischer 344
rats, Proc. Am. Assoc. Cancer Res., 32, 136, 1991.
21. Hsu, I. C., Metcalf, R. A., Sun, T., Welsh, J, A., Wang, N.J., and Harris, C. C.,
Mutational hotspot in the p53 gene in human hepatocellular carcinomas, Nature (London),
350, 427, 1991.
22. Okamoto, A., Sameshima, Y., Yokoyama, S., Terashima, Y., Sugimura, T., Terada,
M., and Yokota, J,, Frequent allelic losses and mutations of the p53 gene in human
ovarian cancer, Cancer Res., 51, 5171, 1991.
23. Kolonel, L. N., Fat and colon cancer: how firm is the epidemiologic evidence?, Am. J.
Clin. Nutr., 45, 336, 1987.
24. Willett, W. C., Stampfer, M. J., Colditz, G. A., Rosner, B. A., and Speizer, F. E.,
Relation of meat, fat, and fiber intake to the risk of colon cancer in a prospective study
among women, New Engl. J. Med., 323, 1664, 1990.
25. Willett, W. C., Implications of total energy intake for epidemiologic studies of breast
and large-bowel cancer, Am. J. Clin. Nutr., 45, 354, 1987.
26. Lyon, J, L., Mahoney, A. W., West, D. W., Gardner, J, W., Smith, K. R., Sorenson,
A. W., and Stanish, W., Energy intake: its relationship to colon cancer risk, J. Nat/.
Cancer lnsl., 78, 853, 1987.
27. Kelsey, J, L. and Berkowitz, G. S., Breast cancer epidemiology, Cancer Res., 48,
5615, 1988.
28. Prentice, R. L., Pepe, M., and Self, S. G., Dietary fat and breast cancer: a quantitative
assessment of the epidemiological literature and a discussion of methodological issues,
Cancer Res., 49, 3147, 1989.
29. Howe, G. R., Hirohata, T., Hislop, T. G., Iscovich, J, M., Yuan, J. M., Katsouyanni,
K., Lubin, F., Marubini, E., Modan, B., Rohan, T., Toniolo, P., and Shunzhang,
Y., Dietary factors and risk of breast cancer: combined analysis of 12 case-control studies,
J. Nat/. Cancer lnsl., 82, 561, 1990.
30. Willett, W. C., Stampfer, M. J., Colditz, G. A., Rosner, B. A., Hennekens, C. H.,
and Speizer, F. E., Dietary fat and the risk of breast cancer, New Engl. J. Med., 316,
22, 1987.
31. Jones, D. Y., Schatzbin, A., Green, S. B., Block, G., Brinton, L.A., Ziegler, R. G.,
Hoover, R., and Taylor, P. R., Dietary fat and breast cancer in the National Health
and Nutrition Examination Survey I. Epidemiological followup study, J. Nat/. Cancer
lnst., 79, 465, 1987.
Glauert 261
32. Howe, G. R., Friedenreich, C. M., Jain, M., and Miller, A. B., A cohort study of
fat intake and risk of breast cancer, J. Nat/. Cancer Inst., 83, 336, 1991.
33. Wynder, E. L., An epidemiological evaluation of the causes of cancer of the pancreas,
Cancer Res., 35, 2228, 1975.
34. Durbec, J, P., Chevilotte, G., Bidart, J, M., Berthezene, P., and Sarles, H., Diet,
alcohol, tobacco and risk of cancer of the pancreas: a case-control study, Br. J. Cancer,
47, 463, 1983.
35. Norell, S. E., Ahlborn, A., Erwald, R., Jacobson, G., Lindberg-Navier, 1., Okin,
R., Tiirnberg, B., and Wiechel, K. L., Diet and pancreatic cancer: a case-control study,
Am. J. Epidemiol., 124, 894, 1986.
36. Carroll, K. K., Lipids and carcinogenesis, J. Environ. Patho/. Toxicol., 3, 253, 1980.
37. National Research Council, Recommended Dietary Allowances, lOth edition, National
Academy Press, Washington, D.C., 1989.
38. Donato, K. and Hegsted, D. M., Efficiency of utilization of various sources of energy
for growth, Proc. Nat/. Acad. Sci. U.S.A., 82, 4866, 1985.
39. Watson, A. F. and Mellanby, E., Tar cancer in mice. II. The condition of the skin
when modified by external treatment or diet, as a factor in influencing the cancerous
reaction, Br. J. Exp. Pathol., II, 311, 1930.
40. Baumann, C. A., Jacobi, H. P., and Rusch, H. P., The effect of diet on experimental
tumor production, Am. J. Hygiene, 30A, 1, 1939.
41. Jacobi, H. P. and Baumann, C. A., The effect of fat on tumor formation, Am. J.
Cancer, 39, 338, 1940.
42. Lavik, P. S. and Baumann, C. A., Dietary fat and tumor formation, Cancer Res., 1,
181, 1941.
43. Lavik, P. S. and Baumann, C. A., Further studies on the tumor-promoting action of
fat, Cancer Res., 3, 749, 1943.
44. Tannenbaum, A., The genesis and growth of tumors. III. Effects of a high fat diet,
Cancer Res., 2, 468, 1942.
45. Tannenbaum, A., The dependence of the genesis of induced skin tumors on the fat
content of the diet during different stages of carcinogenesis, Cancer Res., 4, 683, 1944.
46. Boutwell, R. K., Brush, M. K., and Rusch, H. P., The stimulating effect of dietary
fat on carcinogenesis, Cancer Res., 9, 741, 1949.
47. Mathews-Roth, M. M. and Krinsky, N. I., Effect of dietary fat level on UV-B induced
skin tumors, and anti-tumor action of beta-carotene, Photochem. Photobiol., 40, 671,
1984.
48. Black, H. S., Lenger, W., Phelps, A. W., and Thornby, J, 1., Influence of dietary
lipid upon ultraviolet-light carcinogenesis, Nutr. Cancer, 5, 59, 1983.
49. Holsti, P., Tumor promoting effects of some long chain fatty acids in experimental skin
carcinogenesis in the mouse, Acta Pathol. Microbiol. Scand., 46, 51, 1959.
50. Birt, D. F., Pelling, J. C., Tibbets, M. G., and Schweickert, L., Acceleration of
papilloma growth in mice fed high-fat diets during promotion of two-stage skin carcin-
ogenesis, Nutr. Cancer, 12, 161, 1989.
51. Birt, D. F., White, L. T., Choi, B., and Pelling, J, C., Dietary fat effects on the
initiation and promotion of two-stage skin tumorigenesis in the SEN CAR mouse, Cancer
Res., 49, 4170, 1989.
52. Silberhorn, E. M., Glauert, H. P., and Robertson, L. W., Carcinogenicity of poly-
halogenated biphenyls: PCBs and PBBs, Crit. Rev. Toxicol., 20, 439, 1990.
53. Beer, D. G. and Pitot, H. C., Biological markers characterizing the development of
preneoplastic and neoplastic lesions in rodent liver, Arch. Toxicol., 10 (Suppl.), 68, 1987.
54. Pitot, H. C., Glauert, H. P., and Hanigan, M., The significance of biochemical markers
in fhe characterization of putative initiated cell populations in rodent liver, Cancer Lett.,
29, I, 1985.
262 Nutrition and Gene Expression
55. Hendrich, S., Campbell, H. A., and Pitot, H. C., Quantitative stereological evaluation
of four histochemical markers of altered foci in multistage hepatocarcinogenesis in the
rat, Carcinogenesis, 8, 1245, 1987.
56. Harada, T., Maronpot, R. R., Morris, R. W., Stitzel, K. A., and Boorman, G. A.,
Morphological and stereological characterization of hepatic foci of cellular alteration in
control Fischer 344 rats, Toxicol. Pathol., 17, 579, 1989.
57. Bannasch, P., Enzmann, H., Klimek, F., Weber, E., and Zerban, H., Significance
of sequential cellular changes inside and outside foci of altered hepatocytes during he-
patocarcinogenesis, Toxicol. Pathol., 17, 617, 1989.
58. Kunz, H. W., Tennekes, H. A., Port, R. E., Schwarz, M., Lorke, D., and Schaude,
G., Quantitative aspects of chemical carcinogenesis and tumor promotion in liver, En-
viron. Health Perspect., SO, 113, 1983.
59. Emmelot, P. and Scherer, E., The first relevant cell stage in rat liver carcinogenesis.
A quantitative approach, Biochim. Biophys. Acta, 605, 247, 1980.
60. Kline, B. E., Miller, J, A., Rusch, H. P., and Baumann, C. A., Certain effects of
dietary fats on the production of liver tumors in rats fed p-dimethylaminoazobenzene,
Cancer Res., 6, 5, 1946.
61. Sugai, M., Witting, L.A., Tsuchiyama, H., and Kummerow, F. A., The effect of
heated fat on the carcinogenic activity of 2-acetylaminofluorene, Cancer Res., 22, 510,
1962.
62. McCay, P. B., King, M., Rikans, L., and Pitha, J, V., Interactions between dietary
fats and antioxidants on DMBA-induced hyperplastic nodules and hepatomas, J. Environ.
Pathol. Toxicol., 3, 451, 1980.
63. Baldwin, S. and Parker, R. S., The effect of dietary fat and selenium on the development
of preneoplastic lesions in rat liver, Nutr. Cancer, 8, 273, 1986.
64. Miller, J, A., Kline, B. E., Rusch, H. P., and Baumann, C. A., The carcinogenicity
of p-dimethylaminoazobenzene in diets containing hydrogenated coconut oil, Cancer Res.,
4, 153, 1944.
65. Miller, J, A., Kline, B. E., Rusch, H. P., and Baumann, C. A., The effect of certain
lipids on the carcinogenicity of p-dimethylaminoazobenzene, Cancer Res., 4, 756, 1944.
66. Misslbeck, N. G., Campbell, T. C., and Roe, D. A., Effect of ethanol consumed in
combination with high or low fat diets on the post initiation phase of hepatocarcinogenesis,
J. Nutr., 114, 2311, 1984.
67. Baldwin, S. and Parker, R. S., Influence of dietary fat and selenium in initiation and
promotion of aflatoxin B,-induced preneoplastic foci in rat liver, Carcinogenesis, 8, 101,
1987.
68. Glauert, H. P. and Pitot, H. C., Effect of dietary fat on the promotion of diethylni-
trosamine-induced hepatocarcinogenesis in female rats, Proc. Soc. Exp. Bioi. Med., 181,
498, 1986.
69. Sidransky, H., Verney, E., and Wang, D., Effects of varying fat content of a high
tryptophan diet on induction of gamma-glutamyltranspeptidase positive foci in the livers
of rats treated with hepatocarcinogen, Cancer Lett., 31, 235, 1986.
70. Newberne, P.M., Weigert, J,, and Kula, N., Effects of dietary fat on hepatic mixed-
functions oxidases and hepatocellular carcinoma induced by aflatoxin B I in rats, Cancer
Res., 39, 3986, 1979.
71. Glauert, H. P., Lay, L. T., Kennan, W. S., and Pitot, H. C., Effect of dietary fat
on the initiation of hepatocarcinogenesis by diethylnitrosamine or 2-acetylaminofluorene
in rats, Carcinogenesis, 12, 991, 1991.
72. Druckrey, H., Production of colonic carcinomas by 1,2-dialkyhydrazines and azoalkanes,
in Carcinoma of the Colon and Antecedent Epithelium, Burdette, W. J., Ed., Springfield,
IL, Thomas, 1970, 267.
73. Reddy, B. S. and Ohmori, T., Effect of intestinal microflora and dietary fat on 3,2'-
dimethyl-4-aminobiphenyl-induced colon carcinogenesis in F344 rats, Cancer Res., 41,
1363, 1981.
Glauert 263
74. Rogers, A. E. and Nauss, K. M., Rodent models for carcinoma of the colon, Dig. Dis.
Sci., 30, 87S, 1985.
75. Nauss, K. M., Locniskar, M., Sondergaard, D., and Newberne, P. M., Lack of
effect of dietary fat on N-nitrosomethylurea (NMU)-induced colon tumorigenesis in rats,
Carcinogenesis, 5, 255, 1984.
76. Glauert, H. P. and Weeks, J. A., Dose- and time-response of colon carcinogenesis in
Fischer-344 rats after a single dose of I ,2-dimethylhydrazine, Toxicol. Letters, 48, 283,
1989.
77. Karkare, M. R., Clark, T. D., and Glauert, H. P., Effect of dietary calcium on colon
carcinogenesis induced by a single injection of I ,2-dimethylhydrazine in rats, J. Nutr.,
121, 568, 1991.
78. Schiller, C. M., Curley, W. H., and McConnell, E. E., Induction of colon tumors by
a single oral dose of 1,2-dimethylhydrazine, Cancer Lett., II, 75, 1980.
79. Decaens, C., Gautier, R., Bara, J., and Burtin, P., Induction of rat intestinal carcin-
ogenesis with single doses, low and high repeated doses of I ,2-dimethylhydrazine, Car-
cinogenesis, 10, 69, 1989.
80. Ward, J. M., Dose response to a single injection of azoxymethane in rats, Vet. Pathol.,
12, 165, 1975.
81. Glauert, H. P., Bennink, M. R., and Sander, C. H., Enhancement of 1,2-dimethyl-
hydrazine-induced colon carcinogenesis in mice by dietary agar, Food Cosmetics Toxicol.,
19, 281, 1981.
82. Nauss, K. M., Locniskar, M., and Newberne, P.M., Effect of alterations in the quality
and quantity of dietary fat and I ,2-dimethylhydrazine-induced colon carcinogenesis in
rats, Cancer Res., 43,4083, 1983.
83. Reddy, B.S., Weisburger, J. H., and Wynder, E. L., Effects of dietary fat level and
dimethylhydrazine on fecal bile acid and neutral sterol excretion and colon carcinogenesis
in rats, J. Nat!. Cancer lnst., 52, 507, 1974.
84. Bull, A. W., Soullier, B. K., Wilson, P. S., Hayden, M. T., and Nigro, N. D.,
Promotion of azoxymethane-induced intestinal cancer by high fat diet in rats, Cancer
Res., 39, 4956, 1979.
85. Sakaguchi, M., Minuora, T., Hiramatsu, T., et at., Effects of dietary saturated and
unsaturated fatty acids on bile acids and colon carcinogenesis induced by azoxymethane
in rats, Cancer Res., 46, 61, 1986.
86. Nigro, N. D., Singh, D. V., Campbell, R. L., and Pak, M. S., Effect of dietary fat
on intestinal tumor formation by azoxymethane in rats, J. Natl. Cancer lnst., 54, 439,
1975.
87. Reddy, B.S., Watanabe, K., and Weisburger, J. H., Effect of high-fat diet on colon
carcinogenesis in F344 rats treated with l ,2-dimethylhydrazine, methylazoxymethanol
acetate, or methylnitrosourea, Cancer Res., 37, 4156, 1977.
88. Bull, A. W., Bronstein, j. C., and Nigro, N. D., The essential fatty acid requirement
for azoxymethane-induced intestinal carcinogenesis in rats, Lipids, 24, 340, 1989.
89. Reddy, B. S., Narisawa, T., Vukusich, D., Weisburger, J. H., and Wynder, E. L.,
Effect of quality and quantity of dietary fat and dimethylhydrazine on colon carcinogenesis
in rats, Proc. Soc. Exp. Bioi. Med., 151, 237, 1976.
90. Bansal, B. R., Rhoads, J. E. J., and Bansal, S. C., Effects of diet on colon carcin-
ogenesis and the immune system in rats treated with I ,2-dimethylhydrazine, Cancer Res.,
38, 3293, 1978.
91. Schmaehl, D., Habs, M., and Habs, H., Influence of a non-synthetic diet with a high
fat content on the local occurrence of colonic carcinomas induced by N-nitroso-acetox-
ymethylmethylamine (AMMN) in Sprague-Dawley rats, Hepato-gastroenterol., 30, 30,
1983.
92. Locniskar, M., Nauss, K. M., Kaufmann, P., and Newberne, P. M., Interaction of
dietary fat and route of carcinogen administration on I ,2-dimethylhydrazine-induced colon
tumorigenesis in rats, Carcinogenesis, 6, 349, 1985.
264 Nutrition and Gene Expression
93. Nauss, K. M., Bueche, D., and Newberne, P.M., Effect of beef fat on DMH-induced
colon tumorigenesis: influence of rat strain and nutrient composition, J. Nutr., 117, 739,
1987.
94. Pence, B. C. and Buddingh, F., Inhibition of dietary fat-promoted colon carcinogenesis
in rats by supplement calcium or vitamin 0 3 , Carcinogenesis, 9, 187, 1988.
95. Hardman, W. E., Heitman, D. W., and Cameron, I. L., Suppression of the progression
of I ,2 dimethylhydrazine (DMH) induced colon carcinogenesis by 20% dietary corn oil
in rats supplemented with dietary pectin, Proc. Am. Assoc. Cancer Res., 32, 131, 1991.
96. Sakaguchi, M., Hiramatsu, Y., Takada, H., Yamamura, M., Hioki, K., Saito, K.,
and Yamamoto, M., Effect of dietary unsaturated and saturated fats on azoxymethane-
induced colon carcinogenesis in rats, Cancer Res., 44, 1472, 1984.
97. Reddy, B.S. and Maeura, Y., Tumor promotion by dietary fat in azoxymethane-induced
colon carcinogenesis in female F344 rats: influences of amount and source of dietary fat,
J. Nat/. Cancer Inst., 72, 745, 1984.
98. Reddy, B. S. and Maruyama, H., Effect of different levels of dietary corn oil and lard
during the initiation phase of colon carcinogenesis in F344 rats, J. Nat/. Cancer lnst.,
77, 815, 1986.
99. Reddy, B. S. and Sugie, S., Effect of different levels of omega-3 and omega-6 fatty
acids on azoxymethane-induced colon carcinogenesis in F344 rats, Cancer Res., 48,
6642, 1988.
100. Wargovich, M. J., Allnutt, D., Palmer, C., Anaya, P., and Stephens, L. C., Inhibition
of the promotional phase of azoxymethane-induced colon carcinogenesis in the F344 rat
by calcium lactate: effect of simulating two human nutrient density levels, Cancer Lett.,
53, 17, 1990.
101. Guillem, J. G., Hsieh, L. L., O'Toole, K. M., Forde, K. A., LoGerfo, P., and
Weinstein, I. B., Changes in expression of oncogenes and endogenous retroviral-like
sequences during colon carcinogenesis, Cancer Res., 48, 3964, 1988.
102. Longnecker, D., Experimental pancreatic cancer: role of species, sex and diet, Bull.
Cancer, 77, 27, 1990.
103. Roebuck, B. D., Yager, J.D., Longnecker, D. S., and Wilpone, S. A., Promotion
by unsaturated fat of azaserine-induced pancreatic carcinogenesis in the rat, Cancer Res.,
41' 3961' 1981.
104. Roebuck, B. D., Longnecker, D. S., Baumgartner, K. J., and Thron, C. D., Car-
cinogen-induced lesions in the rat pancreas: effects of varying levels of essential fatty
acid, Cancer Res., 45, 5252, 1985.
105. Roebuck, B. D., Effects of high levels of dietary fats on the growth of azaserine-induced
foci in the rat pancreas, Lipids, 21, 281, 1986.
106. Roebuck, B. D., Kaplita, P. V., Edwards, B. R., and Praissman, M., Effects of
dietary fats and soybean protein on azaserine-induced pancreatic carcinogenesis and plasma
cholecystokinin in the rat, Cancer Res., 47, 1333, 1987.
107. O'Connor, T. P., Roebuck, B. D., and Campbell, T. C., Dietary intervention during
the postdosing phase of !-azaserine-induced preneoplastic lesions, J. Nat/. Cancer Inst.,
75, 955, 1985.
108. Woutersen, R. A. and van Garderen-Hoetmer, A., Inhibition of dietary fat promoted
development of (pre )neoplastic lesions in exocrine pancreas of rats and hamsters by
supplemental vitamins A, C and E, Cancer Lett., 41, 179, 1988.
109. Woutersen, R. A., van Garderen-Hoetmer, A., Bax, J., and Scherer, E., Modulation
of dietary fat-promoted pancreatic carcinogenesis in rats and hamsters by chronic ethanol
ingestion, Carcinogenesis, 10, 453, 1989.
110. Woutersen, R. A., van Garderen-Hoetmer, A., Bax, J., and Scherer, E., Modulation
of dietary fat -promoted pancreatic carcinogenesis in rats and hamsters by chronic coffee
ingestion, Carcinogenesis, 10, 311, 1989.
Glauert 265
Ill. Roebuck, B. D., Yager, J. D. J., and Longnecker, D. S., Dietary modulation of
azaserine-induced pancreatic carcinogenesis in the rat, Cancer Res., 41, 888, 1981.
112. Longnecker, D. S., Roebuck, B. D., and Kuhlmann, E. T., Enhancement of pancreatic
carcinogenesis by a dietary unsaturated fat in rats treated with saline or N-nitroso(2-
hydroxypropyl) (2-oxopropyl)amine, J. Natl. Cancer /nst., 74, 219, 1985.
113. Birt, D. F., Salmasi, S., and Pour, P.M., Enhancement of experimental pancreatic
cancer in Syrian golden hamsters by dietary fat, J. Natl. Cancer /nst., 67, 1327, 1981.
114. Birt, D. F. and Pour, P. M., Increased tumorigenesis induced by N-nitrosobis(2-oxo-
propyl)amine in Syrian golden hamsters fed high-fat diets, J. Nat[. Cancer /nst., 70,
1135, 1983.
115. Birt, D. F., Julius, A. D., White, L. T., and Pour, P.M., Enhancement of pancreatic
carcinogenesis in hamsters fed a high-fat diet ad libitum and at a controlled calorie intake,
Cancer Res., 49,5848,1989.
116. Birt, D. F., Julius, A. D., Dwork, E., Hanna, T., White, L. T., and Pour, P. M.,
Comparison of the effects of dietary beef tallow and com oil on pancreatic carcinogenesis
in the hamster model, Carcinogenesis, 11,745, 1990.
117. Eustis, S. L. and Boorman, G. A., Proliferative lesions of the exocrine pancreas:
relationship to com oil gavage in the National Toxicology Program, J. Nat/. Cancer /nst.,
75, 1067, 1985.
118. Haseman, J. K., Huff, J. E., Rao, G. N., Arnold, J. E., Boorman, G. A., and
McConnell, E. E., Neoplasms observed in untreated and com oil gavage control groups
of F344/N rats and (C57BL/6N X C3H/HeN)F 1 (B6C3F,) mice, J. Nat!. Cancer /nst.,
75, 975, 1985.
119. Freedman, L. S., Clifford, C., and Messina, M., Analysis of dietary fat, calories,
body weight, and the development of mammary tumors in rats and mice: a review, Cancer
Res., 50, 5710, 1990.
120. Beems, R. B. and van Beek, L., Modifying effect of dietary fat on benzo(a)pyrene-
induced respiratory tract tumors in hamsters, Carcinogenesis, 5, 413, 1984.
121. Szepsenwol, J., Carcinogenic effect of ether extract of whole egg, alcohol extract of
egg yolk, and powdered egg free of the ether extractable part in mice, Proc. Soc. Exp.
Bioi. Med., 116, 1136, 1964.
122. Szepsenwol, J., Brain nerve cell tumors in mice on diets supplemented with various
lipids, Pathol. Microbiol., 34, 1, 1969.
123. Pollard, M. and Lockert, P. H., Promotional effects of testosterone and dietary fat on
prostate carcinogenesis in genetically susceptible rats, Prostate, 6, I, 1985.
124. Cave, W. T., Dietary n-3 polyunsaturated fatty acid effects on animal tumorigenesis,
FASEB J., 5, 2160, 1991.
125. O'Connor, T. P., Roebuck, B. D., Peterson, F., and Campbell, T. C., Effect of
dietary intake of fish oil and fish protein on the development of L-azaserine-induced
preneoplastic lesions in the rat pancreas, J. Natl. Cancer /nst., 75, 959, 1985.
126. O'Connor, T. P., Roebuck, B. D., Peterson, F. J., Lokesh, B., Kinsella, J. E., and
Campbell, T. C., Effect of dietary omega-3 and omega-6 fatty acids on development of
azaserine-induced preneoplastic lesions in rat pancreas, J. Nat/. Cancer Inst., 81, 858,
1989.
127. Nelson, R. L., Tanure, J. C., Andrianopoulos, G., Souza, G., and Lands, W. E. M.,
A comparison of dietary fish oil and com oil in experimental colorectal carcinogenesis,
Nutr. Cancer, 11, 215, 1988.
128. Minoura, T., Takata, T., Sakaguchi, M., Takada, H., Yamamura, M., Hioki, K.,
and Yamamoto, M., Effect of dietary eicosapentaenoic acid on azoxymethane-induced
colon carcinogenesis in rats, Cancer Res., 48, 4790, 1988.
129. Reddy, B. S. and Maruyama, H., Effect of fish oil on azoxymethane-induced colon
carcinogenesis in male F344 rats, Cancer Res., 46, 3367, 1986.
266 Nutrition and Gene Expression
130. Abou-El-Ela, S. H., Prasse, K. W., Carroll, R., Wade, A. E., Dharwadkar, S., and
Bunce, 0. R., Eicosanoid synthesis in 7, 12-dimethylbenz(a)anthracene-induced mam-
mary carcinomas in Sprague-Dawley rats fed primrose, menhaden or com oil diets, Lipids,
23, 948, 1988.
131. Abou-El-Ela, S. H., Prasse, K. W., Farrell, R. L., Carroll, R. W., Wade, A. E.,
and Bunce, 0. R., Effects of D.L-2-difluoromethylomithine and indomethacin on mam-
mary tumor promotion in rats fed high n-3 and/or n-6 fat diets, Cancer Res., 49, 1434,
1989.
132. Locniskar, M., Belury, M. A., Cumberland, A. G., Patrick, K. E., and Fischer,
S. M., Lack of a protective effect of menhaden oil on skin tumor promotion by 12-0-
tetradecanoylphorbol-13-acetate, Carcinogenesis, 11, 1641, 1990.
133. Murphy, M.G., Dietary fatty acids and membrane protein function, J. Nutr. Biochem.,
I, 68, 1990.
134. Welsch, C. W., Enhancement of mammary tumorigenesis by dietary fat: review of
potential mechanisms, Am. J. Clin. Nutr., 45, 192, 1987.
135. Narisawa, T., Magadia, N. E., Weisburger, J. H., and Wynder, E. L., Promoting
effect of bile acids on colon carcinogenesis after intrarectal instillation of N-methyl-N '-
nitro-N-nitrosoguanidine in rats, J. Nat/. Cancer Inst., 53, 1093, 1974.
136. Reddy, B.S., Narisawa, T., Weisburger, J. H., and Wynder, E. L., Promoting effect
of sodium deoxycholate on colon adenocarcinomas in germ-free rats, J. Nat/. Cancer
Inst., 56, 441, 1976.
137. Reddy, B. S., Watanabe, K., Weisburger, J. H., and Wynder, E. L., Promoting
effect of bile acids in colon carcinogenesis in germ-free and conventional F344 rats,
Cancer Res., 37, 3238, 1977.
138. Reddy, B. S., Mangat, S., Sheinfil, A., Weisburger, J. H., and Wynder, E. L.,
Colon carcinogenesis. Effect of type and amount of dietary fat and 1,2-dimethylhydrazine
on biliary bile acids and neutral sterols in rats, Cancer Res, 37, 2132, 1977.
139. Glauert, H. P. and Bennink, M. R., Influence of diet or intrarectal bile acid injections
on colon epithelial cell proliferation in rats previously injected with I ,2-dimethylhydra-
zine, J. Nutr., 113,475, 1983.
140. Reddy, B. S., Hanson, D., Mangat, S., Mathews, L., Sbaschnig, M., Sharma, C.,
and Simi, B., Effect of high-fat, high-beef diet and of mode of cooking of beef in the
diet on fecal bacterial enzymes and fecal bile acids and neutral sterols, J. Nutr., 110,
1880, 1980.
141. Gallaher, D. D. and Franz, P. M., Effects of com oil and wheat brans on bile acid
metabolism in rats, J. Nutr., 120, 1320, 1990.
142. Deschner, E. E., Cohen, B. 1., and Raicht, R. F., Acute and chronic effect of dietary
cholic acid on colonic epithelial cell proliferation, Digestion, 21, 290, 1981.
143. Cohen, B. 1., Raicht, R. F., Deschner, E. E., Takahashi, M., Sarwal, A. N., and
Fazzini, E., Effect of cholic acid feeding on N-methyl-N-nitrosourea-induced colon
tumors and cell kinetics in rats, J. Nat/. Cancer Inst., 64, 573, 1980.
144. Skraastad, 0. and Reichelt, K. L., An endogenous colon mitosis inhibitor and dietary
calcium inhibit increased colonic cell proliferation induced by cholic acid, Scand. J.
Gastroenterol., 23, 801, 1988.
145. Wargovich, M. J., Eng, V. W. S., Newmark, H. L., and Bruce, W. R., Calcium
ameliorates the toxic effect of deoxycholic acid on colonic epithelium, Carcinogenesis,
4, 1205, 1983.
146. Wargovich, M. J, Eng, V. W. S., and Newmark, H. L., Calcium inhibits the damaging
and compensatory proliferative effects of fatty acids on mouse colon epithelium, Cancer
Lett., 23, 253, 1984.
147. Craven, P. A., Pfanstiel, J., and DeRubertis, F. R., Role of activation of protein
kinase C in the stimulation of colonic epithelial proliferation and reactive oxygen formation
by bile acids, J. Clin. Invest., 79, 532, 1987.
Glauert 267
148. Rosenthal, M.D., Fatty acid metabolism of isolated mammalian cells, Pro g. Lipid Res ..
26, 87, 1987.
149. Lands, W. E. M., Biosynthesis of prostaglandins, Annu. Rev. Nutr., II, 41, 1991.
150. Dyerberg, J., Linolenate-derived polyunsaturated fatty acids and prevention of athero-
sclerosis, Nutr. Rev., 44, 125, 1986.
151. Nicosia, S. and Patrono, C., Eicosanoid biosynthesis and action: novel opportunities
for pharmacological intervention, FASEB J., 3, 1941, 1989.
152. Fischer, S. M., Baldwin, J, K., Jasheway, D. W., Patrick, K. E., and Cameron,
G. S., Phorbol ester induction of 8-lipoxygenase in inbred SENCAR (SSIN) but not
C57BLI6J mice correlated with hyperplasia, edema, and oxidant generation but not or-
nithine decarboxylase induction, Cancer Res., 48, 658, 1988.
153. Fischer, S. M., Fiirstenberger, G., Marks, F., and Slaga, T. J., Events associated
with mouse skin tumor promotion with respect to arachidonic acid metabolism: a com-
parison between SENCAR and NMRI mice, Cancer Res., 47, 3174, 1987.
154. Denda, A., Ura, H., Tsujiuchi, T., Tsutsumi, M., Eimoto, H., Takashima, Y.,
Kitazawa, S., Kinugasa, T., and Konishi, Y., Possible involvement of arachidonic acid
metabolism in phenobarbital promotion of hepatocarcinogenesis, Carcinogenesis, 10,
1929, 1989.
!55. Reddy, B.S., Wang, C. X., and Maruyama, H., Effect of restricted caloric intake on
azoxymethane-induced colon tumor incidence in male F344 rats, Cancer Res., 47, 1226,
1987.
156. Klurfeld, D. M., Weber, M. M., and Kritchevsky, D., Inhibition of chemically induced
mammary and colon tumor promotion by caloric restriction in rats fed increased dietary
fat, Cancer Res., 47, 2759, 1987.
157. Boissonneault, G. A., Elson, C. E., and Pariza, M. W., Net energy effects of dietary
fat on chemically induced mammary carcinogenesis in F344 rats, J. Natl. Cancer lnst.,
76, 335' 1986.
158. Kritchevsky, D., Weber, M. M., and Klurfeld, D. M., Dietary fat versus caloric
content in initiation and promotion of 7, 12-dimethylbenz(a)anthracene-induced mammary
tumorigenesis in rats, Cancer Res., 44, 3174, 1984.
159. Welsch, C. W., House, J. L., Herr, B. L., Eliasberg, S. J., and Welsch, M. A.,
Enhancement of mammary carcinogenesis by high levels of dietary fat: a phenomenon
dependent on ad libitum feeding, J. Natl. Cancer lnst., 82, 1615, 1990.
160. Boissonneault, G. A., Calories and carcinogenesis: modulation by growth factors, in
Nutrition, Toxicity, and Cancer, Rowland, I. R., Ed., CRC Press, Boca Raton, FL,
1991' 413.
161. Snyder, D. L., Dietary Restriction and Aging, Alan R. Liss, New York, 1989.
162. Baldwin, S. and Parker, R. S., Effects of dietary fat level and aflatoxin B, treatment
on rat hepatic lipid composition, Food Chern. Toxicol., 23, 1049, 1985.
163. Neelands, P. J, and Clandinin, M. T., Diet fat influences liver plasma-membrane lipid
composition and glucagon-stimulated adenylate cyclase activity, Biochem. J., 212, 573,
1983.
164. Wade, A. E., Norred, W. P., and Evans, J. S., Lipids in drug detoxification, in
Nutrition and Drug Interrelations, Hathcock, J. N. and Coon, J., Eds., Academic Press,
New York, 1978, 475.
165. Hammer, C. T. and Wills, E. D., Dependence of the rate of metabolism of benzo(a)pyrene
on the fatty acid composition of the liver endoplasmic reticulum and on dietary lipids,
Nutr. Cancer, 2, 113, 1980.
166. Rutten, A. A. J. j. L. and Flake, H. E., Influence of high dietary levels of fat on rat
hepatic phase I and II biotransformation enzyme activities, Nutr. Rep. Int., 36, 109,
1987.
167. Cassano!, P., Bonnamour, D., Grolier, P., Pelissier, M.A., Amelizad, Z., Albrecht,
R., and Narbonne, J, F., The effect of dietary imbalances on the activation of
benzo[a]pyrene by the metabolizing enzymes from rat liver, Mutat. Res., 191,67, 1987.
268 Nutrition and Gene Expression
168. Kim, H. J., Choi, E. S., and Wade, A. E., Effect of dietary fat on the induction of
hepatic microsomal cytochrome P450 isozymes by phenobarbital, Biochem. Pharmacal.,
39, 1423, 1990.
169. Lam, T. C. L. and Wade, A. E., Influence of dietary lipid on the metabolism of
hexobarbital by the isolated, perfused rat liver, Pharmacology, 21, 64, 1980.
170. Lam, T. C. L. and Wade, A. E., Effect of dietary lipid on benzo(a)pyrene metabolism
by perfused rat liver, Drug Nutr. Interact., 1, 31, 1981.
171. Wade, A. E., Harley, W., and Bunce, 0. R., The effects of dietary com oil on the
metabolism and mutagenic activation of N-nitrosodimethylamine (DMN) by hepatic mi-
crosomes from male and female rats, Mutat. Res., 102, 113, 1982.
172. Clarke, S. D., Armstrong, M. K., and Jump, D. B., Dietary polyunsaturated fats
uniquely suppress rat liver fatty acid synthase and Sl4 mRNA content, J. Nutr., 120,
225, 1990.
173. Brannon, P.M., Adaptation of the exocrine pancreas to diet, Annu. Rev. Nutr., 10, 85,
1990.
174. Ribeiro, A., Mangeney, M., Cardot, P., Loriette, C., Chambaz, J., Rayssiguier,
Y., and Bereziat, G., Nutritional regulation of apolipoprotein genes: effect of dietary
carbohydrates and fatty acids, Adv. Exp. Med. Bioi., 285, 407, 1991.
175. Goodridge, A. G., Dietary regulation of gene expression: enzymes involved in carbo-
hydrate and lipid metabolism, Annu. Rev. Nutr., 7, 157, 1987.
176. Karmali, R. A., Chao, C. C., Basu, A., and Modak, M., II, Effect of n-3 and n-6
fatty acids on mammary H-ras expression and PGE-2 levels in DMBA-treated rats,
Anticancer Res., 9, 1169, 1989.
177. Ronai, Z., Lau, Y. Y., and Cohen, L.A., Dietary n-3 fatty acids do not affect induction
of Ha-ras mutations in mammary glands of NMU-treated rats, Mol. Carcinog., 4, 120,
1991.
178. Elliott, T. S., Swartz, D. A., Visek, W. J., and Kaput, J., Molecular analyses of
differences in gene expression due to changes in the level of dietary fat, FASEB J., 4,
A916, 1990.
179. Swartz, D. A., Visek, W. J., and Kaput, J., Genomic organization of a fat regulated
gene, FASEB J., 5, AI649, 1991.
Chapter 12
TABLE OF CONTENTS
III. The Gene for Acetyl CoA Carboxylase ...................... .... 272
0-8493-6961-4/93/$0.00 +$.50
© 1993 by CRC Press, Inc. 269
270 Nutrition and Gene Expression
I. OVERVIEW
0 Glucoae-G-P Triglyceride
6-P-Giucon?.:tone 1 l
Palmitate
~ Fruiae-8-P
Trloae-P
Malonyl Co A
Pyruvate
/
+
(5) -Malate
! 0l
Acetyl CoA
Oxaloaceta~e
Acetyl CoA @
[M]oU©©i'ml@ au ©hr o©liTl
Oxaloacetate ~ Citrate --+--+ Citrate
FIGURE l. Fatty acid biosynthesis from glucose. 2 Acetyl CoA carboxylase catalyzes the
formation of malonyl CoA (Reaction I) from acetyl CoA produced in cytosol by the action of
ATP-citrate lyase (Reaction 2). Conversion of malonyl CoA to palmitate is catalyzed by fatty
acid synthase (Reaction 3), using NADPH produced by glucose-6-phosphate dehydrogenase
(Reaction 4) and malic enzyme (Reaction 5).
and found RQ values less than 1, whereas the synthesis of fatty acids from
carbohydrate should give values greater than 1. A subsequent studyl 2 con-
firmed that after a single, high-carbohydrate meal equivalent to 479 g of
starch, nonprotein-RQ values did not exceed 1 over the next 8 h. The authors
concluded that the ingested carbohydrate was not converted to fat under these
conditions.
The question was recently addressed more directly by administering ace-
tate labeled with the nonradioactive, heavy isotope of carbon, 13C, to human
volunteers who were given either an infused or oral glucose load. 4 While it
was demonstrated that the administered [ 13C]acetate did label the liver cy-
tosolic acetyl CoA pool, surprisingly little isotope was found in fatty acids
of triglycerides of blood VLDL. It was calculated that less than 2% of the
VLDL palmitate and less than 1% of the VLDL stearate came from de novo
fatty acid biosynthesis. Thus, the authors concluded that the glucose load
failed to stimulate de novo fatty acid biosynthesis.
If this conclusion is extrapolated to the general case, it raises a number
of questions. Not the least of these is, if excessive carbohydrate consumption
does not increase de novo fatty acid synthesis and deposition, what happens
to the calories? Further, since feeding diets high in carbohydrate for extended
periods is known to stimulate fatty acid synthesis in the tissue of a number
of animal models, including rats, 13 • 14 mice, 15 pigs, 16 and even ruminants, 17
the lack of an effect of dietary carbohydrate would imply that humans have
a markedly different lipogenic regulatory system than that found in other
higher animals. It seems much more likely that the apparent lack of a car-
bohydrate-stimulated increase in lipogenesis in humans in these three
reports 4 • 11 • 12 was due to the lack of subject adaptation to an increased car-
bohydrate intake. That is, in each of these human studies, subjects were
allowed to consume their "normal" diet (presumably containing substantially
more fat and less carbohydrate than the test diet) prior to the experiment and
measurements were made in the few hours following a single carbohydrate
load. Numerous studies have made it clear that while some of the regulatory
mechanisms that control lipogenesis in animals occur very rapidly, others
require substantially more time. One such time-dependent, adaptive mecha-
nism is the regulation of the quantity of the active cytosolic form of acetyl
CoA carboxylase. Much of the remainder of this chapter is devoted to a
discussion of the time-dependent adaptive mechanisms which regulate acetyl
CoA carboxylase activity and thus lipogenesis.
probe to study the molecular biology of the rat enzyme. Using these tools in
subsequent experiments, overlapping e-DNA clones were found which co-
vered the entire coding sequence of rat acetyl CoA carboxylase mRNA. 20
The sequence of the coding section, consisting of 7035 bases, was used to
predict the primary structure of rat acetyl CoA carboxylase. By this analysis,
rat acetyl CoA carboxylase contained 2345 amino acids and had a molecular
weight of 265,220. Although the entire predicted primary structure of rat
acetyl Co A carboxylase has been published, 2° Figure 2 shows only selected
segments of the enzyme to illustrate some features which may be important
in its synthesis and regulation.
A e-DNA probe for chicken acetyl CoA carboxylase has also been
prepared 24 and used25 to determine the sequence of bases in the coding section
of DNA for acetyl CoA carboxylase mRNA, from which the primary amino
acid sequence of the chicken enzyme was deduced. Chicken acetyl CoA
carboxylase was predicted to contain 21 fewer amino acid residues than the
rat enzyme and have a molecular weight of 262,706. Not surprisingly, acetyl
CoA carboxylase from the two sources have very similar primary structures.
It is somewhat more surprising that both the rat2° and chicken 25 liver enzyme
exhibit a high degree of homology with carbamoyl phosphate synthetase at
the amino terminal end of the protein.
1 1
61 Leu·Gly·lle·Ser·Ala-Leu·Gln·Asp·Gly·Leu-Ala·Phe·His·Met·Arg·Ser·Ser-Met·Ser·Gly 80
1
81 Leu·His·Leu·Val·Lys·Gln·Gly·Arg·Asp-Arg·Lys-Lys·lle·Asp·Ser·Gln·Arg·Asp·Phe·Thr 100
Biotin
781 lle·Glu·Val-Met·Lys·Met·Val-Met·Thr·Leu·Thr·Ala·Val-Glu·Ser·Gly·Cys·lle·His·Tyr 800
1-----------Deletion------------1 1
1181 Phe·Met·Leu·Pro-Thr·Ser-His·Pro-Asn·Arg-Gly·Asn·lle-Pro·Thr·Leu·Asn-Arg-Met·Ser 1200
1
<:
::::
1201 Phe·Ala-Ser-Asn·Leu·Asn-His-Tyr-Gly·Met-Thr-His·Val·Ala·Ser·Val·Ser·Asp·Val-Leu 1220 3:.
g·
~
~
2340 CJ
2321 Met-Thr·Gln-His·lle-Ser·Pro-Thr-Gln·Arg-Ala·Glu-Val-Val·Arg-lle-Leu-Ser-Thr-Met
"'~
2341 Asp·Ser·Pro·Ser·Thr. 2345
~
"\:i
~
FIGURE 2. Selected segments of the primary structure of acetyl CoA carboxylase, based on the amino acid sequence predicted from analysis of e-DNA clones """"
of the enzymew The arrows ( t ) show the eight serine residues which can be phosphorylated. 21 The lysine residue in the indicated ( Biotin ) tetrapeptide is ~-
biotinylated in the holoenzyme'" 22 and the indicated octapeptide (----Deletion----) is deleted in a putative isozymic forrn of the enzyme."
Allred and Bowers 275
r-----.
/ll\~
Genomic DNA
FIGURE 3. Reported 27 exon-intron structure of genomic DNA coding for the 5' untranslated
region of acetyl CoA carboxylase, showing differential splicing of exons to produce heterogeneous
acetyl CoA carboxylase mRNA. The protein synthesis initiation codon (AUG) is located in exon
5. PI and P2 show the approximate locations of two putative promoters of transcription.
that mRNA coding for it must contain at least 7 kb, 20 it has been concluded
that the 10-kb band represented translatable acetyl CoA carboxylase mRNA
while the 3-kb band represented degradative products. 18
Although electrophoresis can separate species of mRNA which have sub-
stantial differences in molecular weight, detection of more subtle structural
differences require more sophisticated techniques. Using e-DNA probes com-
plementary to specific base sequences in acetyl CoA carboxylase mRNA for
Northern blot analysis and primer extension analysis, acetyl CoA carboxylase
mRNA has been examined in mammary gland from lactating rats 26 •27 and in
rat liver. 28 These analyses established that there was substantial heterogeneity
of the mRNA within the 10-kb band in both tissues. Five different species
of mRNA were found in rat mammary gland all of which had the same base
sequence in the coding region but differed in length and/or base sequence of
the 5' untranslated region. The coding region of genomic DNA for the 5'
untranslated portion of mRNA was found in five exons containing a total of
645 nucleotides, scattered over a 50-kb region. The authors concluded that
these heterogeneous species of mRNA occurred because of differential splic-
ing of the transcripts of these exons (Figure 3). For example, it was proposed
that the 5' untranslated region of the most abundant specie of mRNA in rat
276 Nutrition and Gene Expression
activities of promoters in both liver (Promoter I and II) and adipose tissue
(Promoter I) were depressed in streptozotocin-diabetic rats. Administration
of insulin rapidly increased Promoter I activity in adipose tissue but, sur-
prisingly, had no effect on the activity of either promoter in liver of the same
rats even after a period of 24 h where insulin was administered every 6 h.
It has been proposed 29 that acetyl CoA carboxylase mRNA species can
be classified as either class 1 (represented by the pAU type of mRNA in
Figure 3) or class 2 (represented by the FL56 type of mRNA in Figure 3),
depending upon whether they were produced in response to activation of
Promoter I or II, respectively. Activation of Promoter I in liver occurs only
under increased lipogenic conditions while Promoter II is active in liver of
chow fed rats as well as in other tissue, presumably to maintain a basal level
of acetyl CoA carboxylase. In fact, it has been suggested that Promoter II is
a "house-keeping" promoter. 30 This scenario implies that the presence of
two different promoters may be related to the dual function of acetyl CoA
carboxylase as discussed in previous sections. That is, the class 1 mRNA
transcript produced following the activation of Promoter I may be involved
in increased de novo synthesis of fatty acids destined for stored triglyceride
while the type of mRNA (class 2) produced from activation of Promoter II
may be involved in long chain fatty acyl CoA elongation and/or synthesis of
fatty acids for membrane lipids.
While this scenario represents an interesting hypothesis, it may be an
oversimplification because the reported29 promoter activity of mammary gland
and adipose tissue does not precisely fit. That is, since initiation of lactation
is accompanied by a substantial increase in de novo fatty acid biosynthesis
in the mammary gland, it might be expected on the basis of the proposed
model that Promoter I would become activated during lactation, but Promoter
I was not found to be active in mammary gland under any condition. Instead,
Promoter II activity increased during lactation. In adipose tissue, only Pro-
moter I (and not Promoter II) activity has been found, even under nonlipogenic
conditions. Further, the expression of Promoter II has been found to require
enhancer elements. 30 Enhancer sequences, which activate transcription, are
not usually found in "house-keeping" genes. 3°Finally, additional information
on Promoter II regulation has been obtained by the study of insulin-dependent
differentiation of 30A5 preadipocytes into adipocytes. 31 Differentiation of
these cells, which contain only Class 2 acetyl CoA carboxylase mRNA species
whose production is under the control of Promoter II, is accompanied by an
increased activity/quantity of acetyl CoA carboxylase and the concentration
of acetyl CoA carboxylase mRNA. 32 Insulin apparently activates Promoter
II, but such acute hormonal regulation of "house-keeping genes" would not
be expected.
Surprisingly, insulin induction of acetyl CoA carboxylase and cell dif-
ferentiation required prior exposure of the 30A5 preadipocytes to cyclic-
AMP. 31 Specific regions of genomic DNA were shown to be required for this
278 Nutrition and Gene Expression
cyclic-AMP effect. It is not clear that these results are relevant to the regulation
of acetyl CoA carboxylase mRNA production in cells of growing and adult
rats because hormonal effects during differentiation may be quite different
than their effects on differentiated cells. Semenkovich et al. 33 have shown
that the primary effect of insulin on lipoprotein lipase during the differentiation
of 3T3 preadipocytes into adipocytes was at the transcriptional level, but in
differentiated adipocytes, insulin had no effect on transcription. Rather, in
differentiated cells, exposure to insulin affected lipoprotein lipase production
only at the translational and posttranslational levels. Still, Swierczynski et
al. 34 showed that triiodothyroxine-stimulated accumulation of acetyl CoA
carboxylase mRNA in cultured chicken embryo hepatocytes, as well as in-
creased concentrations of mRNA of other lipogenic enzymes, was blocked
by protein kinase inhibitors. The authors concluded that ongoing protein
phosphorylation was specifically required for triiodothyroxine to stimulate
transcription of genes of lipogenic enzymes.
It is clear from these reports that regulation of acetyl CoA carboxylase
mRNA synthesis is not simple. Understanding of this complex process has
been made even more difficult by evidence that there are isozymic forms of
acetyl CoA carboxylase.
band after SDS-PAGE, indicating the presence of two forms of the enzyme
which differ in subunit molecular weight. That observation has been con-
firmed by Thampy 37 who concluded that the two forms were isozymes on the
basis of antibody reactivity and tissue distribution. More recently, Bianchi et
al. 38 also concluded that these two subunit molecular weight forms of acetyl
CoA carboxylase represent isozymes of the enzyme, based on differences in
binding by two different monoclonal antibody preparations. It was reported
that while liver contained both subunit forms, heart and skeletal muscle con-
tained only the larger subunit form and adipose tissue contained only the
smaller form.
While the reports by Thampy, 37 Kong et al., 23 and Bianchi et al. 38 all
concluded that there are isozymic forms of acetyl CoA carboxylase, a more
detailed analysis of the observations shows very little agreement on how the
putative isozymes differ from each other. First, our observations 36 indicated
that the two high molecular weight forms of acetyl CoA carboxylase in liver
cytosol differed in mass by about 13,000 daltons (13 kDa) while Bianchi et
al. 38 and Thampy 37 estimated a 15 kDa difference. In contrast, the mass of
isozymes with and without the 8 amino acids as shown in Figure 2 would
differ by less than 1 kDa, an amount too small to be detected by SDS-PAGE
in such a large protein. Second, tissue distribution of the isozymic forms were
markedly different. For example, while Bianchi et al. 38 found only one iso-
zymic form in rat adipose tissue, Kong et al. 23 reported that adipocytes con-
tained substantial amounts of mRNA for both isozymes. Finally, the mono-
clonal antibodies used by Bianchi et al. 38 to differentiate the two isozymic
forms of the enzyme were raised against synthetic peptides containing only
the 15 amino acids in theN terminal or C terminal fragments of acetyl CoA
carboxylase where the amino acid composition of the two isozymes reported
by Kong et al. 23 do not differ (Figure 2). In spite of the lack of agreement
on specifics, the available evidence does suggest that rat tissues contain
isozymic forms of acetyl CoA carboxylase which may have different tissue
distribution and regulatory properties.
Although it is clear that there are multiple forms of acetyl Co A carboxy lase
mRNA in tissue, much work remains to be done before the physiological
relevance of them can be understood. In the case of the species of acetyl Co A
carboxylase mRNA which differ only in the 5' untranslated region, translation
of each of them would produce enzymes with identical primary structure.
Since translation of mRNA occurs from the 5' to 3' direction, these 5' regions
are upstream from the coding section. In fact, the protein synthesis initiation
codon, AUG, is located within exon 5 (Figure 3). The function of the 5'
280 Nutrition and Gene Expression
untranslated region of mRNA is not known but it is possible that the length
and base sequence of this section, while not affecting amino acid sequence
of the protein, may affect interaction with ribosomes. In the case of acetyl
CoA carboxylase mRNA species, differences in the 5' untranslated region
were found to affect translation in an in vitro rabbit reticulocyte protein
synthesis assay. 29 With that system, translation was about I 0 times better with
the pAU type of mRNA than with the FL56 type.
If this difference in translation efficiency occurs in vivo, the cell could
effectively control the rate of translation simply by regulating relative amounts
of different types of mRNA produced. Further, from an investigative view-
point, this would mean that experiments to determine the relative importance
of transcriptional vs. posttranscriptional processes in the control of enzyme
synthesis are difficult to interpret unless relative amounts of acetyl CoA
carboxylase mRNA differing in the 5' untranslated region are determined.
That is, a multifold increase in the quantity of class 2 type of acetyl CoA
carboxylase mRNA might produce no more enzyme protein per unit of time
than would be obtained from a much smaller increase in the quantity of class
l type of acetyl CoA carboxylase mRNA. Unfortunately, simple Northern
blot analysis which is most often used for determining dietary/hormonal effects
on acetyl CoA carboxylase mRNA levels does not differentiate between class
l and class 2 types of mRN A.
Similarly, simple Northern blot analysis does not differentiate between
the two acetyl CoA carboxylase mRNAs which code for isozymic forms of
the enzyme. Yet, the relative amounts of the two forms differ among tissues
and their relative concentrations vary under different physiological condi-
tions. 23 The ratio of the two isozymic forms of acetyl CoA carboxylase may
be very important because their regulatory properties differ. It has been shown
that deletion of the eight amino acids from the larger form of acetyl CoA
carboxylase allowed cyclic-AMP-dependent protein kinase to catalyze the
phosphorylation of the serine residue located at position 1200 which makes
the enzyme more dependent upon citrate for activation. 23
CoA carboxylase mRNA was not detectable in liver of fasted (48 h) rats, 39
Batenburg and Whitsett40 reported that fasting (48 h) resulted in only a small,
statistically insignificant decrease in rat liver acetyl CoA carboxylase mRNA
relative to that in liver from fed control animals. Unfortunately, Batenburg
and Whitsett40 did not report changes in acetyl CoA carboxylase quantity or
activity in response to fasting, but Pape et al. 39 reported that fasting rats for
48 h resulted in a 96% decrease in both acetyl CoA carboxylase quantity and
activity in liver relative to that in liver of fed control rats. Thus, it is highly
unlikely that the level of acetyl CoA carboxylase mRNA regulates enzyme
synthesis during fasting, but the data in these reports are compatible with the
hypothesis that refeeding increases transcription of the gene for acetyl CoA
carboxylase, although neither report indicated which of the multiple forms of
acetyl CoA carboxylase mRNA were affected.
Data in these reports 39 •40 showed that both acetyl CoA carboxylase mRNA
and enzyme activity/quantity were increased after rats were refed a diet high
in carbohydrate. In each case, however, acetyl CoA carboxylase mRNA was
measured only at a single point in time (72 h) after refeeding. Such an
experiment does not indicate whether the rise in acetyl CoA carboxylase
activity/quantity was due to increased acetyl CoA carboxylase mRNA. The
relative importance of each potential regulatory mechanism can better be
assessed by determining the time-dependent relationships of transcription rate,
acetyl CoA carboxylase mRNA levels, and quantity of active enzyme. When
Katsurada et al. 41 conducted such an experiment, the results (Figure 4) were
not consistent with the hypothesis of simple transcriptional control. They
found that the rate of acetyl CoA carboxylase mRNA synthesis increased
significantly (p< 0.05) within an hour after refeeding a fat-free, high car-
bohydrate diet and remained at a similarly high rate until 8 h after refeeding.
Meanwhile, acetyl CoA carboxylase mRNA concentration steadily increased
over the first 8 h and then remained relatively constant through 24 h of
refeeding. Over the next 24 and 48 h, acetyl CoA carboxylase mRNA con-
centration steadily dropped to reach a level at 72 h after refeeding that was
significantly (p< 0.05) below the 8- to 24-h plateau. In contrast, cytosolic
acetyl CoA carboxylase activity (and quantity as judged by immunoreactive
protein) increased at a much slower rate than the rate of accumulation of
mRNA and did not reach a plateau until 48 h after refeeding. It should be
noted that at a point in time (72 h) after refeeding, both acetyl CoA carboxylase
mRNA and acetyl CoA carboxylase activity/quantity were elevated relative
to that in liver of fasted rats, in agreement with the results of Pape et al. 39
and Batenburg and Whitsett, 40 but measurement of changes in these parameters
over time leads to a different conclusion. That is, since there was a time lag
of several hours between measurable increased acetyl CoA carboxylase mRNA
and the increased enzyme activity/quantity, the data are not consistent with
the hypothesis that formation of cytosolic enzyme is limited simply by acetyl
CoA carboxylase mRNA template availability. Rather, posttranscriptional
Allred and Bowers 283
11 ~--------------------------~
. .
I 1.~. I.
'
+
lt . . . , ..······--···
fl
1 ..
T••">-,,
~ ',.l
..
I r: I
I
,.. ...~
..~
l
0 Ww~~~~~~~~~~~~~~
0
0 8 18
Refeecllng Time (Hrs)
FIGURE 4. Effect of refeeding a high carbohydrate diet on acetyl CoA carboxylase activity,
mRNA synthesis rate, and mRNA levels in rat liver, adapted from Katsurada et al. 41 Enzyme
activity was shown to be directly proportional to quantity of immunoprecipitated enzyme.
While the studies described in the preceding paragraphs dealt with intact
rats, a number of investigations on hormonal regulation of acetyl CoA car-
boxylase have been conducted with primary cultures of hepatocytes. Use of
such cell cultures has an advantage over studies with intact animals in that
the substrate and hormonal milieu of the cells can be controlled, but have the
disadvantage that it is not certain whether results from the use of these systems
accurately reflect in vivo regulatory mechanisms. With that caveat, it appears
that insulin stimulates the synthesis of acetyl CoA carboxylase in cultured rat
liver hepatocytes. Giffhorn and Katz42 found that the activity of acetyl CoA
carboxylase per mg DNA was increased 24 h after insulin and glucose were
added to cultured hepatocytes. Acetyl CoA carboxylase synthesis, as measured
by incorporation of [35 S]-methionine into immunoprecipitated enzyme, was
also stimulated by a combination of insulin (.01 J.1M) and glucose (20 mM).
Spence et a!Y conducted a more in-depth study of regulation of acetyl
CoA carboxylase in cultured rat hepatocytes in which they determined the
time-dependent changes in acetyl CoA carboxylase activity and synthesis
rates. Enzyme activity was measured44 by a [' 4 C]-bicarbonate fixation assay
in cytosol after preincubation with 20 mM citrate for 30 min at 37°. Protein
synthesis rates were determined at each time point by measuring the amount
of [3H]-leucine in 20 min into protein precipitated by polyclonal antiacetyl
CoA carboxylase antibody. Enzyme activity began to increase immediately
after insulin was added and reached a plateau after 6 h, but the rate of protein
synthesis increased much more slowly (Figure 5). Although the quantity of
acetyl CoA carboxylase was not measured in this study, preincubation of the
enzyme preparation with citrate has been found by others 39 to negate the
effects of any short-term regulatory mechanisms, in which case activity is
directly proportional to quantity. Therefore, it is likely that the increase in
acetyl CoA carboxylase activity well before synthesis of the enzyme was
increased indicates that the initial effects of insulin were on posttranslational
processes rather than transcription or translation.
Instead of measuring acetyl CoA carboxylase mRNA by Northern blot,
Spence et al. 43 determined time-dependent changes in acetyl CoA carboxylase
mRNA template activity. Template activity ("translatable acetyl CoA car-
boxylase mRNA' ') was measured at each time point by determining the
relative synthesis rates of immunoprecipitated acetyl CoA carboxylase vs.
synthesis rates of total soluble protein in a rabbit reticulocyte-lysate assay,
using total RNA extracted from hepatocytes as template. Following insulin
exposure, the acetyl CoA carboxylase mRNA activity increased at a sub-
stantially slower rate than enzyme activity but at a considerably faster rate
than enzyme synthesis (Figure 5). The temporal relationship between enzyme
synthesis rates and acetyl Co A carboxylase mRNA template activity was much
closer in cultured hepatocytes (Figure 5) than that found between enzyme
quantity and acetyl CoA carboxylase mRNA determined by Northern blot
analysis in liver of intact rats (Figure 4). Although this may be due, at least
Allred and Bowers 285
5 5
- "':"
4 4
ic
.11
·--
. _. ··-
••
0
.I•.J
lJoo
:;:~-
.!! 3 3
·-·
.. 1,
.I II
:sc:c 0
2 2 ~~--
~ II II.~
:loo:loo II:
.i ~
Ill
1 1
E
I
0 0
0 2 4 8 8 10
lnaulln Expoaure (Hra)
FIGURE 5. Activity, synthesis, and mRNA template activity of acetyl CoA carboxylase over
time after exposure of cultured rat hepatocytes to insulin, adapted from Spence et at"' Activity
is very likely directly proportional to enzyme quantity since it was detem1ined by the bicarbonate
fixation assay after preincubation with 20 mM citrate. Protein synthesis rates were determined
at each time point by measuring the amount of [-'H)-leucine in 20 min into protein precipitated
by polyclonal antiacetyl CoA carboxylase antibody. Template activity of mRNA was measured
at each time point by determining the relative synthesis rates of immunoprecipitated acetyl CoA
carboxylase vs. synthesis rates of total soluble protein in a rabbit reticulocyte-lysate assay, using
total RNA extracted from hepatocytes as template.
in part, to the fact that these are two different systems, it could also be argued
that measurement of acetyl CoA carboxylase mRNA template activity gives
a more accurate assessment of the relative importance of transcription and
translation in regulating the synthesis of acetyl CoA carboxylase. Unlike
results from standard Northern blot analysis, measurement of acetyl CoA
carboxylase mRNA template activity would reflect both the quantity of mRNA
and variations in translation efficiency of the heterogeneous forms of acetyl
CoA carboxylase mRNA. Thus, the data in Figure 5 could be interpreted to
mean that acetyl CoA carboxylase synthesis over several hours may be reg-
ulated by an insulin effect, at least in part, at the transcriptional level, but
again there was a substantial time lag between the rise in acetyl CoA car-
boxylase mRNA and increased acetyl CoA carboxylase synthesis.
Using similar primary cultures of rat hepatocytes, Spence et al. 43 also
made the interesting observation that incubation of cultured cells with 25 rnM
286 Nutrition and Gene Expression
fructose (but not glucose). in the absence of insulin, had an effect on enzyme
activity, rates of enzyme synthesis, and acetyl CoA carboxylase mRNA tem-
plate activity almost identical to the insulin response, except for an initial 2-
h time lag following fructose addition before any of the three parameters
changed. Combination experiments showed that insulin and fructose effects
on all three parameters were additive. The results of this study indicate that
effects of fructose, like insulin, can be observed in primary cultures of rat
hepatocytes on both transcription and posttranscriptional processes. Studies
with intact rats have similarly indicated that feeding diets high in fructose
(but not glucose) to streptozotocin-diabetic rats had almost the same effect
on acetyl CoA carboxylase activity/quantity, 4 I.43 acetyl CoA carboxylase mRNA
template activity, 43 acetyl CoA carboxylase mRNA concentration, 41 and acetyl
CoA carboxylase synthesis rates 41 in liver as insulin injection. The mechanism
by which dietary fructose affects these parameters has not been established.
In addition to fructose, the effects of other dietary components on reg-
ulation of acetyl CoA carboxylase have been studied. Katsurada et a!. 45 com-
pared the effects in liver on activity of lipogenic enzymes, including acetyl
CoA carboxylase, after fasted rats were refed a diet containing 85% sucrose
without protein vs. rats refed a diet composed of 67% sucrose/18% casein.
In each case, the remainder of the diet was composed of cellulose (9.9%),
vitamins, and minerals. Acetyl CoA carboxylase activity increased at the
same rate for the first 2 days of refeeding and remained at this high level
(about sixfold greater than that of fasted rats) until day 4 regardless of protein
content of the diet. In contrast, maximal increased activity of fatty acid
synthase, glucose-6-phosphate dehydrogenase and malic enzyme was ob-
served only when the diet contained protein. At the end of the 4 days, [3 H]-
leucine was injected 2 h before rats were killed, and radioactivity in immu-
noprecipitated cytosolic enzymes was determined as a measure of enzyme
synthesis rates. While the presence or absence of dietary protein had no effect
on acetyl CoA carboxylase synthesis rates, the rates of synthesis of both
glucose-6-phosphate dehydrogenase and malic enzyme were significantly lower
(p< 0.01) in liver of rats refed the protein-free diet. The levels of mRNA
template activity for glucose-6-phosphate dehydrogenase and malic enzyme45
were unaffected by dietary protein, indicating that dietary protein affected
the synthesis of these two enzymes at posttranscriptional level. Subsequent
experiments, 41 in which fasted rats were refed these identical diets, confirmed
that the presence or absence of dietary protein had no effect on either the
increase in acetyl CoA carboxylase activity/quantity, acetyl CoA carboxylase
mRNA synthesis rates, or acetyl CoA carboxylase mRNA concentration.
The effects of dietary triglycerides on acetyl CoA carboxylase depend
upon the chain length and degree of unsaturation of the fatty acids present.
For example, after rats were fed a diet in which 32% of total calories were
derived from medium chain triglycerides (predominant fatty acids = c8:0 and
C 10,0 ) for 21 d, acetyl CoA carboxylase activity in liver was not different than
Allred and Bowers 287
that in liver of rats fed a high carbohydrate (corn starch), low fat (I% corn
oil) diet. 46 In contrast, when an equivalent amount of energy was derived
from com oil, liver acetyl CoA carboxylase activity was reduced by a factor
of 2.
Other studies have shown that dietary polyunsaturated fatty acids are
much more effective than saturated or monounsaturated fatty acids in de-
pressing lipogenesis and acetyl Co A carboxylase activity. For example, dietary
safflower oil had a greater inhibitory effect on rat liver acetyl CoA carboxylase
activity than tallow47 or palmitate. 47.4 8 Emken et al. 49 compared the effect of
feeding various long chain fatty acids to mice on the activity in liver of acetyl
CoA carboxylase and other lipogenic enzymes. They found that linoleate
dramatically decreased the activity of all of the lipogenic enzymes, including
acetyl CoA carboxylase, but neither palmitate nor stearate had any effect on
the activity of acetyl CoA carboxylase or any of the other lipogenic enzymes.
The mechanism by which polyunsaturated fatty acids decrease acetyl CoA
carboxylase activity remains unknown although Katsurada et al. 41 reported
that when rats were fed a diet containing 57% sucrose, 18% casein, and 10%
com oil, acetyl CoA carboxylase activity, acetyl CoA carboxylase mRNA
synthesis, and acetyl CoA carboxylase mRNA concentration in liver were all
significantly lower (p< 0.001) than when rats were fed a diet containing 67%
sucrose and 18% casein but no fat. While these data might suggest that dietary
corn oil affected transcription, time-dependent changes in these parameters
were not reported. The authors suggested that dietary polyunsaturated fatty
acids might influence insulin secretion or insulin binding to cellular receptors.
Salati et al., 50 who studied the effects of long chain fatty acids on induction
of acetyl CoA carboxylase and other lipogenic enzymes in rat hepatocyte
cultures, similarly concluded that fatty acids might affect insulin interaction
with the hepatocytes. It should be noted, however, that the effect of feeding
long chain fatty acids to rats is different than the effects observed when
primary cultures of hepatocytes are exposed to them. When Salati and Clarke51
determined the effects of adding either 0.2 mM palmitate, oleate, linoleate,
or arachidonate to the media of primary cultures of rat hepatocytes, they found
that each of them caused a small (30 to 35%) albeit statistically significant
reduction in acetyl CoA carboxylase activity. But, in contrast to results from
studies with intact rats, the effect did not vary with either chain length or
degree of unsaturation.
Witters et a!. 52 published the first report in recent years which suggested
that there may be particulate forms of acetyl CoA carboxylase. They found
substantial acetyl CoA carboxylase activity was precipitated when rat liver
288 Nutrition and Gene Expression
again with no change in the total quantity of acetyl CoA carboxylase. The
study of acute (3 d) alloxan-diabetic rats 59 indicated that mobilization/acti-
vation of mitochondrial forms of acetyl CoA carboxylase was insulin depen-
dent. Examination of the subcellular distribution of acetyl CoA carboxylase
in the liver of Zucker rats 60 indicated that obese rats had no more enzyme
than that of lean litter mates, but rather more of the enzyme was in the active
cytosolic form.
Evidence of particulate forms of two other lipogenic enzymes which are
normally active in cytosol has been found. A form of ATP-citrate lyase has
been shown to be associated with mitochondria. 61 The distribution of this
enzyme between cytosol and mitochondria is dietary dependent. 62 More re-
cently, it was concluded63 that epidermal growth factor increased the activity
of glucose-6-phosphate dehydrogenase of rat renal cortical cells by causing
its release from a particulate form within the cell into the cytosol.
Although the mechanism by which acetyl CoA carboxylase might be
bound to the outer mitochondrial membrane has not been established, several
enzymes have now been shown to be attached to the cellular plasma membrane
via a glycosylated phosphatidyl inositol linkage. 64 The discovery that acetyl
CoA carboxylase is a glycoprotein65 ·66 raises the possibility that it may be
attached to the mitochondrial membrane by a similar mechanism. Analysis
of the carbohydrates present in purified acetyl CoA carboxylase by GC-MS
confirmed a previous report 67 that the enzyme contained inositol and estab-
lished that it also contained mannose and galactose, as well as trace amounts
of N-acetyl glucosamine, fucose, and 6-deoxy mannose. It was calculated
that the combined mass of the carbohydrates amounted to about 6 kDa. While
glycosylation is most often associated with exported proteins, 64 it has been
established that another enzyme intimately involved in lipid metabolism,
HMG CoA reductase, is a glycoprotein. 68 HMG CoA reductase remains within
the cell in which it is produced and, unlike exported glycoproteins, is not
processed in the Golgi apparatus. 69
Ifrat liver contains mitochondrial storage forms of acetyl CoA carboxylase
which can be mobilized under lipogenic conditions, it could provide a mech-
anism by which cells could more rapidly respond to dietary/hormonal changes
than de novo enzyme synthesis might allow. Such a mechanism could account
for the rapid rise in enzyme quantity implied by the data in Figure 5 well
before acetyl CoA carboxylase synthesis is increased after cultured rat he-
patocytes are exposed to insulin. In this scenario, insulin stimulation of acetyl
CoA carboxylase transcription and translation would still be necessary in
order to replace mobilized mitochondrial enzyme. While the existence of
storage forms of acetyl CoA carboxylase represents a plausible hypothesis,
definitive proof of the physiological significance of particulate forms of the
enzyme has not been obtained.
290 Nutrition and Gene Expression
IX. BIOTINYLATION
XI. CONCLUSION
It is clear that dietary/hormonal regulation of acetyl CoA carboxylase
ultimately invovles gene expression. It is equally apparent, however, that
regulation of the enzyme is remarkably complex. The available data are not
compatible with the hypothesis that the quantity of active forms of the cytosolic
enzyme is solely dependent upon the rate of gene transcription. Rather, di-
etary/hormonal regulation of acetyl CoA carboxylase can occur at the tran-
scriptional, translational, and posttranslationallevels.
REFERENCES
I. Volpe, J. J. and Vagelos, P.R., Mechanisms and regulation of biosynthesis of saturated
fatty acids, Physiol. Rev.. 56, 339, 1976.
2. Gibson, D. M., Lyons, R. T., Scott, D. F., and Muto, Y., Synthesis and degradation
of the lipogenic enzymes of rat liver, Adv. Enzyme Regul., 10, 187, 1970.
3. Brady, L., Regulation of hepatic camitine palmitoyltransferase, Nutrition and Gene
Expression, CRC Press, Boca Raton, FL, 1992.
4. Hellerstein, M. K., Christiansen, M., Kaempfer, S., Kletke, C., Wu, K., Reid,
J, S., Mulligan, K., Hellerstein, N. S., and Shackleton, C. H. L., Measurement of
de novo hepatic lipogenesis in humans using stable isotopes, J. Clin. Invest., 87, 1841,
1991.
5. Mead, J. F., Alfin-Siater, R. B., Howton, D. R., and Popjak, G., Lipids: Chemistry,
Biochemistry and Nutrition, Plenum Press, New York, 1986, chaps. 8 and 9.
6. Needleman, P., Turk, J., Jakschik, B. A., Morrison, A. R., and Lefkowith, J. B.,
Arachidonic acid metabolism, Annu. Rev. Biochem., 55, 69, 1986.
7. Bijleveld, C., Vaartjes, W. J., and Geelen, M. J,, Time course of hormonal effects
on acetyl-CoA carboxylase as measured in digitonin-permeabilized rat hepatocytes, Horm.
Metab. Res., 21,602,1989.
8. Milatovich, A., Plattner, R., Heerema, N. A., Palmer, C. G., Lopez-Casillas, F.,
and Kim, K.-H., Localization of the gene for acetyl-CoA carboxylase to human chro-
mosome 17, Cytogenet. Cell. Genet., 48, 190, 1988.
9. Blom, W., DeMuinck-Keizer, S.M. P. F., and Scholte, H. R., Acetyl CoA c.arboxylase
deficiency: an inborn error of de novo fatty acid synthesis, New Engl. J. Med., 305, 465,
1981.
292 Nutrition and Gene Expression
10. Shrago, E., Spennetta, T., and Gordon, E., Fatty acid synthesis in human adipose
tissue, J. Bioi. Chern., 244, 2761, 1969.
II. Passmore, R. and Swindells, Y. E., Observations on the respiratory quotients and weight
gain of man after eating large quantities of carbohydrate, Brit. J. Nutr., 17, 331, 1963.
12. Acheson, K. J,, Flatt, J, P., and Jequier, E., Glycogen synthesis versus lipogenesis
after a 500 gram carbohydrate meal in man, Metabolism, 31, 1234, 1982.
13. Tepperman, H. M. and Tepperman, J,, Adaptive hyperlipogenesis, Fed. Proc., 23,
73, 1964.
14. Nakanishi, S. and Numa, S., Purification of rat liver acetyl CoA carboxylase and
immunochemical studies on its synthesis and degradation, Eur. J. Biochem., 16, 161,
1970.
15. Buckley, M. G. and Rath, E. A., Regulation of fatty acid synthesis and malonyl-CoA
content in mouse brown adipose tissue in response to cold-exposure, starvation or re-
feeding, Biochem. J., 243, 437, 1987.
16. Mersmann, H. J, and Koong, L.-J., Effect of plane of nutrition on adipose tissue
metabolism in genetically obese and lean pigs, J. Nutr., 114, 862, 1986.
17. Smith, S. B., Prior, R. L., Ferrell, C. L., and Mersmann, H. J,, Interrelationships
among diet, age, fat deposition and lipid metabolism in growing steers, J. Nutr., 114,
153, 1984.
18. Lopez-Casillas, F., Pape, M. E., Bai, D. H., Kuhn, D. N., Dixon, J, E., and Kim,
K. H., Preparation of functional acetyl-CoA carboxylase mRNA from rat mammary
gland, Arch. Biochem. Biophys., 257, 63, 1987.
19. Bai, D. H., Pape, M. E., Lopez-Casillas, F., Luo, X. C., Dixon, J, E., and Kim,
K. H., Molecular cloning of eDNA for acetyl-coenzyme A carboxylase, J. Bioi. Chern.,
261, 12395, 1986.
20. Lopez-Casillas, F., Bai, D. H., Luo, X. C., Kong, I. S., Hermodson, M. A., and
Kim, K. H., Structure of the coding sequence and primary amino acid sequence of acetyl-
coenzyme A carboxylase, Proc. Nat/. Acad. Sci. U.S.A., 85, 5784, 1988.
21. Davies, S. P., Sim, A. T., and Hardie, D. G., Location and function of three sites
phosphorylated on rat acetyl-CoA carboxylase by the AMP-activated protein kinase, Eur.
J. Biochem., 187, 183, 1990.
22. Bai, D. H., Moon, T. W., Lopez-Casillas, F., Andrews, P. C., and Kim, K. H.,
Analysis of the biotin-binding site on acetyl-CoA carboxylase from rat, Eur. J. Biochem.,
182, 239, 1989.
23. Kong, I. S., Lopez-Casillas, F., and Kim, K. H., Acetyl-CoA carboxylase mRNA
species with or without inhibitory coding sequence for Ser-1200 phosphorylation, J. Bioi.
Chern., 265, 13695, 1990.
24. Takai, T., Wada, K., and Tanabe, T., Primary structure of the biotin-binding site of
chicken liver acetyl-CoA carboxylase, FEBS. Lett., 212, 98, 1987.
25. Takai, T., Yokoyama, C., Wada, K., and Tanabe, T., Primary structure of chicken
liver acetyl-CoA carboxylase deduced from eDNA sequence, J. Bioi. Chern., 263, 2651,
1988.
26. Lopez-Casillas, F., Luo, X. C., Kong, I. S., and Kim, K. H., Characterization of
different forms of rat mammary gland acetyl-coenzyme A carboxylase mRNA: analysis
of heterogeneity in the 5' end, Gene, 83,311, 1989.
27. Luo, X. C., Park, K., Lopez-Casillas, F., and Kim, K. H., Structural features of the
acetyl-CoA carboxylase gene: mechanisms for the generation of mRNAs with 5' end
heterogeneity, Proc. Nat/. Acad. Sci. U.S.A., 86, 4042, 1989.
28. Lopez-Casillas, F. and Kim, K. H., Heterogeneity at the 5' end of rat acetyl-coenzyme
A carboxylase mRNA. Lipogenic conditions enhance synthesis of a unique mRNA in
liver, J. Bioi. Chern., 264, 7176, 1989.
29. Lopez-Casillas, F., Ponce-Castaneda, M. V., and Kim, K. H., In vivo regulation of
the activity of the two promoters of the rat acetyl coenzyme-A carboxylase gene, En-
docrinology, 129, 1049, 1991.
Allred and Bowers 293
30. Luo, X. C. and Kim, K. H., An enhancer element in the house-keeping promoter for
acetyl-CoA carboxylase gene. Nucleic Acids Res., 18, 3249, 1990.
31. Park, K. and Kim, K. H., Regulation of acetyl-CoA carboxylase gene expression:
Insulin induction of acetyl-CoA carboxylase and differentiation of 30A5 preadipocytes
require prior cAMP action on the gene, J. Bioi. Chern., 266. 12249, 1991.
32. Pape, M. E. and Kim, K. H., Transcriptional regulation of acetyl coenzyme A car-
boxylase gene expression by tumor necrosis factor in 30A-5 preadipocytes, Mol. Cell.
Bioi., 9, 974,1989.
33. Semenkovich, C. F., Wims, M., Noe, L., Etienne, J., and Chan, L., Insulin regulation
of lipoprotein lipase activity in 3T3-Ll adipocytes is mediated at the posttranscriptional
and posttranslationallevels, J. Bioi. Chern., 264. 9030, 1989.
34. Swierczynski, J., Mitchell, D. A., Reinhold, D. S., Salati, L. M., Stapleton, S. R.,
Klautky, S. A., Struve, A. E., and Goodridge, A. G., Triiodothyronine-induced ac-
cumulations of malic enzyme, fatty acid synthase, acetyl-coenzyme A carboxylase, and
their mRNAs are blocked by protein kinase inhibitors. Transcription is the affected step,
J. Bioi. Chern., 266, 17459, 1991.
35. Allred, J. B., Harris, G. J., and Goodson, J. Regulation of purified rat liver acetyl
CoA carboxylase by phosphorylation, J. Lipid Res., 24, 449, 1983.
36. Goodson, J., Pope, T. S., and Allred, J. B., Molecular weights of subunits of acetyl
CoA carboxylase in rat liver cytoplasm. Biochern. Biophys. Res. Cornrnun., 122, 694.
1984.
37. Thampy, K. G., Formation of malonyl coenzyme A in rat heart. Identification and
purification of an isozyme of acetyl CoA carboxylase from rat heart, J. Bioi. Chern.,
264, 17631, 1989.
38. Bianchi, A., Evans, J. L., Iverson, A. J., Nordlund, A. C., Watts, T. D., and
Witters, L. A., Identification of an isozymic form of acetyl-CoA carboxylase, J. Bioi.
Chern., 265, 1502, 1990.
39. Pape, M. E., Lopez-Casillas, F., and Kim, K. H., Physiological regulation of acetyl-
CoA carboxylase gene expression: effects of diet, diabetes, and lactation on acetyl-CoA
carboxylase mRNA, Arch. Biochern. Biophys., 267, 104, 1988.
40. Batenburg, J. J. and Whitsett, J. A., Levels of mRNAs coding for lipogenic enzymes
in rat lung upon fasting and refeeding and during perinatal development, Biochirn. Bio-
phys. Acta., 1006, 329, 1989.
41. Katsurada, A., Iritani, N., Fukuda, H., Matsumura, Y., Nishimoto, N., Noguchi,
T., and Tanaka, T., Effects of nutrients and hormones on transcriptional and post-
transcriptional regulation of acetyl-CoA carboxylase in rat liver, Eur. J. Biochern., 190,
435, 1990.
42. Giftlmrn, S. and Katz, N. R., Glucose-dependent induction of acetyl-CoA carboxylase
in rat hepatocyte cultures, Biochern. J., 221, 343, 1984.
43. Spence, J. T., Koudelka, A. P., and Tseng-Crank, J. C., Role of protein synthesis
in the carbohydrate-induced changes in the activities of acetyl CoA carboxylase and
hydroxymethylglutaryl CoA reductase in cultured rat hepatocytes, Biochern. J., 227, 939,
1985.
44. Craig, M. C., Dugn, R. E., Muesing, R. A., Slakey, L. L., and Porter, J. W.,
Comparative effects of dietary regimen on the levels of enzymes regulating the synthesis
of fatty acids and cholesterol in rat liver, Arch. Biochern. Biophys., 151, 128, 1972.
45. Katsurada, A., Iritani, N., Fukuda, H., Noguchi, T., and Tanaka, T., Effects of
dietary nutrients on lipogenic enzyme and mRNA activities in rat liver during induction,
Biochirn. Biophys. Acta., 877, 350, 1986.
46. Chanez, M., Bois-Joyeux, B., Arnaud, M. J., and Peret, J., Metabolic effects in rats
of a diet with a moderate level of medium-chain triglycerides, J. Nutr., 121, 585, 1991.
47. Wilson, M. D., Hays, R. D., and Clarke, S. D., Inhibition of liver lipogenesis by
dietary polyunsaturated fat in severely diabetic rats, J. Nutr., 116, 1511, 1986.
294 Nutrition and Gene Expression
48. Clarke, S. D., Wilson, M. D., and Ibnoughazala, T., Resistance of lung fatty acid
synthesis to inhibition by dietary fat in the meal-fed rat, J. Nutr., 114, 598, 1984.
49. Emken, E. A., Abraham, S., and Lin, C. Y., Metabolism of cis-12-octadecenoic acid
and trans-9.trans-12-octadecadienoic acid and their influence on lipogenic enzyme activ-
ities in mouse liver. Biochim. Biophys. Acta., 919. Ill, 1987.
50. Salati, L. M., Adkins-Finke, B., and Clarke, S. D., Free fatty acid inhibition of the
insulin induction of glucose-6-phosphate dehydrogenase in rat hepatocyte monolayers,
Lipids, 23, 36. 1988.
51. Salati, L. M. and Clarke, S. D., Fatty acid inhibition of hormonal induction of acetyl-
coenzyme A carboxylase in hepatocyte monolayers. Arch. Biochem. Biophys.. 246, 82,
1986.
52. Witters, L. A., Friedman, S. A., and Bacon, G. W., Microsomal acetyl CoA car-
boxylase: evidence for association of enzyme polymer with liver microsomes. Proc. Nat/.
Acad. Sci. U.S.A., 78, 3639, 1981.
53. Imesch, E., Wolczunowicz, M., and Rous, S., Enzymatic activities of cytoplasmic and
of microsomal acetyl CoA carboxylase of rat epididymal adipose tissue; different regu-
latory effects of a short-term fast and palmitoyl CoA on these two enzymes, Int. J.
Biochem., 15, 977, 1983.
54. Evans, J. L. and Witters, L.A., Quantitation by immunoblotting of the in vivo induction
and subcellular distribution of hepatic acetyl-CoA carboxylase, Arch. Biochem. Biophys.,
264, 103, 1988.
55. Iverson, A. J., Bianchi, A., Nordlund, A. C., and Witters, L. A., Immunological
analysis of acetyl-CoA carboxylase mass, tissue distribution and subunit composition,
Biochem. J., 269, 365, 1990.
56. Roman-Lopez, C. R., Goodson, J., and Allred, J. B., Determination of the quantity
of acetyl CoA carboxylase by [14C]methyl avidin binding, J. Lipid Res., 28, 599, 1987.
57. Allred, J, B. and Roman-Lopez, C. R., Enzymatically inactive forms of acetyl-CoA
carboxylase in rat liver mitochondria, Biochem. J., 251, 881, 1988.
58. Roman-Lopez, C. R., Shriver, B. J., Joseph, C. R., and Allred, J. B., Mitochondrial
acetyl-CoA carboxylase: time course of mobilization/activation in liver of refed rats,
Biochem. J., 260, 927, 1989.
59. Roman-Lopez, C. R. and Allred, J. B., Acute alloxan diabetes alters the activity but
not the total quantity of acetyl CoA carboxylase in rat liver, J. Nutr., 117, 1976, 1987.
60. Allred, J. B., Roman-Lopez, C. R., Jurin, R. R., and McCune, S. A., Mitochondrial
storage forms of acetyl CoA carboxylase: mobilization/activation accounts for increased
activity of the enzyme in liver of genetically obese Zucker rats, J. Nutr., 119, 478, 1989.
61. Janski, A.M. and Cornell, N. W., Association of ATP citrate lysase with mitochondria,
Biochem. Biophys. Res. Commun., 92, 305, 1980.
62. Cornell, N. W., Janski, A.M., and Rendon, A., Compartmentation of enzymes: ATP
citrate lyase in hepatocytes from fed and fasted rats, Fed. Proc., 44, 2448, 1985.
63. Stanton, R. C., Seifters, J. L., Boxer, D. C., Zimmerman, E., and Cantley, L. C.,
Rapid release of bound glucose-6-phosphate dehydrogenase by growth factors. Correlation
with increased enzymatic activity, J. Bioi. Chern., 266, 12442, 1991.
64. Low, M. G., Biochemistry of the glycosyl-phosphatidylinositol membrane protein an-
chors, Biochem. J., 244, I, 1987.
65. Bowers, D. F. and Allred, J. B., Acetyl CoA carboxylase is a glycoprotein, FASEB
J., 5, A462, 1991.
66. Bowers, D. F., Identification and Characterization of Acetyl CoA Carboxylase as a
Glycoprotein, Ph.D. dissertation, The Ohio State University, Columbus, 1991.
67. Heger, H. W. and Peter, H. W., Phosphatidylinositol as an essential constituent of the
acetyl CoA carboxylase from rat liver, Int. J. Biochem., 8, 841, 1977.
Allred and Bowers 295
68. Liscum, L., Cummings, R. D., Anderson, R. G. W., DeMartino, G. N., Goldstein,
J. L., and Brown, M.S., 3-Hydroxy-3-methylglutaryl CoA reductase: a transmembrane
glycoprotein of the endoplasmic reticulum with N-linked "high-mannose" oligosac-
charide, Proc. Nat/. Acad. Sci. U.S.A .. 80, 7165, 1983.
69. Haro, D., Marrero, P. F., Ayte, J,, and Hegardt, F. G., Identification of a cholesterol-
regulated 180-kDa microsomal protein in rat hepatocytes, Eur. 1. Biochern .. 188, 123,
1990.
70. Moss, J, and Lane, M. D., The biotin-dependent enzymes, Adv. Enzyrnol .. 35, 321,
1971.
71. Shenoy, B. C., Paranjape, S., Murtif, V. L., Kumar, G. K., Samols, D., and Wood,
H. G., Effect of mutations at met-88 and met-90 on the biotinylation of lys-89 of the
apo 1.3S subunit of transcarboxylase, FASEB J., 2, 2505, 1988.
72. Lamhonwah, A.-M., Quan, F., and Gravel, R. A., Sequence homology around the
biotin-binding site of human propionyl CoA carboxylase and pyruvate carboxylase, Arch.
Biochern. Biophys.. 254, 631, 1987.
73. Carlson, C. A. and Kim, K.-H., Regulation of hepatic acetyl CoA carboxylase by
phosphorylation and dephosphorylation, 1. Bioi. Chern., 248, 378, 1973.
74. Allred, J, B. and Roehrig, K. L., Inhibition of hepatic lipogenesis by cyclic-3', 5'-
nucleotide monophosphate, Biochern. Biophys. Res. Cornrnun., 46, 1135, 1972.
75. Allred, J, B. and Roehrig, K. L., Metabolic oscillations and food intake, Fed. Proc ..
32, 1727, 1973.
76. Allred, J, B. and Roehrig, K. L., Inhibition of rat liver acetyl CoA carboxylase in
N6, 0 2-dibutyryl cyclic-3', 5' -adenosine monophosphate in vitro, J. Bioi. Chern., 248,
4131, 1973.
77. Haystead, T. A., Moore, F., Cohen, P., and Hardie, D. G., Roles of the AMP-
activated and cyclic-AMP-dependent protein kinases in the adrenaline-induced inacti-
vation of acetyl-CoA carboxylase in rat adipocytes, Eur. J. Biochern., 187, 199, 1990.
78. Thampy, K. G. and Wakil, S. J,, Regulation of acetyl-coenzyme A carboxylase. II.
Effect of fasting and refeeding on the activity, phosphate content, and aggregation state
of the enzyme, J. Bioi. Chern., 263, 6454, 1988.
Chapter 13
Nicholas 0. Davidson
TABLE OF CONTENTS
0·8493-6961-4/93/$0.00 +$.50
© 1993 by CRC Pre». 1nc. 297
298 Nutrition and Gene Expression
biology that DNA encodes an RNA template which is identical and which
subsequently specifies a predictable protein. Over the last few years, several
examples of RNA editing have been described in lower eukaryotes, and these
have been classified according to the underlying mechanism (for detailed
reviews, see References 18 and 19). These mechanisms include the posttran-
scriptional insertion or deletion of uridine residues in trypanosome mito-
chondrial genes which results in the production of a translationally competent
open reading frame. This form of RNA editing is of interest also because it
is mediated by small guide RNAs. 20 Other forms of RNA editing include the
posttranscriptional insertion of guanosine residues in paramyxovirus and cy-
tidine residues in physarum polycephalum. These examples are discussed in
detail in the reviews cited. 18 • 19
Apo B mRNA editing was described in 1987 when several groups si-
multaneously reported the site-specific modification of apo B mRNA as the
underlying mechansim for the production of distinct isoforms of the protein
from the human liver and small intestine. 21 . 23 Powell et al. 21 prepared intestinal
cDNAs and demonstrated that nucleotide 6666 in human and rabbit intestinal
apo B eDNA was changed from the genomically temp1ated cytidine to a
uridine residue. This change modified codon 2153 from a CAA which encodes
glutamine to UAA which specifies an in-frame stop codon. Chen et aJ.2 2
reported the identical conclusion but used a strategy consisting of tryptic
fragment analysis and direct RNA sequencing. These findings indicated that
intestinal apo B was the product of a single apo B gene in which codon 2153
is altered to produce a translational stop codon and thereby specify a truncated
apo B (apo B48) as the primary translation product. Recently, a second
example of mammalian RNA editing was described in which certain subunits
of the brain glutamate receptor transcripts undergo a single base change in
which a glutamine codon (CAG) is changed to an arginine codon (CGG). 24
The significance of the observation that both forms of RNA editing involve
a glutamine codon may be fortuitous since the base involved is different
(cytidine vs. adenosine) although the mechanism may involve a deamination
in both instances. The consequences following editing of the brain glutamate
receptor transcripts are profound since the arginine residue regulates the func-
tional characteristics of these gated receptor channels. Thus, in both situations
currently known of mammalian RNA editing, the process has enormous func-
tional consequences for gene expression. It is likely that an understanding of
RNA editing mechanisms and their evolution will provide important insight
into a novel area of regulation of gene expression and will reveal new functions
for this process.
in apo B mRNA editing is distinct for the liver and small intestine with a
striking increase in intestinal editing prenatally while the hepatic transcript is
largely unedited until postnatal day 20. Thus, the emergence of edited apo
B mRNA in both the rat liver and intestine appears to coincide with devel-
opmental changes in triglyceride metabolism. Parallel conclusions cannot be
proposed for either human or porcine intestine since fetal hepatic and intestinal
triglyceride metabolism have yet to be characterized in these species. In regard
to the observation that apo B mRNA editing is developmentally induced in
the fetal small intestine, studies conducted in our laboratory have addressed
the question of whether apo B mRNA editing changes in the adult as enter-
ocytes migrate up the villus during their 3- to 5-day life span. Using adult
rats, sequential enterocyte isolation from villus tip to crypt was undertaken
by citrate-EDTA chelation and RNA extracted from isolated cells whose
position was inferred from sucrase activity enrichment (Figure 1). Apo B
mRNA editing was determined by reverse-transcription and coupled poly-
merase chain reaction amplification of apo B eDNA followed by single nu-
cleotide discrimination using differential oligonucleotide hybridization. As
can be seen in Figure I, the small intestinal enterocyte edits apo B mRNA
as soon as it is transcribed in cells emerging from the crypt.
As a model of the developing fetal small intestine, workers have used a
human colon cancer-derived cell line (Caco-2) which, in culture, undergoes
a form of' 'spontaneous differentiation'' and displays certain phenotypic char-
acteristics of developing enterocytes. Teng et al. 28 used this cell line to study
apo B mRNA editing and found that during the course of differentiation from
pre- to late postconfluency, apo B mRNA abundance increased 20-fold but
the proportions of edited to unedited transcript remained unaltered at less than
5% at all times studied. Thus, in this cell line apo B mRNA abundance
appears to be regulated by mechanisms distinct from those which influence
apo B mRNA editing. Other investigators 36 using this cell line have found
that apo B mRNA editing increases when the cells are grown on semiperme-
able filters rather than plastic. The specific cues which mediate this response
are presently unknown but may provide insight into the mechanisms under-
lying developmental changes in human small intestinal apo B mRNA editing.
•
68
•• • •
71
•• • •
36
•• •
TAA
••
CAA
•
Ileum Sucrase TAA CAA
••
••
6
••
4
•• "
FIGURE 1. Gradients of apo B mRNA editing in rat small intestine. Isolated rat enterocytes
were prepared from both jejunum and ileum by citrate-EDTA chelation with estimates of the
relative position of each fraction on the villus-crypt axis determined by sucrase-specific activity.
RNA was prepared from each fraction and subjected to reverse-transcription and polymerase
chain reaction amplification of apo B eDNA followed by differential oligonucleotide hybridization
to detect the single nucleotide substitution. It is evident that intestinal apo B mRNA is extensively
edited in all regions of the small intestine with no horizontal or vertical gradient detectable.
thentic uridine 5' -monophosphate indicates strongly that the process involves
a cytosine deaminase. The question of where in the cell this modification
occurs was addressed in an imaginative series of experiments from Lau et
a!. 41 These workers addressed this issue using the rat liver which is known
to contain both edited and unedited apo B mRNA. The authors first prepared
nuclear and cytoplasmic RNA and demonstrated that nuclear RNA contained
less than 20% edited apo B mRNA while cytoplasmic apo B mRNA-either
total or polysomal-contained approximately 60% edited species. Lau et al.~ 1
then went on to fractionate nuclear RNA into poly A+ and poly A- RNA in
order to ascertain the relationship between polyadenylation and editing of apo
B mRNA. They found that poly A+ RNA contained approximately 50% edited
apo B mRNA while poly A- RNA contained only 10% edited species. These
workers then identified an intron in the rat apo B genomic DNA sequence
5% to the edited base in a similar position to that reported for the human apo
B gene. Using a primer downstream of the edited base and another primer
at the 3' terminus of the upstream exon 25, Lau et al. 41 were able to use the
polymerase chain reaction to amplify approximately 3 kb of apo B flanking
the intron-exon junction in both genomic DNA and unspliced RNA. Splicing
was shown to remove the 500-bp intron, and thus the amplification of a 2.5-
kb fragment could be distinguished from the 3-kb fragment alluded to above.
These workers demonstrated that unspliced nuclear pre-RNA contained less
than 10% edited apo B mRNA and that unspliced poly A- RNA contained
essentially no edited apo B mRNA. Thus, the conclusion from these exper-
iments is that apo B mRNA editing is not a cotranscriptional process, but
occurs predominantly in the nucleus and is completed by the time the RNA
is spliced and polyadenylated. These findings have important implications
for the identity and characteristics of the postulated editing machinery which
should presumably be nuclear, although it is well recognized that passage of
proteins and other macromolecules occurs through nuclear pores, which thus
does not preclude the possibility of the editing machinery originating in the
cytoplasmic compartment. The topology of apo B mRNA editing in other cell
types, particularly the small intestine, has not been evaluated to date, at least
in part because of difficulties associated with the preparation of intestinal
nuclei and intact nuclear RNA.
the changes in apo B mRNA editing in both the rat and human small intestine
precede any developmental increase in fetal thyroid hormone activity and
maternal T3 does not cross the placenta. Additionally in the adult rat small
intestine, there was no change in apo B mRNA editing following changes in
thyroid hormone status. 25
above. It was found that apo A-1 and A-IV mRNA abundance and protein
biosynthesis were increased approximately two- to fourfold in the animals
refed a high carbohydrate diet for 48 h. These changes are thus of a lesser
magnitude but in the same direction as encountered42 with T3 treatment. The
time course of these changes has not been fully characterized since the earliest
time point examined was 24 h after exposure to a high-carbohydrate diet and
the experiment only continued up to 48 h of refeeding. Thus, important
questions still remain, such as the earliest point that a response to carbohydrate
is detectable and whether equivalent responses can be obtained with different
carbohydrate sources such as fructose and glucose. Additionally, it will be
of interest to examine longer periods of exposure to a high-carbohydrate diet
to determine whether the liver is capable of effectively unloading the accu-
mulated lipid after sustained intake of this modified isocaloric diet. Finally
it will be important to evaluate the synthesis of nascent intrahepatic lipopro-
teins by isolating Golgi VLDLs and examining their morphology, lipid con-
tent, and apo B isomorph distribution. These studies are currently underway
in the laboratory.
A. METHODOLOGICAL CONSIDERATIONS
The major question for investigators examining the biology of apo B
mRNA editing is the underlying mechanism of this interesting process. As
indicated above the most likely explanation is a site-specific deamination of
the cytosine at nucleotide 6666, but the basis for this conclusion was attained
experimentally as a result of a technical advance pioneered by Driscoll et
al., 51 which described an in vitro system to examine apo B mRNA editing.
This sytem used cytoplasmic S 100 supernatants prepared from MeA 7777
cells, a rat hepatoma line which synthesizes and secretes both apo BlOO and
apo B48 and which therefore provides a ready source of "editing activity"
to examine possible in vitro RNA modification. Driscoll et al. 51 incubated
MeA 7777 extracts with various lengths of synthetic apo B RNA prepared
from the region flanking the edited base. In order to detect the single nucleotide
change, these workers devised an ingenious scheme whereby an antisense
apo B oligonucleotide (35-mer) is annealed downstream from the edited base
at position 6666 and extended with reverse transcriptase in the presence of
dATP, dCTP, dTTP, and dideoxy-GTP as a chain terminator. There being no
other "C" residues between the 5' end of the antisense oligonucleotide and
the edited base, the primer is extended until it reaches the first upstream "C".
If nucleotide 6666 is unedited the primer undergoes chain termination and
produces an extension product of 42 bp. If nucleotide 6666 is edited to a
"U" then the primer extends to the next upstream "C", at 6655 in human
apo B eDNA and 6661 in rat and mouse apo B eDNA. The extended and
terminated primers are resolved effectively by urea-acrylamide electropho-
resis. In the original description by Driscoll et al., 51 in vitro editing was
demonstrated with S I00 extracts from MeA 7777 cells, but the efficiency of
editing was less than 5% of the input RNA. The reasons for the low activity
are of interest since endogenous apo B mRNA from these cells is edited
approximately 20%. One possibility is that subtle sequence differences be-
tween human and rat apo B eDNA may be of importance (see below and
Figure 2). There may be additional factors, however, since an intriguing
observation is that MeA 7777 cells transfected with a human full-length apo
B eDNA edit a substantial proportion of the human apo B mRNA (greater
than 50%) while the endogenous transcript is edited less than half of this
value. 52 ·53 Thus issues relating to differential accessibility and transcript sta-
bility require consideration. The relatively low level of in vitro apo B mRNA
editing observed with MeA 7777 cells has been observed by others in nuclear
extracts prepared from rat liver. 41 The question arises as to whether this
particular source (i.e., rat hepatic or hepatoma-derived nuclear or SIOO ex-
tract) contains relatively low levels of editing activity or whether the puri-
fication scheme used is inappropriate. Investigators using rat liver nuclear
extracts to examine in vitro editing have had to optimize their methods for
310 Nutrition and Gene Expression
64 82 6522
Human CCCACAGCAAGCTAATGATTATCTGAATTCATTCAATTGGGAGAGACAAGTTTCACATGC
Pig ------------T--------------G-A------G------A----------TGAG---
Rabbit -----------T--------------G-A-------------------------CAG---
Rat T--T-----GAT-C--------------G---CTG-C--------------AG-TGG---
Mouse T--T-----GAT-C--C-----------G---CTG-C--------------AG-TGG---
6542 6582
Human CAAGGAGAAACTGACTGCTCTCACAAAAAAGTATAGAATTACAGAAAATGATATACAAAT
Pig ---AA------AT-G--A-T---TGG--G-T---------------------G---G---
Rabbit ------------A---A--T----------T----A---------G-------------C
Rat ------A---T-A---T--T---TGG----C--------------T------G---T---
Mouse ------A---A-A---T--T---TGG----T--------------T------G---T---
6602 6642
Human TGCATTAGATGATGCCAAAATCAACTTTAATGAAAAACTATCTCAACTGCAGACATATAT
Pig ---------CA--------------C--------------A----G--A--A------G-
Rabbit ------G---A----------------A-----------G--------T---------G-
Rat ---C------AG---------------G-----------C--------TG-------CGC
Mouse ---CA-----AG---------------C-----------C--------TG-------CGC
6662 6702
Human GATACAATTTGATCAGTATATTAAAGATAGTTATGATTTACATGATTTGAAAATAGCTAT
Pig -----------------------------A------------------T--G-C------
Rabbit -----------------------------A--T----C----------T-----------
Rat -----------------------G-----A-------GC---G--C--A----G-A----
Mouse -----------------------------A-------CC------C--A----G-A----
6722 6762
Human TGCTAATATTATTGATGAAATCATTGAAAAATTAAAAAGTCTTG
Pig -----GG-------------------C--C---G----T-----
Rabbit A----G------A---C-------G-------------T-----
Rat ----C-G---------AG------------GC------TG----
Mouse ----G-G---------CG------------G-------TG----
2152 2182
Human IQFOQYIK05YOLHOLKIAIANIIOEIIEKLK5L
Pig ---------N-----F-T---R------AT--I-
Rabbit ---------NF----F-----5---Q-M----I-
Rat -------R-N--AQ---RT--0---R------M-
Mouse ---------N--P----RT--E---R------M-
FIGURE 2. (A) Aligned nucleotide sequence of apo B eDNA flanking the edited base. Four
mammalian apo B cDNAs are contrasted to the human sequence with identity represented by a
hyphen. 30 (B) Aligned amino acid sequence deduced from four mammalian apo B cDNAs in
comparison to human apo B flanking the edited base. The single Jetter code is employed for
amino acid identity. 30
reliable detection of low levels of apo B mRNA editing which in some cases
are less than 1 to 2% of input RNA. 54 Studies from a number of labs indicate
that the yield and functional activity of small intestinal S 100 extracts is
considerably higher than that reported for the rat liver. Using isolated rabbit, 40
rat, 55 or baboon56 enterocyte S100 extracts, workers have reported in vitro
Davidson 311
conversion rates of~ 10 to 40% of input RNA. Whether this editing activity
is developmentally induced in the small intestine awaits further study. There
appear to be some inconsistencies between different reports concerning the
optimal conditions for in vitro editing with the original report describing a
cryolabile activity which required 50 mM EDTA. 51 Later reports have found
a number of differences from the earlier study, and most recently Greeve et
al. 55 have detailed extensive findings in regard to the characterization of rat
enterocyte SlOO editing activity. The major conclusions emerging from this
and other reports is that the editing activity is most likely a protein with no
cofactor or additional RNA requirements. Attempts to purify this editing
activity from crude enterocyte S 100 extracts have resulted in severalfold
enrichment, but no consensus has emerged as to its apparent molecular mass.
Driscoll has suggested that baboon enterocyte S 100 extracts contain an editing
activity of approximately 125 kDa. 56 Another controversial aspect of the in
vitro editing methodology is whether apo B mRNA assembles into a higher
order molecular complex following incubation with a source of editing ac-
tivity. Following an initial report of this phenomenon, 5 7 others55 have failed
to confirm the existence of a so-called ''editosome' '. Distinct from the editing
activity, there appears to be an apo B mRNA binding activity in rat liver
nuclear extracts. 5 8 This binding activity has an apparent molecular weight of
40 kDa following UV cross-linking and specifically binds a 26-nucleotide
apo B RNA fragment spanning the edited base. It will be of interest to
investigate the relationship of this apparent RNA binding protein to the editing
machinery by way of mutagenesis studies in which editing is abolished.
Species considerations may be relevant in this regard also, since studies59
from our laboratory have indicated that chick small intestinal enterocytes
contain an editing enhancing factor(s), and studies are currently underway to
purify and compare these factor(s) to the binding activity found in rat tissue.
from MeA 7777 cells. Thus it is likely that there are true biological differences
in editing activity within different cell types which may, additionally, be more
or less preserved during the preparation of S 100 extracts. In regard to the
nucleotide specificity for apo B mRNA editing, recent studies have provided
some important advances. Chen et al. 54 used a coupled transcription editing
reaction to demonstrate that in a series of mutations in a 9-nucleotide sequence
flanking the edited base, in only 2 of 22 such mutants (a transversion mutant
changing the wild type ATACAATIT to ATTCT ATIT and a deletion mutant
ATAC-TTTT) was editing abolished. The authors contend that this indicates
a relatively lax sequence specificity for apo B mRNA editing. An important
limitation of this study was the low level of editing demonstrated by rat liver
nuclear extracts which was less than 2% of the input apo B RNA. In order
to investigate whether more distant sequence information conferred important
specificity Shah et a!. 61 systematically mutagenized a 55-bp region of human
apo B eDNA and demonstrated that the minimal sequence information required
for in vitro editing by rat enterocyte S 100 extracts is contained with a 26-
nucleotide stretch flanking the edited base. However, these workers found
an 11-nucleotide region downstream of position 6666 in which all but one
point mutations abolished editing. This result is of interest since recent work
from our laboratory 59 has demonstrated that chick apo B mRNA is not edited
in vivo by small intestinal enterocytes, and that the chick apo B eDNA
sequence is divergent from mammalian apo B cDNAs with 3 changes from
the conserved mammalian sequence noted in the 11-nucleotide cassette de-
scribed by Shah et alY These changes would be predicted to abolish in vitro
apo B RNA editing according to the data of Shah et a!., and, indeed, no
editing could be detected when chick apo B RNA was incubated in vitro with
S 100 extracts prepared from rat enterocytes while human, rat, and porcine
apo B RNAs were all efficiently edited. A further point of interest in regard
to the sequence specificity of apo B RNA editing concerns the demonstration
that a second edited base is detectable in human apo B RNA at position 6802
which changes threonine (ACA) to isoleucine (AUA) at codon 2198. 62 This
is of no functional consequence since the transcript is terminated upstream
of this nucleotide in all instances examined. The finding is of interest in view
of the sequence homology flanking both edited sites, and it reinforces the
hypothesis that the requisite information for editing is contained within the
26 nucleotides flanking position 6666.
ACKNOWLEDGMENTS
Work cited from the author's laboratory was supported by Grants HL-
38180, HL K0-4 02166 from the NHLBI and DK-42086 from NIDDK. I
would like to recognize the contributions of BaBie Teng, Charles Baum, Ruth
Carlos, and Annalise Hausman to the work presented in this review. This
chapter is dedicated to my father, Jack Davidson, M.B. Ch.B., on the occasion
of his 75th birthday.
REFERENCES
1. Chan, L., VanTuinen, P., Ledbetter, D. H., Daiger, S. P., Gotto, A.M., and Chen,
S. H., The human apolipoprotein B-100 gene: a highly polymorphic gene that maps to
the short arm of chromosone 2, Biochem. Biophys. Res. Comm.• 133, 248, 1985.
2. Mehrabian, M., Sparkes, R. S., Mohandas, T., Klisak, I. J., Schumaker, V. N.,
Heinzmann, C., Zollman, S., Ma, Y., and Lusis, A. J., Human apolipoprotein B:
chromosmal mapping and DNA polymorphisms of hepatic and intestinal species, Somatic
Cell Mol. Genet., 12. 245, 1986.
3. Luo, C. C., Li, W. H., Moore, M. N., and Chan, L., Structure and evolution of the
apolipoprotein multigene family, J. Mol. Bioi., 187, 325, 1986.
4. Li, W. H., Tanimura, M., Luo, C. C., Datta, S., and Chan, L., The apolipoprotein
multigene family: biosynthesis, structure, structure-function relationships, and evolution,
J. Lipid Res.. 29, 245, 1988.
5. Knott, T. J., Pease, R. J., Powell, L. M., Wallis, S. C., Rail, S. C., Innerarity,
T. L., Blackhart, D., Taylor, W. H., Marcel, Y., Milne, R., Johnson, D., Fuller,
M., Lusis, A. J., McCarthy, B. J., Mahley, R. W., Levy-Wilson, B., and Scott, J.,
Complete protein sequence and identification of structural domains of human apolipo-
protein B, Nature (London), 323, 734, 1986.
6. Yang, C. Y., Chen, S. H., Gianturco, S. H., Bradley, W. A., Sparrow, J. T.,
Tanimura, M., Li, W. H., Sparrow, D. A., DeLoof, H., Roseneu, M., Lee, F. S.,
Gu, Z. W., Gotto, A.M., and Chan, L., Sequence, structure, receptor-binding domains
and internal repeats of human apolipoprotein B-100. Nature (London), 323, 738, 1986.
7. Chen, S. H., Yang, C. Y., Chen, P. F., Setzer, D., Tanimura, M., Li, W. H., Gotto,
A. M., and Chan, L., The complete eDNA and amino acid sequence of human apoli-
poprotein B-100, J. Bioi. Chern., 261, 12918, 1986.
8. Cladaras, C., Hadzopoulou-Cladaras, M., Nolte, R. T., Atkinson, D., and Zannis,
V. I., The complete sequence and structural analysis of human apolipoprotein B-100:
relationship between apoB-100 and apoB-48 forms, EMBO J., 5, 3495, 1986.
9. Ludwig, E. H., Blackhart, B. D., Pierotti, V. R., Caiati, L., Fortier, C., Knott, T.,
Scott, J., Mahley, R. W., Levy-Wilson, B., and McCarthy, B. J., DNA sequencing
of the human apolipoprotein B gene, DNA, 6, 363, 1987.
314 Nutrition and Gene Expression
10. Innerarity, T. L., Young, S. G., Poksay, K. S., Mahley, R. W., Smith, R. S., Milne,
R. W., Marcel, Y. L., and Weisgraber, K. H., Structural relationship of human apo-
lipoprotein B48 to apolipoprotein BIOO, J. Clin. Invest., 80, 1794, 1987.
11. Hardman, D. A., Protter, A. A., Chen, G. C., Schilling, J. W., Sato, K. Y., Lau,
K., Yamanaka, M., Mikita, T., Miller, J., Crisp, T., McEnroe, G., Scarborough,
R. M., and Kane, J. P., Structural comparison of human apolipoproteins B-48 and B-
100, Biochemistry, 26, 5478, 1987.
12. Yang, C. Y., Kim, T. W., Weng, S. A., Lee, B., Yang, M., and Gotto, A. M., Jr.,
Isolation and characterization of sulfhydryl and disulfide peptides of human apolipoprotein
B-100, Proc. Nat!. Acad. Sci. U.S.A., 87, 5523, 1990.
13. Law, A. and Scott, J., A cross-species comparison of the apo1ipoprotein B domain that
binds to the LDL receptor, J. Lipid Res., 31, 1109, 1990.
14. Pease, R. J., Milne, R. W., Jessup, W. K., Law, A., Provost, P., Fruchart, J. C.,
Dean, R. T., Marcel, Y. L., and Scott, J., Use of bacterial expression cloning to
localize the epitopes for a series of monoclonal antibodies against apolipoprotein B-1 00,
J. Bioi. Chern., 265, 553, 1990.
15. Herz, J., Hamann, U., Rogne, S., Myklebost, 0., Gausepohl, H., and Stanley,
K. K., Surface location and high affinity for calcium of a 500-kd liver membrane protein
closely related to the LDL-receptor suggest a physiological role as lipoprotein receptor,
EMBO J., 7, 4119, 1988.
16. Kowal, R. C., Herz, J., Goldstein, J. L., Esser, V., and Brown, M.S., Low density
lipoprotein receptor-related protein mediates uptake of cholesterol esters derived from
apoprotein E-enriched lipoproteins, Proc. Nat!. Acad. Sci. U.S.A., 86, 5810, 1989.
17. Beisegel, U., Weber, W., Ihrke, G., Herz, J., and Stanley, K. K., The LDL-receptor-
related protein, LRP, is an apolipoprotein E-binding protein, Nature (London), 341, 162,
1989.
18. Cattaneo, R., Different types of messenger RNA editing, Annu. Rev. Genet., 25, 71,
1991.
19. Sollner-Webb, B., RNA editing, Curr. Op. Cell Bioi., 3, 1056, 1991.
20. Blum, B., Bakalara, N., and Simpson, L., A model for RNA editing in kinetoplastid
mitochondria: "Guide" RNA molecules transcribed from maxicircle DNA provide the
edited information, Cell, 60, 189, 1990.
21. Powell, L. M., Wallis, S.C., Pease, R. J., Edwards, Y. H., Knott, T. J., and Scott,
J., A novel form of tissue-specific RNA processing produces apolipoprotein B48 in
intestine, Cell. 50, 831, 1987.
22. Chen, S. H., Habib, G., Yang, C. Y., Gu, Z. W., Lee, B. R., Weng, S., Silberman,
S. R., Cai, S. J., Deslypere, J. P., Rosseneu, M., Gotto, A. M., Li, W. H., and
Chan, L., Apolipoprotein B-48 is the product of a messenger RNA with an organ-specific
in-frame stop codon, Science, 238, 363, 1987.
23. Hospattanker, A. V., Higuchi, K., Law, S. W., Meglin, N., and Brewer, H. B.,
Identification of a novel in-frame translational stop codon in human intestine ApoB
mRNA, Biochem. Biophys. Res. Commun., 148, 279, 1987.
24. Sommer, B., Kohler, M., Sprengel, R., and Seeburg, P. H., RNA editing in brain
controls a determinant of ion flow in glutamate-gated channels, Cell, 67, II, 1991.
25. Davidson, N. 0., Powell, L. M., Wallis, S. C., and Scott, J., Thyroid hormone
modulates the introduction of a stop codon in rat liver apolipoprotein B messenger RNA,
J. Bioi. Chern., 263, 13482, 1988.
26. Tennyson, G. E., Sabatos, C. A., Higuchi, K., Meglin, N., Brewer, H. B., Expression
of apolipoprotein B mRNAs encoding higher- and lower-molecular weight isoproteins in
rat liver and intestine, Proc. Nat[. Acad. Sci. U.S.A., 86, 500, 1989.
Davidson 315
27. Higuchi, K., Hospattankar, A. V., Law, S. W., Meglin, N., Cortright, J,, and
Brewer, H. B., Human apolipoprotein B (apo B) mRNA: identification of two distinct
apo B mRNAs, an mRNA with the apo B-100 sequence and an apoB mRNA containing
a premature in-frame translational stop codon, in both liver and intestine, Proc. Natl.
Acad. Sci. U.S.A., 85, 1772, 1988.
28. Teng, B., Verp, M., Salomon, J., and Davidson, N. 0., Apolipoprotein B messenger
RNA editing is developmentally regulated and widely expressed in human tissues, J.
Bioi. Chern., 265, 20616, 1990.
29. Wu, J, H., Semenkovich, C. F., Chen, S. H., Li, W. H., and Chan, L., Apolipoprotein
B mRNA editing: validation of a sensitive assay and developmental biology of RNA
editing in the rat, J. Bioi. Chern., 265, 12312, 1990.
30. Teng, B., Black, D. D., and Davidson, N. 0., Apolipoprotein B messenger RNA
editing is developmentally regulated in pig small intestine: nucleotide comparison of
apolipoprotein B editing regions in five species, Biochem. Biophys. Res. Comm., 173,
74, 1990.
31. Glickman, R. M., Rogers, M., and Glickman, J, N., Apolipoprotein B synthesis by
human liver and intestine in vitro, Proc. Nat!. Acad. Sci. U.S.A., 83, 5296, 1986.
32. Levy, E., Rochette, C., Londono, I., Roy, C. C., Milne, R. W., Marcel, Y. L., and
Bendayan, M., Apolipoprotein B-1 00: immunolocalization and synthesis in human in-
testinal mucosa, J. Lipid Res., 31, 1937, 1990.
33. Hoeg, J, M., Sviridov, D. D., Tennyson, G. E., Demosky, S. J,, Meng, M. S.,
Bojanovski, D., Safonova, I. G., Repin, V. S., Kuberger, M. B., Smirnov, V. N.,
Higuchi, K., Gregg, R. E., and Brewer, H. B., Both apolipoproteins B-48 and B-100
are synthesized and secreted by the human intestine, J. Lipid Res., 31, 1761, 1990.
34. Dixon, J, L., Furukawa, S., and Ginsberg, H. N., Oleate stimulates secretion of
apolipoprotein B-containing lipoproteins from Hep 02 cells by inhibiting early intracel-
lular degradation of apolipoprotein B, J. Bioi. Chern., 266, 5080, 1991.
35. Coleman, R. A., Haynes, E. B., Sand, T. M., and Davis, R. A., Developmental
coordinate expression of triacylglycerol and small molecular weight apoB synthesis and
secretion by rat hepatocytes, J. Lipid Res., 29, 33, 1988.
36. Jiao, S., Moberly, J, B., and Schonfeld, G., Editing of apolipoprotein B messenger
RNA in differentiated Caco-2 cells, J. Lipid Res .. 31, 695, 1990.
37. Demmer, L.A., Levin, M.S., Elovson, J., Reuben, M.A., Lusis, A. J,, and Gordon,
J, I., Tissue-specific expression and developmental regulation of the rat apolipoprotein
B gene, Proc. Nat!. Acad. Sci. U.S.A., 83, 8102, 1986.
38. Hopkins, B., Brice, A. L., Schofield, P. N., Baralle, F. E., and Graham, C. F.,
Identity of cells containing apolipoprotein B messenger RNA, in 6- to 12-week postfer-
tilization human embryos, Development, 100, 83, 1987.
39. Bostrom, K., Lauer, S. J., Poksay, K. S., Garcia, Z., Taylor, J. M., and Innerarity,
T. L., Apolipoprotein B48 RNA editing in chimeric apolipoprotein EB mRNA, J. Bioi.
Chern., 264, 15701, 1989.
40. Bostrom, K., Garcia, Z., Poksay, K. S., Johnson, D. F., Lusis, A. J,, and Innerarity,
T. L., Apolipoprotein B mRNA editing: direct determination of the edited base and
occurrence in non-apolipoprotein B producing cell lines, J. Bioi. Chern., 265, 22446,
1990.
41. Lao, P. P., Xiong, W., Zhu, H. J,, Chen, S. H., and Chan, L., Apolipoprotein B
mRNA editing is an intracellular event that occurs posttranscriptionally coincident with
splicing and polyadenylation, J. Bioi. Chern., 266, 20550, 1991.
42. Davidson, N. 0., Carlos, R. C., Drewek, M. J., and Parmer, T. G., Apolipoprotein
gene expression in the rat is regulated in a tissue-specific manner by thyroid hormone,
J. Lipid Res., 29, 1511, 1988.
316 Nutrition and Gene Expression
43. Davidson, N. 0., Carlos, R. C., Sherman, H. L., and Hay, R. V., Modulation of
apolipoprotein B-100 mRNA editing: effects on hepatic very low density lipoprotein
assembly and intracellular apoB distribution in the rat, J. Lipid Res., 31, 899, 1990.
44. Davidson, N. 0., Carlos, R. C., and Lukaszewicz, A. M., Apolipoprotein B mRNA
editing modulated by thyroid hormone analogs but not growth hormone administration
in the rat, Mol. Endocrinol., 4, 779, 1990.
45. Underwood, A. H., Emmett, J. C., Ellis, D., Flynn, S. B., Leeson, P. D., Benson,
G. M., Novelli, R., Pearce, N. J., and Shah, V. P., A thyromimetic that decreases
plasma cholesterol levels without increasing cardiac activity, Nature (London), 324, 425,
1986.
46. Sjoberg, A., Oscarsson, J., Bostrom, K., Innerarity, T. L., Eden, S., and Olofsson,
S. 0., Growth hormone participates in the regulation of the editing of apolipoprotein B
mRNA in the rat liver, Arterioscler. Thromb., II, 1402a, 1991.
47. Patterson, A. P., Eggerman, T. L., Demosky, S. J., and Brewer, H. B., T3 Nuclear
receptor and growth hormone modulate apolipoprotein B editing, Arteriosclerosis and
Thrombosis, II , 1403a, 1991.
48. Baum, C. L., Teng, B., and Davidson, N. 0., Apolipoprotein B messenger RNA
editing in the rat liver: modulation by fasting and refeeding a high carbohydrate diet, J.
Bioi. Chon., 265, 19263, 1990.
49. Seishima, M., Bisgaier, C. L., Davies, S. L., and Glickman, R. M., Regulation of
hepatic apolipoprotein synthesis in the 17 alpha-ethinyl estradiol-treated rat, J. Lipid Res.,
32, 941, 1991.
50. Jiao, S., Yanagi, K., Shimomura, I., Kobatake, T., Keno, Y., Kobo, M., Tokunaga,
K., Matsuzawa, Y., and Tarui, S., Enhancement of apoB mRNA editing in the liver
of non-insulin-dependent diabetic rats, Arteriosclerosis and Thrombosis, II, 1424a. 1991.
51. Driscoll, D. M., Wynne, J. K., Wallis, S. C., and Scott, J., An in vitro system for
the editing of apolipoprotein B mRNA, Cell, 58, 519, 1989.
52. Blackhart, B. D., Yao, Z., and McCarthy, B. J., An expression system for human
apolipoprotein BIOO in a rat hepatoma cell line, J. Bioi. Chern., 265. 8358, 1990.
53. Yao, Z., Blackbart, B. D., Linton, M. F., Taylor, S.M., Young, S. G., and McCarthy,
B. J., Expression of carboxyl-terminally truncated forms of human apolipoprotein B in
rat hepatoma cells: Evidence that the length of apolipoprotein B has a major effect on
the buoyant density of the secreted lipoproteins, J. Bioi. Chern., 266, 3300, 1991.
54. Chen, S. H., Li, X., Liao, W. S., Wu, j. H., and Chan, L., RNA editing of apoli-
poprotein B mRNA: sequence specificity detem1ined by in vitro coupled transcription
editing, J. Bioi. Chern., 265, 6811, 1991.
55. Greeve, J., Navaratnam, N., and Scott, J., Characterization of the apolipoprotein B
mRNA editing enzyme: no similarity to the proposed mechanism of RNA editing in
kinetoplasid protozoa, Nucleic Acids Res., 19, 3569, 1991.
56. Driscoll, D. M. and Casanova, E., Characterization of the apolipoprotein B mRNA
editing activity in enterocyte extracts, J. Bioi. Chern., 265, 21401, 1990.
57. Smith, H. C., Kuo, S. R., Backus, J. W., Harris, S. G., Sparks, C. E., and Sparks,
j. D., In vitro apolipoprotein B mRNA editing: Identification of a 27S editing complex,
Proc. Nat/. Acad. Sci. U.S.A., 88, 1489, 1991.
58. Lau, P. P., Chen, S. H., Wang, J. C., and Chan, L., A 40 ki1odalton rat liver nuclear
protein binds specifically to apolipoprotein B mRNA around the RNA editing site, Nucleic
Acids Res., 18, 5817, 1991.
59. Teng, B. B. and Davidson, Evolution of intestinal apolipoprotein B mRNA editing:
chicken apolipoprotein B mRNA is not edited but chicken enterocytes contain in-vitro
editing enhancement factor(s). J. Bioi. Chern .. in press, 1992.
60. Davies, M. S., Wallis, S. C., Driscoll, D. M., Wynne, J. K., Williams, G. W.,
Powell, L. M., and Scott, J., Sequence requirements for apolipoprotein B RNA editing
in transfected rat hepatoma cells, J. Bioi. Chern., 264, 13395, 1989.
Davidson 317
61. Shah, R. R., Knott, T. J., Legros, J. E., Navaratnam, N., Greeve, J. C., and Scott,
J., Sequence requirements for the editing of apo1ipoprotein B mRNA, J. Bioi. Chern.,
266, 16301, 1991.
62. Navaratnam, N., Patel, D., Shah, R. R., Greeve, J. C., Powell, L. M., Knott, T. J.,
and Scott, J., An additional editing site present in apo1ipoprotein B mRNA, Nucleic
Acids Res., 19, 1741, 1991.
Chapter 14
TABLE OF CONTENTS
0-8493-6961-4/93/$0.00 +$.50
© 1993 by CRC Press, Inc. 319
320 Nutrition and Gene Expression
I. INTRODUCTION
ro-Oxidation
(microsomal)
FIGURE 1. Pathways of hepatic fatty acid utilization. When a long chain fatty acid (here
represented as palmitic acid) enters the liver, two major metabolic fates are available, oxidation
via the mitochondria or peroxisomes and reesterification to triglyceride or phospholipid.
modulates the flux of long chain acyl CoA into the mitochondria. Figure 2
presents a classical view of CPT function within the mitochondria. It is clear
that CPT activity is found on the inner surface of the mitochondrial inner
membrane. This activity has been referred to as CPT-B, CPT-II, CPT2 , or
CPT; (for CPT inner). It is also clear that CPT activity is found on the cytosolic
side of the mitochondrion. The outer activity has been referred to as CPT-
A, CPT-I, CPT,, or CPTa (for CPT outer). Further, the outer CPT is inhibited
by malonyl CoA, a substrate in the fatty acid synthetic pathway. Unfortu-
nately, while localization of activities is possible, it remains very difficult to
determine how many CPT proteins are involved in producing these CPT
activities. The problem of definition of the number of distinct CPT proteins,
as well as the problems mentioned in the following section, has confounded
efforts to study the regulation of CPT at the gene level. Only recently has a
68-kDa CPT been purified, cloned, and its regulation studied.H
Palmitoylcarnitine
+
Co A
1-Carnitine
+
Palmitoyi-CoA
1-Carnitine
~-Oxidation
Outer Inner
Membrane Membrane
chondria often show two bands, particularly where CPT activity is induced.
Calculation of M, led us to conclude that the less abundant, upper band
represented the 68-kDa precursor. In support of this, mRNA yields a single
immunoprecipitable band with M, corresponding to the higher weight band
observed on Western blots. Hoppel recently presented the possibility that
outer membrane malonyl CoA binding protein and inner membrane 68-kDa
CPT are associated at mitochondrial membrane pores or junctions. 25 Such
junctions are known to serve as sites for protein import. His suggestion may,
indeed, explain the enrichment of the outer mitochondrial membrane with a
68-kDa CPT precursor. Can we, then, be sure that there is a single mito-
chondrial CPT protein associated with a malonyl CoA-binding regulatory
protein? The answer remains an unfortunate no. The good fortune in this is
that the activity of the 68-kDa CPT is regulated. If this is the only mito-
chondrial CPT, we are well on our way to understanding its regulation. If
there is a second mitochondrial CPT, the data available from in situ studies
indicate that it too is regulated; that is, outer CPT activity changes. It would,
then, be important to determine how this change in activity is mediated. Such
data are currently available for the 68-kDa CPT.
Regulation of the activity of any enzyme can be divided into a number
of events (Figure 3). In the most general sense, these events can be subdivided
into (1) short-term events which alter catalytic efficiency but do not alter the
quantity of protein, and (2) longer term events which specifically alter protein
quantity. If protein quantity is regulated, this regulation can be accomplished
at a number of levels. As with many regulatory enzymes, CPT exhibits both
forms of regulation. The specific difficulty which arises with CPT is that the
majority of in situ studies examine the outer CPT activity, while all studies
of purified CPT involve the 68-kDa protein.
~no~ ••
• Transcription
••
•
Primary
RNA
Transcript
•
: Post-transcriptional
• Processing
••
mRNA
in
Cytosol
•
Translation :
•
Nascent
Protein
Mature
Protein
••
••
Degradation ~· • • • •• • • •••
to 50%) decline in the inner CPT activity. We had previously suggested such
a connection in studies with 2-tetradecylglycidyl CoA (TDGA-CoA). 12
2-Tetradecylglycidyl CoA is presumed to inhibit only the outer CPT
activityY However, if one exposes the intact mitochondria to TGDA-CoA,
washes them, and then inverts them, the inner CPT activity is inhibited. 12
Still, we also reported that the inner CPT activity is, of itself, sensitive to
this inhibitor. Further, the purified 68-kDa CPT protein can be rendered
TGDA-CoA-sensitive under proper conditions. Thus, there was always the
possibility that TDGA-CoA leaked from the outer to the inner surface.
Current and ongoing work from two groups 22 •23 demonstrates that the
combining of 68-kDa CPT and outer mitochondrial membrane or malonyl
CoA-binding protein, under defined conditions, results in a malonyl CoA-
sensitive CPT activity. These same conditions neither result in CPT activity
of the outer membrane preparation in the absence of the 68-kDa CPT nor
malonyl CoA sensitivity of the 68-kDa CPT in the absence of the outer
membrane preparation. Whether they are observing regained CPT activity of
the outer membrane malonyl CoA-binding protein or recovered malonyl CoA
sensitivity of the 68-kDa CPT is unclear. Putting aside the question of one
vs. two CPT proteins, these data would serve to confirm the observation that
mitochondrial outside and inside could communicate relative to a malonyl
CoA inhibition of CPT activities on both faces.
While it remains uncertain how malonyl CoA sensitivity is conferred, a
recurring theme in CPT regulation has been that when CPT activity increases,
malonyl CoA sensitivity declines. This effect is likely quite important in
governing total flux through CPT.
B. REGULATION BY PHOSPHORYLATION
Harano and co-workers have reported that CPT is phosphorylated in
response to cAMP in cultured cells. 28 To our knowledge, this initial obser-
vation has not been pursued. It remains unclear if the phosphorylation has
physiologic significance, but the possibility of this form of regulation will
need to be further investigated.
bution, with a very high cardiolipin content on the inner face. Exposure of
the outer face to cardiolipin also results in increased CPT activity. 11 Second,
a number of physiologic states where CPT activity is elevated, such as star-
vation or severe streptozotocin-diabetes in the rat. also produce increased
mitochondrial fluidity. 12 Thus, membrane fluidity can and does have an effect
on CPT activity without an effect on malonyl CoA sensitivity. The extent to
which membrane fluidity does contribute to overall CPT regulation remains
uncertain. More recent observations on the regulation of the 68-kDa CPT
regulation show that a primary mechanism of control is via the control of
protein synthesis. At the same time, the largest observed increase in 68-kDa
CPT protein is fivefold with a two- to threefold increase a more typical
observation. In assessing the overall control of flux through CPT, the com-
bination of short-term and long-term effects must be considered.
Carnitine Octanoyltransferase
593
659
ceed to determine if these changes are the product of alterations in the quantity
of mRNA and, ultimately, the product of alterations in transcription rates.
D. PROMOTER ANALYSIS
The regulation of the 68-kDa CPT protein must, therefore, involve in-
teraction of specific nuclear regulatory proteins with the promoter region for
the CPT gene. Such genomic regulatory elements are generally found up-
stream or 5' to the structural elements. Therefore, we have undertaken the
isolation of promoter regions for both the rat and human 68-kDa CPTs. 35
Given the high degree of homology of the eDNA for these two proteins, the
probability of successful simultaneous isolation seemed high. Using poly-
merase chain reaction approaches to the isolation of these promoters, we were
able to initially isolate regions of approximately 500 bp upstream from the
mRNA coding region. The 68-kDa CPT, and CPT activity, in general, are
quite interesting because not only do they respond to expected stimuli, such
as cAMP-mediated induction, but, also, they respond to apparently unrelated
stimuli, such as the peroxisomal proliferating agents. We have taken three
simultaneous approaches to the study of the promoter. First, we have examined
the promoter region directly using sequence comparison to known consensus
nuclear protein binding elements, footprinting with purified nuclear binding
TABLE 1 w
w
0
Regulation of the Hepatic Mitochondrial 68-kDa Carnitine Palmitoyltransferase
Starvation Rat i t i 17
Diabetes Rat ti i i i t 17,31
Riboflavin deficiency Rat ii i i NO 4
cAMP Rat i t i t ii 36
H4IIE Cell ii i t NO 32
Sepsis Rat t t NO 34
Interleukin-6 H4IIE Cell t i NO 37
DEHP Rat ti t i ti i t t 5,15,17
Clofibrate Rat t i ii t 38
Note: The quantity of 68 kDa carnitine palmitoyltransferase protein is regulated primarily at the transcriptional level.
Increases in the protein, determined by densitometric scanning of Western blots. increases in mRNA. determined
by blot or translational activity, and transcription rates correlate quite well. The table illustrates the approximate
degree of response ( j = 1.5- to 2-fold increase; j t = 2- to 3-fold increase; i i i 2: 4-fold; L = 50% ~
decrease; NO = not determined). The table also indicates something of the variety of stimuli to which the 68- ~-
kDa CPT responds.
c;·
:::::
1::>
:::::
1::>...
c:J
~
:::::
~
~
""\::i
~
"'c;·
"':::::
Brady eta/. 331
-500 +1
A.
B.
Acute
Phase
c.
FIGURE 5. Mapping the 68-kDa camitine palmitoyltransferase promoter. We have isolated
an approximately 600-bp region of the CPT promoter: (A) shows the areas of protection by rat
liver nuclear extract in footprint analysis; (B) represents regions of protection by purified nuclear
binding proteins CREB, C/EBPa, and C/EBP~; (C) shows the predicted binding regions for a
variety of binding proteins based on consensus nucleotide sequences in the promoter.
proteins where available, and gel mobility shift assays. These studies have
indicated that the 68-kDa CPT promoter contains a single cAMP response
element (CRE), binding sites for C/EBPa (CCAAT/enhancer binding protein),
and for C/EBPI3 (the DBP-IL6 dependent binding protein). Studies with rat
liver nuclear extracts also define a large region of binding which incudes a
C/EBPa and a C/EBP binding region, but one which is much broader and
more complex, containing consensus binding elements for other proteins,
which we have not directly tested. A map of the CPT promoter is presented
(Figure 5).
The second major approach to promoter analysis is functional. The CPT
promoter has been introduced into a plasmid containing a reporter gene,
chloramphenicol acetyltransferase. The CPT promoter is then introduced into
HepG2 cells and its response to various stimuli examined. These can include
hormonal factors as cAMP or thyroid hormone, peroxisomal proliferators,
such as clofibrate, or expression vectors for nuclear regulatory proteins, such
as c-fos, c-jun, or C/EBPa. 35
The final approach represents a descriptive analysis of changes in the
level of nuclear binding proteins. Rat liver or cell nuclear extracts are prepared
following stimulus with cAMP or peroxisomal proliferators. The changes in
specific nuclear proteins can be assayed by Western blotting. Preliminary
data indicate that c-fos, c-jun, and C/EBPI3 increase in sepsis.
The difficulty with the analysis of any promoter is that what may bind
can be easily defined. What does bind is more difficult to determine. Current
and ongoing work in this area is aimed at answering this question.
332 Nutrition and Gene Expression
REFERENCES
I. Kondrup, J. and Lazarow, P. B., Peroxisomal 13-oxidation in intact rat hepatocytes:
quantitation of its flux, Ann. N.Y. Acad. Sci., 386, 404, 1982.
2. Bieber, L. L., Carnitine, Annu. Rev. Biochem., 57, 261, 1988.
3. Ramsay, R. R., Microsomal, mitochondrial and peroxisomal carnitine palmitoyltrans-
ferase activities change differently in starvation and diabetes, Biochim. Biophys. Acta,
(submitted.)
4. Brady, P. S., Feng, Y.-X., and Brady, L. J., Transcriptional regulation of carnitine
palmitoyltransferase synthesis in riboflavin deficiency in rats, J. Nutr., 118, 1128, 1988.
5. Woeltje, K. F., Esser, V., Weis, B. C., Sen, A., Cox, W. F., McPhaul, M. J.,
Slaughter, C. A., Foster, D. W., and McGarry, J.D., Cloning, sequencing, and
expression of a eDNA encoding rat liver mitochondrial carnitine palmitoyltransferase II,
1. Bioi. Chern., 265, 10720, 1990.
6. Finocchiaro, G., Taroni, F., Rocchi, M., Martin, A. L., Colombo, I., Tarelli, G. T.,
and DiDonato, S., eDNA cloning, sequence analysis, and chromosomal localization of
the gene for human carnitine palmitoyltransferase, Proc. Nat/. Acad. Sci. U.S.A., 88,
661, 1991.
7. Murthy, M.S. R. and Pande, S. V., Malonyl-CoA binding site and the overt carnitine
palmitoyltransferase activity reside on the opposite sides of the outer mitochondrial mem-
brane, Proc. Nat/. Acad. Sci. U.S.A., 84, 378, 1987.
8. Nicolay, K., Timmers, R., Spoelstra, E., VanderNeut, R., Fok, J., Huigen, Y.,
Verkleij, A., and deKruiff, B., Biochim. Biophys. Acta, 778, 359, 1984.
9. Brady, L. J,, Silverstein, L. J., Hoppel, C. L., and Brady, P. S., Hepatic mitochondrial
inner membrane properties and carnitine palmitoyltransferase A and B: effect of diabetes
and starvation, Biochem. 1., 232, 445, 1985.
10. Brady, L. J., Hoppel, C. L., and Brady, P. S., Hepatic mitochondrial inner membrane
properties, 13-oxidation and carnitine palmitoyltransferases A and B, Biochem. 1., 233,
427, 1986.
II. Brady, L. J, and Brady, P. S., Hepatic and cardiac carnitine palmitoyltransferase
activity: effects ofadriamycin and galactosamine, Biochem. Pharmacal., 36, 3419, 1987.
12. Brady, P. S. and Brady, L. J., Action in vivo and in vitro of 2-tetradecylglycidic acid,
2-tetradecylglycidyl-CoA and 2-tetradecylglycidylcarnitine on hepatic carnitine palmi-
toyltransferase, Biochem 1., 238, 801-809, 1986.
13. Yates, D. W. and Garland, P. B., Carnitine palmitoyltransferase activities
(EC 2.3.1.-) of rat liver mitochondria, Biochem. 1., 119,547, 1970.
14. Hoppel, C. L. and Brady, L., Carnitine palmitoyltransferase and transport of fatty acids,
in The Enzymes of Biological Membranes, Martonosi, A., Ed., Vol. 2, Plenum Press,
NY, 1985, 139.
15. Miyazawa, S., Ozasa, H., Osumi, T., and Hashimoto, T., Purification and properties
of carnitine octanoyltransferase and carnitine palmitoyltransferase from rat liver, 1. Biochem.
(Tokyo), 94, 529, 1983.
16. Clarke, P.R. H. and Bieber, L. L., Isolation and purification of mitochondrial carnitine
octanoyltransferase from beef heart, 1. Bioi. Chern., 256, 9861, 1981.
17. Brady, P. S. and Brady, L. J,, Hepatic carnitine palmitoyltransferase turnover and
translation rates in fed, starved, streptozotocin-diabetic and diethylhexylphthalate-treated
rats, Biochem. 1., 246, 641, 1987.
18. Kerner, J, and Bieber, L. L., Studies on the association of b-oxidation enzymes with
CPTi and CPTo of heart mitochondria, FASEB 1., 4 (Abstr. 3104), A802, 1990.
19. Declerq, P. E., Falck, J. R., Kuwajima, M., Tyminski, H., Foster, D. W., and
McGarry, J. D., Characterization of the mitochondrial carnitine palmitoyltransferase
enzyme system: use of inhibitors, 1. Bioi. Chern., 262, 9812, 1987.
334 Nutrition and Gene Expression
20. Woeltje, K. F., Kuwajima, M., Foster, D. W., and McGarry, J.D., Characterization
of the mitochondrial camitine palmitoyltransferase enzyme system: use of detergents and
antibodies, J. Bioi. Chem., 262, 9822, 1987.
21. Zammit, V. A., Corstorphine, C. G., and Kolodziej, M. P., Evidence for distinct
functional molecular sizes of camitine palmitoyltransferases I and II in rat liver mito-
chondria, Biochem. J., 263, 89, 1989.
22. Ghagiminejad, I. and Saggerson, E. D., Camitine palmitoyltransferase (CPT2) from
mitochondrial inner membrane becomes inhibitable by malonyl-CoA if reconstituted with
outer membrane malonyl-CoA binding protein, FEBS, 269, 406, 1990.
23. Chung, C., Woldegiorgis, G., and Bieber, L. L., Restoration of malonyl-CoA sen-
sitivity to purified rat heart mitochondrial camitine palmitoyltransferase by addition of
protein fraction(s) from an 86 kD malonyl-CoA binding affinity column, FASEB J., 5
(Abstr. 1290), A592, 1990.
24. Ghagiminejad, I. and Saggerson, E. D., The relationship of rat liver overt camitine
palmitoyltransferase to the mitochondrial malonyl-CoA binding entity and to the latent
palmitoyltransferase, Biochem. J., 270, 787, 1990.
25. Hoppel, C. L., Camitine palmitoyltransferases, in Current Concepts in Carnitine Re-
search, Carter, A., Ed., CRC Press, Boca Raton, FL, 1992, 153.
26. McGarry, J, D., Leatherman, G. F., and Foster, D. W., Camitine palmitoyltransferase.
I, J. Bioi. Chem., 253, 4128, 1978.
27. Kiorpes, T. C., Hoerr, D., Ho, W., Weaner, L. E., Inman, M. G., and Tutwiler,
G. F., Identification of 2-tetradecylglycidyl coenzyme A as the active form of methyl 2-
tetradecylglycidate (methyl palmoxirate) and its characterization as an irreversible, active
site-directed inhibitor of camitine palmitoyltransferase A in isolated rat liver mitochondria,
J. Bioi. Chem., 259, 9750, 1984.
28. Harano, Y., Kashiwago, H., Kojima, H., Suzuki, M., Hashimoto, T., and Shigeta,
Y., Phosphorylation of camitine palmitoyltransferase and activation by glucagon in iso-
lated rat hepatocytes, FEBS Lett., 188, 267-272, 1985.
29. Ozasa, H., Miyazawa, S., and Osumi, T., Biosynthesis of camitine octanoyltransferase
and camitine palmitoyltransferase, J. Biochem. (Tokyo), 94, 543, 1983.
30. Brady, P. S., Marine, K. A., Brady, L. J., and Ramsay, R. R., Co-ordinate induction
of hepatic mitochondrial and peroxisomal camitine acyltransferase synthesis by diet and
drugs, Biochem. J., 260, 93, 1989.
31. Brady, L. J, and Brady, P. S., Regulation of camitine palmitoyltransferase synthesis
in spontaneously diabetic BB Wistar rats, Diabetes, 38, 65,
32. Wang, L., Brady, P. S., and Brady, L. J., Turnover of camitine palmitoyltransferase
mRNA and protein in H4IIE cells: effect of cyclic AMP and insulin, Biochem. J., 263,
703, 1989.
33. Chatterjee, B., Song, C. S., Kim, J, M., and Roy, A. K., Cloning, sequencing and
regulation of rat liver camitine octanoyltransferase: transcriptional stimulation of the
enzyme during peroxisome proliferation, Biochemistry, 27, 9000, 1988.
34. Birklid, S.D., Brady, P. S., Brady, L. J., and Barke, R. A., Hepatic gene expression
in a rat intra-abdominal sepsis model: fatty acid oxidation, FASEB J., 4 (Abstr. 2250),
A654, 1990.
35. Brady, P. S., Liu, J.-S., Park, E. A., Hanson, R. W., and Brady, L. J., Isolation
and characterization of the promoter for the gene coding for camitine palmitoyltransferase,
Biochem. J., 286, 779, 1992.
36. Brady, L. J, and Brady, P. S., Regulation of camitine palmitoyltransferase by glucagon
and insulin, Biochem. J., 258, 677, 1989.
37. Barke, R. A., Brady, L. J., and Brady, P. S., The Ca2+ second messenger system
and IL-l-alpha modulation of hepatic gene transcription and mitochondrial fatty acid
oxidation, Surgery, 110, 285, 1991.
38. Brady, L. J., Ramsay, R. R., and Brady, P. S., Regulation of camitine acyltransferase
synthesis in lean and obese Zucker rats by DHEA and clofibrate, J. Nutr., 121, 525,
1991.
Chapter 15
Malcolm Watford
TABLE OF CONTENTS
References ............................................................... 34 7
0-8493-6961-4/93/$0.00 +$.50
1993 by CRC Press. Inc. 335
336 Nutrition and Gene Expression
I. INTRODUCTION
drolysis of glutamine occur in the liver concomitantly and the net flux is,
therefore, the difference between these two pathways. 24 Although species
differences exist, it appears that the liver is a site of glutamine synthesis in
the normal physiological conditions 14 but changes to a site of net utilization
in diabetes, 25 •26 high protein feeding, 15 •22 exercise, 23 and during some stages
of feeding and starvation. 27 This is in keeping with its role as a direct substrate
for gluconeogenesis and urea synthesis. Glutamine also serves as an important
precursor for liver urea synthesis by the indirect pathway of glutamine ca-
tabolism, in extrahepatic cells, such as enterocytes, to alanine, ammonia, and
other amino acids which are then taken up by the liver. 8
B. GLUTAMINASE
The degradation of glutamine can occur by many reactions, including
many amidotransferases. However, there is only one true glutaminase, giving
rise to stoichiometric amounts of glutamate and ammonia. 30 Unfortunately,
many enzymes will produce ammonia from glutamine and therefore reliable
methods for the assay of glutaminase activity in nonpurified preparations must
rely on the measurement of glutamate production. Phosphate activated glu-
taminase, sometimes known as phosphate-dependent glutaminase or glutam-
inase l (EC 3.5.1.2, L-glutamine amidohydrolase), is the only true glutam-
inase found in mammalian tissues (Equation 2). It is the major enzyme
responsible for glutamine catabolism in the tissues mentioned in this review,
with the possible exception of lactating mammary gland. 21 ·31 It is hereafter
referred to simply as glutaminase. However, there are two isozymes of glu-
taminase, described here as liver-type and kidney-type; the latter is sometimes
referred to as brain-type in the literature.
ARGININO- ASL
A S / SUCCINATE~
OTC
~ ORNITHINE
_j '-II UREA I
CARBAMOYL PHOSPHATE
HCO) ~CPS I
GLUTAMATE+ NH 3
t GLUTAMINASE
II GLUTAMINE II
FIGURE 1. Hepatic glutaminase and the urea cycle.
The function of the urea cycle is ammonia detoxification but since it also
removes bicarbonate it has been proposed to play a significant role in the
regulation of systemic pH. The complete cycle is found only in the liver
although partial reactions are found in the kidney and small intestine consti-
tuting tissue-compartmented system for arginine synthesis. 61 These aspects
have been the subject(s) of many recent reviews. 55 •62 · 65
A. SHORT-TERM REGULATION
Both hepatic glutamine metabolism and the urea cycle are subject to
regulation by both short-term and long-term mechanisms. Short-term regu-
lation can be inferred from the observation that the concentration of glutamine
within the liver is held constant at about 5 1-Lmoles/g wet weight. The con-
centrations of urea cycle substrates, ammonia, and bicarbonate are also not
reduced to zero and do not vary widely. A number of key regulatory points
have been identified in the pathway of hepatic glutamine metabolism, transport
into the cell, glutaminase and a-ketoglutarate dehydrogenase activities, and
entry into the urea cycle. 34 •35 •52 A variety of evidence has been cited to
demonstrate a regulatory role for hepatic glutaminase activity. Recently Pog-
son et al. 52 have applied the calculations of quantitative regulatory theory to
this problem in isolated rat hepatocytes. They report that glutaminase exhibits
a control strength of close to 1.0 (indicative of a major regulatory role), while
that of transport system N is 0.31 and system Lis -0.4 (indicating a negative
effect). Despite problems with the application of this theory to enzymes within
the mitochondrial matrix the results are further indication that glutaminase
activity is of regulatory significance.
Although glutamine at high concentrations (>2 mM) is readily utilized
by experimental systems in vitro, such as the perfused liver and isolated
hepatocytes, no net glutamine utilization is observed at physiological gluta-
mine concentrations ( < 1 mM). K A number of substances will stimulate
glutamine utilization in such systems, for example, glucagon, ammonia, leu-
cine, cysteine, bicarbonate, low pH, cAMP, catecholamines, vasopressin,
angiotensin II, and thyroxine. 34 ·66 . 69 The mechanisms by which these agents
exert their effects is not well understood and their relevance in vivo is not
clear. They may act by changing the interaction of glutaminase with the inner-
mitochondrial membrane. 69 Recent work has suggested that hormonal regu-
lation of cell (and mitochondrial) volume may play a role. 70 •71 Ochs72 has
proposed that glucagon and cAMP act at the level of glutaminase activity but
that hormones acting via changes in calcium (a-adrenergic, vasopressin, and
angiotensin II) act at the level of a-ketoglutarate dehydrogenase. As mentioned
in Section III, hepatic glutaminase activity is activated by one of its products,
ammonia. This was first described by Charles 73 and subsequently shown to
be an absolute requirement for the enzyme. 34 •74 Welboume 75 has proposed an
Watford 341
B. LONG-TERM REGULATION
Since the pathways of glutamine catabolism and urea synthesis within the
liver are tightly regulated in the short-term, the capacity of these pathways
is, by definition, in excess under normal circumstances. But there is also
extensive evidence of long-term regulation, although for hepatic glutaminase
this has only recently been investigated in depth.
As pointed out by Folin81 in 1905, urinary urea represents the most
variable nitrogenous excretion product. In what must now be considered a
potentially dangerous series of experiments, Polin showed that urea production
was variable and regulated by diet. He fed himself, and others, a very low
protein diet of arrowroot starch and cream for 14 d and found that urea
excretion fell to low levels, but it rose again within 24 h of resuming his
normal diet. Although based on anecdotal evidence, this downregulation of
urea production has been reported to be of pathological importance in the
refeeding of high protein diets to severely malnourished prisoners at the end
of World War 11. 82
The long-term regulation of hepatic glutaminase has only been recently
recognized. Some earlier reports indicated that the activity was increased after
feeding high protein diets 83 and decreased on feeding low protein diets. 84
342 Nutrition and Gene Expression
TABLE 1
Long-Term Regulation of Glutaminase in Rat Liver
Note: All values reported as% of control values and are means ± SEM
for 4 to 6 animals in each study. Control activity = 4.1 f.Lmol!
min/g wet wt. Other values are based on arbitrary densitometry
units.
These results were based on questionable assay methods but did reflect changes
in activity although the absolute values were incorrect. Examination of the
regulation of hepatic glutaminase required the establishment of an accurate
and reliable assay procedure based on that of Curthoys and Lowry. 85 The
original method was modified to accommodate the kinetic properties of the
hepatic enzyme. This meant the inclusion of high substrate concentration
(100 mM glutamine), limited phosphate (50 mM), and the inclusion of am-
monium chloride (2 rnM) to ensure complete activation of the enzyme. 86
Using this method, it has been shown that hepatic glutaminase activity is
increased during diabetes, 86 high protein feeding, 87 and starvation, 87 and de-
creased during low protein feeding 88 (Table 1). These changes are unrelated
to changes in acid-base status, the major determinant of renal glutaminase
activity. Using a protein-free diet, McGivan et a!. 89 found reduced activity
of hepatic glutaminase. Similar results are seen with a 5% protein diet 88 which
gives a more consistent response since rats consuming a protein-free diet lose
weight and are obviously in a catabolic state. The observed pattern of reg-
ulation of hepatic glutaminase activity is similar to the pattern observed for
net hepatic glutamine catabolism in vivo.
Long-term changes in hepatic glutaminase activity parallel those observed
in the activity of all five urea cycle enzymes and in the activity of a key
regulatory enzyme of gluconeogenesis, phosphoenolpyruvate carboxyki-
nase. 65 Thus changes in the maximal capacity of hepatic glutaminase are in
keeping with the role of glutamine as a substrate for urea synthesis and
gluconeogenesis. It is well established that hormones, such as glucagon,
insulin, and glucocorticoids, are the agents acting at the level of the liver cell
to regulate expression of the gluconeogenic and urea cycle enzymes. 65 ·90 To
date the question of hormonal regulation of liver glutaminase has received
little attention. In rat hepatocytes in primary culture, the activity is reported
to increase in response to glucagon and to exogenous ammonia. 89 However,
the activity decreases in cells in primary culture in the absence of hormones,
and even with hormones the activity does not rise above that seen in freshly
isolated hepatocytes. It is therefore not clear what mechanisms are involved
in this response or if it has any bearing on the situation in vivo.
Watford 343
V. REGULATION OF EXPRESSION OF
GLUTAMINASE GENES
A. GLUTAMINASE cDNAs
Liver glutaminase is encoded by a rnRNA of approximately 2.8 kb in
length which is expressed only in postnatal liver. 48 It is absent until a few
hours prior to birth but rapidly reaches adult levels within 2 days of birth. 48
As mentioned elsewhere, kidney-type glutaminase is comprised of two dif-
ferent sized subunits. Curthoys 46 has recently shown that both subunits are
the product of the same gene, located on chromosome 9 in the rat. 94 Tran-
scription of the gene gives rise to two species of mRNA, 6.0 and 3.3 kb in
length. Both encode a 72-kDa precursor polypeptide which is then differ-
entially processed to give the 65- and 68-kDa subunits. 46
Comparison of the known eDNA sequences of liver- and kidney-type
glutaminases shows little similarity at the nucleotide level. When the predicted
amino acid sequences are compared a region of -150 residues with high
identity (>85%) has been found. 46 •48 The complete sequence of liver-type
glutaminase has not yet been determined. The results to date indicate that the
two isozymes are the products of different but evolutionary related genes.
Interestingly, the family of CPS enzymes has been shown to have evolved
from the fusion of a glutaminase with a synthetase gene, 95 and many of the
amidotransferases exhibit a conserved active site triad of cysteine/histidine/
aspartate. 96 The presence of such a site has not been detected in the mammalian
glutaminase sequences determined to date46 .48 and may therefore indicate a
separate line of evolution.
ivenous cells. 55 •98 •99 Haussinger and co-workers, 64 using the isolated perfused
rat liver, proposed an intercellular glutamine cycle, with glutamine being
degraded in the periportal cells and synthesized in perivenous cells. The model
required that glutaminase be located predominantly in periportal cells. Studies 100
of enzyme activity and mRNA abundance in isolated periportal and perivenous
cells show glutaminase to be mainly located in the periportal cells along with
other enzymes of glutamine catabolism, phosphoenolpyruvate carboxykinase,
and the urea cycle enzymes. The pattern of expression of the carbamoyl
synthetase I changes during different physiological conditions, 101 and the
availability of eDNA probes will allow experiments to examine if this is also
true for liver-type glutaminase.
C. REGULATION OF EXPRESSION
Long-term increases in the activity of enzymes may be the result of the
modification of existing protein or due to changes in the amount of the protein.
In the rat the activity of hepatic glutaminase is increased fourfold in strep-
tozotocin diabetes. 86 Using equivalence point titration and Western blotting,
the increase in glutaminase activity during diabetes was shown to be com-
pletely accounted for by an increase in the amount of glutaminase protein
with no change in the specific activity of the protein. 45 The availability of a
eDNA has allowed determination of the relative abundance of glutaminase
mRNA and this too increases fourfold. 48 Recent work has established that
this is due to a fourfold increase in the rate of transcription of the hepatic
glutaminase gene (Table 1). 88 The abundance of the mRNA is also increased
in response to injection of large doses of dibutyryl cAMP. 101 After high (60%)
protein feeding or starvation, there is an increase in liver glutaminase activity,
mRNA abundance, and rate of transcription. 88 Conversely, low (0 or 5%)
protein feeding results in decreases in glutaminase activity, mRNA abundance,
and transcription rate. 88 These results establish that liver glutaminase is reg-
ulated in the long-term primarily by changes in the rate of transcription of
the gene, and no changes in protein or mRNA turnover need be postulated.
This is in striking contrast to the expression of kidney-type glutaminase
in rat kidney. Expression is regulated by changes in the amount of enzyme
and rate of enzyme synthesis and this correlates with changes in the relative
abundance of the mRNA. 103 However, it appears that mRNA turnover, rather
than changes in transcription rate, is the predominant mechanism of regula-
tion. 103 • 104 In metabolic acidosis the abundance of the message increases five-
to sevenfold and appears to be very stable. On recovery from acidosis, the
abundance of the message falls rapidly and no changes in the rate of tran-
scription are seen. In LLPC-PK 1-F+ cells in culture the abundance of the
mRNA is increased >20-fold at pH 6.9 compared to pH 7.4, and again no
changes in the rate of transcription of the gene are seen. The apparent half-
life of the mRNA changes from 2.5 hat pH 7.4 to 15 hat pH 6.9 and thus
accounts for the rise in abundance. 104 The long (6 kb) kidney-type glutaminase
Watford 345
regulatory elements. For CPS I and OTC and arginase, the presence of ele-
ments recognized by C/EBP or related factors has been demonstrated, and it
is likely that CRE and GRE will be present. 65 A unique sequence has been
found, "UCE" (urea cycle element), 65 • 115 in the regions 5' to at least 4 of
the genes (OTC, AS, ASL, and arginase) and this may play a role in the
coordinate regulation. It has been hypothesized that this element may be
involved in the coordinate expression of other genes involved in nitrogen
metabolism.
The common expression of glutaminase with urea cycle enzymes (and
gluconeogenic enzymes) in periportal cells, and the similar ontogenic devel-
opment pattern, appearing around birth in the liver, is in keeping with the
role of hepatic glutamine metabolism. However, differences do exist. The
urea cycle enzymes are expressed in other tissues, CPS I and OTC in the
intestine, and ASS and ASL in kidney, while hepatic glutaminase is only
expressed at measurable levels in postnatal liver. Evidence from experiments
in vivo and in vitro indicate that glutaminase will have similar elements for
the tissue-specific and hormonal regulation of expression. The presence or
absence of these, and the UCE, await the availability of genomic clones and
sequence analysis.
VI. SUMMARY
ACKNOWLEDGMENTS
Work done in the author's laboratory was supported by Grant DK-37301
from the National Institutes of Health. The author is grateful to Drs. Brosnan,
Curthoys, and Morris for providing manuscripts in press.
REFERENCES
l. Lacey, J. M. and Wilmore, D. W., Is glutamine a conditionally essential amino acid?,
Nutr. Rev., 48, 297, 1990.
2. Lowe, D. K., Benfell, K., Smith, R. J., Jacobs, D. 0., Murawski, B., Ziegler, T. R.,
and Wilmore, D. W., Safety of glutamine-enriched parenteral nutrient solutions in
humans, Am. J. Clin. Nutr., 52, 1101, 1990.
3. Furst, P., Alpers, S., and Stehle, P., Dipeptides in clinical nutrition, Proc. Nutr. Soc.,
49, 343, 1990.
4. Windmueller, H. G. and Spaeth, A. E., Uptake and metabolism of plasma glutamine
by the small intestine, J. Bioi. Chern., 249, 5070, 1974.
5. Newsholme, E. A. and Parry-Billings, M., Properties of glutamine release from skeletal
muscle and its importance to the immune system, J. Parent. Ent. Nutr., 14, 63S, 1990.
6. Souba, W. W., Glutamine. A key substrate for the splanchnic bed, Annu. Rev. Nutr.,
11,285, 1991.
7. Souba, W. W., Smith, R. J., and Wilmore, D. W., Glutamine metabolism by the
intestinal tract, J. Parent. Ent. Nutr., 9, 608, 617, 1985.
8. Lund, P. and Watford, M., Glutamine as a precursor of urea, in The Urea Cycle,
Grisolia, S., Baguena, R., and Mayor, F., Eds., John Wiley & Sons, New York, 1976,
479.
9. Squires, E. J. and Brosnan, J. T., Measurement of the turnover rate of glutamine in
normal and acidotic rats, Biochem. J., 210, 277, 1983.
10. Lund, P. and Williamson, D. H., Inter-tissue nitrogen fluxes, Brit. Med. Bull., 41,
251, 1985.
11. Welbourne, T. C., Role of the lung in glutamine homeostatis, Contr. Nephrol., 63, 178,
1988.
12. Souba, W. W., Herkowitz, K., and Plumley, D. A., Lung glutamine metabolism, J.
Parent. Ent. Nutr., 14, 68S, 1990.
13. Frayn, K. N., Khan, K., Coppack, S. W., and Elia, M., Amino acid metabolism in
human subcutaneous adipose tissue, Clin. Sci., 80, 471, 1991.
14. Aikawa, T., Matsuka, H., Yamamoto, H., Okuda, T., Ishikawa, E., Kawano, T.,
and Matsumura, E., Gluconeogenesis and amino acid metabolism. Interorganal relations
and roles of glutamine and alanine in the amino acid metabolism of fasted rats, J.
Biochem., 74, 1003, 1974.
15. Fafournoux, P., Remesy, C., and Demigne, C., Fluxes and membrane transport of
amino acids in rat liver under different protein diets, Am. J. Physiol., 259, E614, 1990.
16. Tannen, R. L., Ammonia metabolism, Am. J. Physiol., 235, F265, 1978.
17. Watford, M., Lund, P., and Krebs, H. A., Isolation and metabolic characteristics of
rat and chicken enterocytes, Biochem. J., 178, 589, 1979.
18. Ardawi, M. S. M. and Newsholme, E. A., Metabolism in lymphocytes and its im-
portance in the immune response, Essays Biochem., 21, I, 1985.
348 Nutrition and Gene Expression
19. Newsholme, E. A., Crabtree, B., and Ardawi, M.S. M., The role of high rates of
glycolysis and glutamine metabolism in rapidly dividing cells. Biosci. Rep., 5, 393, 1985.
20. Watford, M., Erbelding, E. J., and Smith, E. M., Glutamine metabolism in rat small
intestine: response to lactation. Biochem. Soc. Trans., 14. 1058, 1986.
21. Horowitz, M. L. and Knox, W. E., A phosphate activated glutaminase in rat liver
different from that in kidney and other tissues, Enrymol. Bioi. Clin., 9, 214, 1968.
22. Yamamoto, H., Aikawa, T., Matsumura, H., Okuda, T., and Ishikawa, E., Inter-
organa! relationships of amino acid metabolism in fed rats, Am. J. Physiol., 244, 1428,
1974.
23. Wasserman, D. H., Greer, R. J., Williams, P. E., Becker, T., Lacy, D. B., and
Abumrad, N. N., Interaction of gut and nitrogen metabolism during exercise, Metab-
olism, 40, 307, 1991.
24. Vincent, N., Martin, G., and Baverel, G., Simultaneous synthesis and degradation of
glutamine in isolated rat liver cells. Effect of vasopressin. Biochim. Biophys. Acta, 1014,
184, 1989.
25. Schrock, H. and Goldstein, L., Interorgan relationships for glutamine metabolism in
normal and acidotic rats, Am. J. Physiol., 240, E519. 1981.
26. Brosnan, J. T., Man, K.-C., Hall, D. E., Colbourne, S. A., and Brosnan, M. E.,
Interorgan metabolism of amino acids in streptozotocin-diabetic ketoacidotic rat, Am. J.
Physiol., 244, El51, 1983.
27. Yamamoto, H., Aikawa, T., Matsutaka, H., Okuda, T., and Ishikawa, E., Inter-
organa! relationships of amino acid metabolism in fed rats, Am. J. Physiol., 226, 1428,
1974.
28. Krebs, H. A., Metabolism of amino acids. The synthesis of glutamine from glutamic
acid and ammonia, and the enzymic hydrolysis of glutamine in animal tissues, Biochem.
J., 29, 1951, 1935.
29. Cooper, A. j. L., Glutamine synthetase, in Glutamine and Glutamate in Mammals,
Kvamme, E., Ed., CRC Press. Boca Raton, FL, 1988, 7.
30. Meister, A., Function of glutathione in kidney via the gamma-glutamyl cycle, Med.
Clin. N. Am., 59, 649, 1975.
31. Erbelding, E. J., Glutamine Metabolism in the Small Intestine. MS thesis. Cornell
University, Ithaca, NY, 1985.
32. Huang, Y.-Z. and Knox, W. E., A comparative study of glutaminase isozymes in rat
tissues, Enzyme, 21, 408, 1976.
33. Curthoys, N. P., Cellular distribution and induction of the enzymes of renal ammoni-
agenesis and gluconeogenesis, in pH Homeostasis, Haussinger, D .. Ed. , Academic Press,
New York, 323, 1988.
34. McGivan, j. D., Metabolism of glutamine and glutamate in liver-regulation and phys-
iological significance, in Glutamine and Glutamate in Mammals, Kvamme. E., Ed., CRC
Press, Boca Raton, FL, 1988, 183.
35. Kovacevic, Z. and McGivan, J. D., Mitochondrial metabolism of glutamine and glu-
tamate and its physiological significance, Physiol. Rev., 63, 547, 1983.
36. Kalra, J. and Brosnan, J. T., Localization of glutaminase in rat liver, FEBS Lett., 32,
325, 1973.
37. McGivan, J. D., Lacey, J. H., and Joseph, S. K., Localization and some properties
of phosphate-dependent glutaminase in disrupted liver mitochondria. Biochem. J., 192,
537, 1980.
38. Kvamme, E., Tveit, B., and Svenneby, G., Glutaminase from pig renal cortex. I.
Purification and general properties, J. Bioi. Chern., 245. 1871, 1970.
39. Curthoys, N. P., Kuhlenschmidt, T., and Godfrey, S. S., Regulation of renal am-
moniagenesis. Purification and characterization of phosphate-dependent glutaminase from
rat kidney, Arch. Biochem. Biophys., 174. 82, 1976.
40. Svenneby, G., Pig brain glutaminase: purification and identification of different enzyme
forms. J. Neurochem., 17, 1591, 1970.
Watford 349
41. Quesada, A. R., Sanchez-Jimenez, F., Perez-Rodriguez, J., Marquez, J., Medina,
M. A., and Nunez de Castro, I., Purification of phosphate-dependent glutaminase from
isolated mitochondria of Ehrlich ascites-tumour cells, Biochem. J., 255, 1031, 1988.
42. Curthoys, N. P., Kuhlenschmidt, K., Godfrey, S. S., and Weiss, R. F., Phosphate-
dependent glutaminase from rat kidney. Cause of increased activity in response to acidosis
and identity with glutaminase from other tissues, Arch. Biochem. Biophys., 172, 162,
1976.
43. Patel, M. and McGivan, j. D., Partial purification and properties of rat liver glutaminase,
Biochem. J., 220, 583, 1984.
44. Heini, H. G., Gebhardt, R., and Mecke, D., Purification and characterization of rat
liver glutaminase, Eur. J. Biochem., 162, 541, 1987.
45. Smith, E. M. and Watford, M., Rat hepatic glutaminase: purification and immuno-
chemical characterization, Arch. Biochem. Biophys., 260, 740, 1988.
46. Shapiro, R. A., Farrell, L., Srinivasan, M., and Curthoys, N. P., Isolation, char-
acterization, and in vitro expression of a eDNA that encodes the kidney isoenzyme of
the mitochondrial glutaminase, J. Bioi. Chern., 266, 18792, 1991.
47. Banner, C., Hwang, J.-J., Shapiro, R. A., Wenthold, R. J., Nakatani, Y., Lampe!,
K. A., Thomas, J. W., Huie, D., and Curthoys, N. P., Isolation of a eDNA for rat
brain glutaminase, Mol. Brain Res., 3, 247, 1988.
48. Smith, E. M. and Watford, M., Molecular cloning of a eDNA for rat hepatic glutam-
inase. Sequence similarity to kidney-type glutaminase, J. Bioi. Chern., 265, 10631, 1990.
49. Under-Horowitz, M., Changes in glutaminase activities of rat liver and kidney during
pre- and post-natal development, Biochem. J., 114,65, 1969.
50. Kilberg, M. S., Handlogten, M. E., and Christensen, H. N., Characteristics of an
amino acid transport system in rat liver for glutamine, asparagine, histidine and closely
related analogs, J. Bioi. Chern., 255,4011, 1980.
51. Farfournoux, P., Demigne, C., Remesy, C., and Le Cam, A., Bidirectional transport
of glutamine across the cell membrane in rat liver, Biochem. 1 .. 216. 401, 1983.
52. Pogson, C. 1., Low, S. Y., Knowles, R.N., Salter, M., and Rennie, M. J., Application
of metabolic control theory to amino acid metabolism in liver, Alfred Benson Symp., 30,
262, 1991.
53. Blackburn, E. H. and Hird, F. J. R., Metabolism of glutamine and glutamate by rat
liver mitochondria, Arch. Biochem. Biophys., 152, 258, 1972.
54. Jungas, R. L., Halperin, M. L., and Brosnan, J. T., A quantitative analysis of amino
acid oxidation and related gluconeogenesis in man, Physiol. Rev., in press.
55. Meijer, A. J., Lamers, W. H., and Chamuleau, R. A. F. M., Nitrogen metabolism
and ornithine cycle function, Physiol. Rev., 70. 701, I990.
56. Cohen, N. S., Cheung, C.-W., and Raijman, L., Channeling of extramitochondrial
ornithine to matrix ornithine transcarbamoylase, J. Bioi. Chern., 262, 203, 1987.
57. Cheung, C.-W., Cohen, N. S., and Raijman, L., Channeling of urea cycle intermediates
in situ in permeabilized hepatocytes, J. Bioi. Chern., 264, 4038, 1989.
58. Watford, M., Channeling in the urea cycle: a metabolon spanning two compartments,
Trends Biochem. Sci., 14, 313, 1989.
59. Watford, M., The urea cycle: a two-compartment system, Essays Biochem., 26, 49,
1990.
60. Meijer, A. j., Channeling of ammonia from glutaminase to carbamoyl-phosphate syn-
thetase in liver mitochondria, FEBS Lett., 191. 249, 1985.
61. Dhanakoti, S. N., Brosnan, J. T., Herzberg, G. R., and Brosnan, M. E., Renal
arginine synthesis: studies in vitro and in vivo, Am. J. Physioi .. 259. E437, 1990.
62. Atkinson, D. E. and Bourke, E., The role of ureagenesis in pH homeostasis, Trends
Biochem. Sci., 9, 291, 1984.
63. Brosnan, J. T., Lowry, M., Vinay, P., Gougoux, A., and Halperin, M. L., Renal
ammonium production-une vue canadienne. Can. J. Physioi. Pharamacoi., 65, 489,
1987.
350 Nutrition and Gene Expression
64. Haussinger, D., Nitrogen metabolism in liver: structural and functional organization and
physiological relevance, Biochem. J., 267, 281. 1990.
65. Morris, S. M., Regulation of enzymes of urea and arginine synthesis, Annu. Rev. Nutr.,
12, in press.
66. Corvera, S. and Garcia·Sainz, j. A., Hormonal stimulation of mitochondrial glutam-
inase. Effects of vasopressin, angiotensin II, adrenaline and glucagon, Biochem. J., 210,
957, 1983.
67. Kashiwagura, T., Erecinska, M., and Wilson, D. F., pH Dependence of hormonal
regulation of gluconeogenesis and urea synthesis from glutamine in suspensions of he-
patocytes, J. Bioi. Chern., 260, 407. 1985.
68. Verhoeven, A. j., Estrela, j. M., and Meijer, A. j., Adrenergic stimulation of glu-
tamine metabolism in isolated rat hepatocytes, Biochem. J., 230, 457, 1985.
69. McGivan, j. D., Vadher, M., Lacey, J., and Bradford, N., Rat liver glutaminase.
Regulation by reversible interaction with the mitochondrial membrane. Eur. J. Biochem.,
148, 323, 1985.
70. Haussinger, D. and Lang, F., The mutual interaction between cell volume and cell
function: a new principle of metabolic regulation, Biochem. Cell. Bioi., 69, I, 1991.
71. vonDahl, S., Hallbrucker, C., Lang, F., and Haussinger, D., Regulation of liver cell
volume and proteolysis by glucagon and insulin, Biochem. J., 278, 771, 1991.
72. Ochs, R. S., Glutamine metabolism in isolated hepatocytes. Evidence for catecholamine
stimulation of a-ketoglutarate dehydrogenase, J. Bioi. Chern., 259, 13004, 1984.
73. Charles, R., Mitochondriale Citrulline Synthese: een Ammonik Fixerend en ATP Ver-
bruikend Proces, Ph.D. thesis, University of Amsterdam, Rototype, Amsterdam, 1968.
74. McGivan, j. D. and Bradford, N. M. Characteristics of the activation of glutaminase
by ammonia in sonicated rat liver mitochondria. Biochim. Biophys. Acta, 759, 241, 1983.
75. Welbourne, T. C., lnterorgan glutamine flow in metabolic acidosis, Am. J. Physiol.,
253, F1069, 1987.
76. Kaiser, S., Gerok, W., and Haussinger, D., Ammonia and glutamine metabolism in
human liver slices. New aspects of the pathogenesis of hyperammonemia in chronic liver
disease, Eur. J. Clin. Invest., 18, 535, 1988.
77. Quesada, A. R., Medina, M.A., Marquez, J., Sanchez-Jimenez, F. M., and Nunez
de Castro, I., Contribution by host tissue to circulating glutamine in mice inoculated
with Ehrlich ascites tumor cells. Can. Res., 48, 1551, 1988.
78. Watford, M., A swell way to regulate metabolism, Trends Biochem. Sci., 15, 329,
1990.
79. Tannen, R. L. and Sastrasinh, S., Response of ammonia metabolism to acute acidosis,
Kidney Int., 25, I, 1984.
80. Schoolwerth, A. C. and LaNoue, K., Transport of metabolic substrates into renal
mitochondria, Annu. Rev. Physiol., 47, 143, 1985.
81. Folio, 0., Laws governing the chemical composition of urine. Am. J. Physiol., 13, 66,
1905.
82. Freedland, R. A. F. and Briggs, S., A Biochemical Approach to Nutrition, Chapman
and Hall, London, 1977, 46.
83. Freedland, R. A. F. and Taylor, A. R., Studies on glucose-6-phosphatase and glutam-
inase in rat liver and kidney, Biochim. Biophys. Acta, 92, 567, 1964.
84. Simell, 0., Effect of low protein diet and hyperammonemia on liver glutaminase activity
in the rat, Experientia, 30, 324, 1974.
85. Curthoys, N. P. and Lowry, 0. H., The distribution of glutaminase isoenzymes in the
various structures of the nephron in normal. acidotic, and alkalotic rat kidney, J. Bioi.
Chern., 248, 162, 1973.
86. Watford, M., Smith, E. M., and Erbelding, E. j., Regulation of phosphate-activated
glutaminase activity and glutamine metabolism in the streptozotocin-diabetic rat, Biochem.
J., 224, 207, 1984.
Watford 351
87. Watford, M., Erbelding, E. J., Shapiro, A. C., Zakow, A. C., and Smith, E. M.,
The adaptive response of phosphate-activated glutaminase in the rat. Contr. Neph., 47,
140, 1985.
88. Watford, M., Zhan, Z., and Vincent, N. C., unpublished.
89. McGivan, J. D., Boon, K., and Doyle, F. A., Glucagon and ammonia influence the
long-term regulation of phosphate-dependent glutaminase activity in primary cultures of
rat hepatocytes, Biochem. J., 274, 103. 1991.
90. Snodgrass, P. J., Lin, R. C., Muller, W. A., and Aoki, T. T., Induction of urea cycle
enzymes of rat liver by glucagon, J. Bioi. Chern., 253, 2748, 1978.
91. Watford, M., Regulation of expression of the genes for glutaminase and glutamine
synthetase in the acidotic rat, Contr. Neph., 92, 211, 1991.
92. Watford, M., Erbelding, E. J., and Smith, E. M., The regulation of glutamine and
ketone body metabolism in the small intestine of the long-term (40-day) streptozotocin-
diabetic rat, Biochem. J., 242, 61, 1987.
93. Nagy, L. E. and Kretchmer, N., Effect of diabetic ketosis on jejunal glutaminase,
Arch. Biochem. Biophys., 248, 80, 1986.
94. Mock, B., Kozak, C., Seldin, M. F., Ruff, N., D'Hoostelaere, L., Szpirer, C.,
Seuanez, H., O'Brien, S., and Banner, C., A glutaminase (Gls) gene maps to mouse
chromosome l, rat chromosome 9, and human chromosome 2. Genomics, 5. 291, 1989.
95. Nyunoya, H., Broglie, K. E., and Lusty, C. J., The gene coding for carbamoyl-
phosphate synthetase I was formed by fusion of an ancestral glutaminase gene and a
synthetase gene, Proc. Nat/. Acad. Sci. U.S.A., 82, 2244, 1985.
96. Zalkin H., Argos, P., Narayana, S. V. L., Tiedeman, A. A., and Smith, j. M.,
Identification of a trpG-related glutamine amide transfer domain in Escherichia coli GMP
synthetase, J. Bioi. Chern., 260, 3350. 1985.
97. Gebhardt, R. and Mecke, D., Heterogeneous distribution of glutamine synthetase among
rat liver parenchymal cells in situ and in primary culture, EMBO J., 2, 567. 1983.
98. Jungermann, K. and Katz, N., Functional specialization of different hepatocyte pop-
ulations, Physiol. Rev., 69, 708, 1989.
99. Gaasbeek-Janzen, J. W., Lamers, W. H., Moorman, A. F. M., deGraaf, A., Los,
J. A., and Charles, R., Immunohistochemical localization of carbamoylphosphate syn-
thetase (ammonia) in adult rat liver. Evidence for a heterogeneous distribution, J. His-
tochem. Cytochem., 32, 557, 1984.
100. Watford, M. and Smith, E. M., Distribution of hepatic glutaminase activity and mRNA
in perivenous and periportal hepatocytes, Biochem. J., 267, 265, 1990.
101. Moorman, A. F. M., deBoer, P. A. j., Charles, R., and Lamers, W. H., Diet and
hormone induced reversal of the carbamoylphosphate synthetase mRNA gradient in the
rat liver lobulus, FEBS Leu., 276, 9, 1990.
102. Watford, M. and Smith, E. M., Regulation of hepatic glutaminase mRNA levels in
the rat, Biochem. Soc. Trans., 17, 175, 1988.
103. Tong, J., Shapiro, R. A., and Curthoys, N. P., Changes in the levels of translatable
glutaminase mRNA during onset and recovery from metabolic acidosis, Biochemistry,
26, 2773, 1987.
104. Kaiser, S. and Curthoys, N. P., Effect of pH and bicarbonate on phosphoenolpyruvate
carboxykinase and glutaminase mRNA levels in cultured renal epithelial cells, J. Bioi.
Chern., 266, 9397, 1991.
lOS. Schimke, R. and Doyle, D., Control of enzyme levels in animal tissues, Annu. Rev.
Biochem., 39, 929, 1970.
106. Morris, S. M., Moncman, C. L., Rand, K. D., Dizikes, G. J., Cederbaum, S. D.,
and O'Brien, W. E., Regulation of mRNA levels for five urea cycle enzymes in rat
liver by diet, cyclic AMP and glucocorticoids, Arch. Biochem. Biophys., 256. 343, 1987.
107. Bond, J. S., Failla, M. L., and Unger, D. F., Elevated manganese concentration and
arginase activity in livers of streptozotocin-induced diabetic rats, J. Bioi. Chern., 258,
8004, 1983.
352 Nutrition and Gene Expression
108. Schimke, R. T., The importance of both synthesis and degradation in the control of
arginase levels in rat liver, J. Bioi. Chern .. 239. 3808, 1964.
109. vanRoon, M.A., Zonneveld, D., Charles, R., and Lamers, W. H., Accumulation of
carbamoylphosphate synthetase and phosphoenolpyruvate carboxykinase mRNA in em-
bryonic rat hepatocytes. Evidence for translational control during the initial phases of
hepatocyte specific gene expression in vitro. Eur. J. Biochern., 178. 191, 1988.
110. Nebes, V. L. and Morris, S. M., Regulation of messenger ribonucleic acid levels for
five urea cycle enzymes in cultured rat hepatocytes. Requirements for cyclic adenosine
monophosphate, glucocorticoids and ongoing protein synthesis, Mol. Endocrinol., 2,
444, 1988.
Ill. Jackson, M. J., Beaudet, A. L., and O'Brien, W. J., Mammalian urea cycle enzymes,
Annu. Rev. Genet., 20, 431, 1986.
112. Takiguchi, M., Matsubasa, T., Amaya, Y., and Mori, M., Evolutionary aspects of
urea cycle genes, BioEssays, 10, 163, 1989.
113. Boyce, F. M., Anderson, G. M., Rusk, C. D., and Freytag, S. 0., Human argini-
nosuccinate synthetase minigenes are subject to arginine mediated repression but not to
trans induction, Mol. Cell. Bioi., 6, 1244, 1986.
114. Jackson, M. J., O'Brien, W. E., and Beaudet, A. L., Arginine-mediated regulation
of an argininosuccinate synthetase minigene in normal and canavanine resistant human
cells, Mol. Cell. Bioi., 6, 2257, 1986.
115. Engelhardt, J, F., Steel, G., and Valle, D., Transcriptional analysis of the ornithine
aminotransferase promoter, J. Bioi. Chern., 266, 752, 1990.
Chapter 16
TABLE OF CONTENTS
0-8493-6961-4193/$0.00 + $.50
© 1993 by CRC Press, Inc. 353
354 Nutrition and Gene Expression
When meals that contain protein are ingested, the liver is exposed to
elevated concentrations of amino acids and hormones in the blood of the
hepatic portal vein. This produces two important effects that alter the regu-
lation of specific genes. The first of these effects is an increase in the size
of the liver and kidneys. Hepatic and renal cells proliferate, and this involves
an increase in DNA synthesis, RNA synthesis, and the activation of enzymes
needed for synthesis of cellular constituents. '·2 In addition to cell division,
cell growth occurs with accumulation of protein. The second effect is an
increase in the concentration of enzymes needed to catabolize the excess
amino acids, dispose of the unneeded amino group, and utilize the resulting
carbon chain. 3 . 6 Due to this adaptation, the concentration of amino acids in
the general circulation does not increase to the degree noted in the portal
circulation.
Although amino acids can be converted to glucose through the processes
of trans- or deamination and gluconeogenesis, fatty acids do not fuel glu-
coneogenesis efficiently in liver. This causes dietary protein and carbohydrate
to have a special, opposing relationship on the synthesis of enzymes involved
in amino acid catabolism. Fasting or consuming diets that contain relatively
large amounts of protein causes adaptive increases in the concentrations of
enzymes in liver and kidney that convert amino acids to precursors for syn-
thesis of glucose and fatty acids; 4 · 6 conversely, dietary carbohydrate decreases
the activity of enzymes that participate in gluconeogenesis and amino acid
catabolism. 7•8 The ability of carbohydrates to decrease the synthesis of en-
zymes that participate in catabolism of amino acids is superficially similar to
catabolite repression in bacteria and has been referred to as glucose repression.
These responses have been characterized best in animals fed experimental
diets that provide extreme levels of protein or carbohydrate and produce large
changes in enzyme levels. Although these diets are extreme in comparison
to the mixed diets typical of North Americans, adaptive changes in gene
expression probably occur in response to diets of any composition. In the
words of Helen Teppermann et a!., 9 "the business of differentiation in the
liver is eternally unfinished", and this comment no doubt applies to cells of
the endocrine system and central nervous system that contribute to nutrient-
initiated signaling.
The means by which the diet affects genetic activity probably differs for
individual organs, and depends on the duration of any prior fast and the
composition of the ensuing meals. Levels of nutrients in the blood plasma
are significant even during starvation, and their concentrations increase only
moderately during the absorptive phase. For example, it is unusual for the
concentration of glucose or amino acids in the general circulation to increase
by more than 50% after a meal in nondiabetic subjects.' 0 · 13 The concentration
Chan and Hargrove 355
The liver and kidneys both play important roles in regulating levels of
amino acids in the blood plasma and in synthesizing glucose from excess
amino acids. Perhaps for this reason, dietary protein promotes growth of these
organs more strongly than it stimulates growth of the rest of the body. This
topic has received considerable attention due to the concern that excessive
amino acids may cause kidney function to deteriorate; 15 in addition, excess
amino acids stimulate kidney growth, and this could be beneficial in some
cases. 2 Depending on the level of protein, the weight of these organs in rats
may increase from 10 to 50% over a period of several days, and the rate of
DNA replication increases, as judged by incorporation of radiolabeled de-
oxythymidine. 16 The amount of protein relative to DNA also increases, sug-
gesting that protein or amino acids promote both hypertrophy and hyperplasia;
hyperplasia predominates in young rats and hypertrophy predominates in older
onesY In addition to their effects on organ growth, amino acids rapidly
increase the rate of renal blood flow (hyperemia) and glomerular filtration
rate (GFR). 15 ·18 When the subjects are refed diets containing lower levels of
protein, the blood flow and organ weights return to normal values.
Amino acids are thought to affect the function of kidney and liver by
direct and indirect mechanisms. Mixtures of amino acids that cause renal
hyperemia also stimulate secretion of insulin, glucagon, and growth hor-
mone.18 When infused alone, glucagon increases GFR. However, a mixture
of branched-chain amino acids increased secretion of insulin and growth
hormone without affecting GFR. 18 This evidence suggests that glucagon par-
ticipates in the hyperemic response to dietary protein. In addition to the
hormonal responses, amino acids may generate other mediators that affect
organ function. Arginine not only stimulates glucagon secretion, but is con-
verted to nitric oxide by the vascular endothelium. Nitric oxide increases
blood flow and alters levels of cyclic guanosine 3' ,5 '-monophosphate in
glomeruli. 19 It counteracts the action of vasoconstrictors, such as the local
356 Nutrition and Gene Expression
TABLE I
Effects of Energy-Yielding Nutrients on Hormone Secretion
Gut
Neurotensin o· 0 ++ 25
Peptide YY + + ++ 26
Gastric inhibitory peptide ++ 01+ ++ 27
Cholecystokinin 01+ ++ ++ 28
Pancreas
Insulin ++ + +10 29-32
Glucagon ++ 24, 32-34
Pancreatic polypeptide +1-b ++ 26
Anterior pituitary
Growth hormone ++ 23, 34-37
Adrenocorticotropin 0/-b ++ 0 38-42
Thyroid stimulating hormone 43
Adrenal
Cortisol + ++ 0 32, 39--41
Thyroid gland
T, + 0 32, 40, 44
T. + 0 44
T,IT4 + 0
cretion during the initial phase, but changes that occur after refeeding depend
on composition of the diet, and the hormones participate in activating specific
genes. Table I summarizes effects of glucose, amino acids, and fatty acids
on secretion of a variety of hormones from the gut, pancreas, and anterior
pituitary gland. It is evident that each class of energy-yielding macronutrient
produces a distinct pattern of hormone secretion from the intestinal mucosa,
pancreas, and pituitary. Peptide hormones are gene products of the endocrine
system, and nutrients frequently affect gene expression in hormone-secreting
cells; glucose regulation of transcription from the insulin gene is an important
example. 24
from the intestine and pancreas. and augment secretion of glucagon and
insulin; dietary protein or amino acids also increase secretion of growth hor-
mone, adrenocorticotropin (ACTH), and cortisol. Fatty acids have minor
effects on insulin and glucagon but strongly stimulate secretion of neurotensin,
peptide YY, gastic inhibitory peptide, and cholecystokinin.
Tissues that contain receptors for these peptide hormones may respond
to diet-induced secretion of the gut peptides. For example, the endocrine and
exocrine pancreas contains receptors for many peptides. 45 Insulin output is
increased by gastric inhibitory peptide, pancreatic polypeptide, neurotensin,
and bombesin, and may be decreased by galanin. The liver contains receptors
for secretin, pancreatic polypeptide, and vasoactive intestinal peptide, 46 and
starvation would be expected to decrease the activity of signaling pathways
linked to these receptors. At present, it is not clear to what extent peptides
from the intestinal mucosa affect gene expression in the liver. However,
vasoactive intestinal peptide activates adenylate cyclase, 47 and cyclic adeno-
sine-3'5'-monophosphate (cAMP) regulates numerous hepatic genes. During
the initial phase of starvation in humans and rats, secretion of insulin from
the pancreas declines as levels of glucose and other secretagogues decrease
in the blood plasma. 24 .48 - 51 The output of pancreatic glucagon changes less
than insulin, but fasting causes an initial increase in glucagon secretion in
the human. 50 In both species, the ratio of insulin to glucagon in the blood
plasma declines. Fasting increases the activity of the sympathetic nervous
system and leads to increased secretion of epinephrine from the adrenal med-
ulla. 52 Rising levels of catecholamines and ACTH stimulate output of glu-
cocorticoids from the adrenal cortex and antagonize the action of insulin while
enhancing the action of glucagon on expression of genes that participate in
gluconeogenesis.
Studies done with experimental animals suggest that the relative levels
of carbohydrate and protein in the diet modify the ratio of insulin to glucagon
secreted into the plasma. 53 Glucose is the primary stimulus to insulin secretion
and is necessary for secretion in response to most amino acids; in contrast,
glucose inhibits glucagon secretion, whereas amino acids stimulate this pro-
cess. 33 In portal blood, the ratio of glucagon to insulin was correlated with
the concentrations of total amino acids and branched-chain plus aromatic
amino acids. 53 Diets containing high carbohydrates produce different meta-
bolic effects on liver than diets with high protein content. With mixed or high
carbohydrate diets, the level of glycogen in liver increases during the feeding
period, and cyclic AMP levels remain low. However, diets containing elevated
protein stimulate production of cAMP, and little glycogen is deposited. 54 This
is confirmed by measurements of enzyme activities that are regulated in
opposing fashion by insulin compared to glucagon or cAMP. Diets containing
high protein caused phosphoenolpyruvate carboxykinase (PEPCK) levels to
increase, whereas pyruvate kinase declined, and carbohydrates promoted the
opposite effect. 55
Chan and Hargrove 359
reproductive cycles cease, and the metabolic rate declines because the secre-
tion of thyroid-stimulating hormone, prolactin, luteinizing hormone, and fol-
licle-stimulating hormone is depressed. Decreased secretion of gonadotropic
hormones represses the preovulatory surge of LH secretion in the female and
causes a decline in output of gonadal steroids in both sexes. Sex steroids are
anabolic agents, and their decreased secretion reduces the capacity to maintain
synthesis of proteins that are essential to normal functions of muscle and
bone. In the rat, secretion of GH declines, whereas in humans output of GH
increases during fasting or hypoglycemia.
The response of the thyroid gland to stimulation by thyrotropin diminishes
with food deprivation in man and experimental animals. 44 •57 Although plasma
concentrations of thyroid-stimulating hormone (TSH) may not differ greatly,
levels of 3,5,3' -triiodothyronine (T3 ) and T4 decline during long-term food
deprivation, and TSH levels do not rise to compensate. Pituitary function is
affected, as indicated by a lower level of mRNA for thyroid-stimulating
hormone in the anterior pituitary gland of rats. The mRNA for thyrotropin-
releasing hormone in the hypothalamus declines in rats. 58
Acute hypoglycemia, which can be caused by injecting insulin, increases
the secretion of counter-regulatory hormones from the pituitary gland, in-
cluding ACTH, GH, and prolactin. This effect has been postulated to involve
glucoreceptors in specific areas of the brain, and is thought to reflect the lack
of glucose and not the presence of insulin. For instance, administering suf-
ficient glucose to prevent hypoglycemia inhibited the release of ACTH and
360 Nutrition and Gene Expression
Q)
"'
c::
0
100
•••••
•• ......
............
.......... r/:l.
h\
I
Q.
Ill
Q)
•o ............... 4····· Serine
80
a: ••••• •••• De hydratase
. ·•· ··...
.... . ·· ··.•.·.
E
::l
60
E #' ••
)(
ca IS. ••••••
--
,i'
:::E
0
40
. ······ ·•···•. •
0
c::
Q)
20
A~
. /
/ Glucose 6-Phosphate~··.
Dehydrogenase •••• .b
...
Q) 0 ···············IS.
a.. 0 20 40 60 80 100
FIGURE 1. Patterns of induction by dietary protein differ for various enzymes and peptides.
The threshold of induction is quite low for xanthine oxidase (squares) and glucose-6-phosphate
dehydrogenase (circles), but is substantially higher for serine dehydratase (triangles) and most
transaminases (not shown). Maximal expression of xanthine oxidase occurs at a dietary casein
level of about 20%, whereas serine dehydratase continues to increase in proportion to protein
consumed, and glucose-6-phosphate declines at high levels of protein. Data were replotted from
articles by Harper,' Potter and Ono," and Ogawa et al 82
-...
c 80
Q) A. Glucokinase
0 70
Q,
60
Dl
-
E 50
Iii
c 40
::::>
-
>- 30
-
>
u
c(
20
10
Fasting
0 2 3 4
-...
Q)
0
Q,
200
B. PEP Carboxykinase
150
E Fasting
...
as
-
Dl
._
ltl 100
c
::::>
->- 50
->
u
c( 0
Refeeding
Time (Days)
whereas levels of PEP carboxykinase increase (Figure 2). 84 ·85 After feeding,
the level of glucokinase increases and PEP carboxykinase decreases in pro-
portion to dietary carbohydrate. In contrast, dietary protein fails to produce
an increase in glucokinase in fasted rats, and increases the activity of PEP
carboxykinase (Figure 2). The decline in PEP carboxykinase activity results
364 Nutrition and Gene Expression
?:-
'
.:: 100
u
C( A
I
I
I
80 Birth I
I
Promoter Status: I
1
I
I
60 I
I
Inactive Active Inducible I
I
I
40
~ 20
Ill
~
~ o6-~~-o~~~~~~~~~~~~~~~~-r~~~
·10 .g ·8 ·7 ·6 ·5 ·4 ·3 ·2 ·1 0 2 3 4
Days
?:- Fasted
,.,.
·;: 200
60%
u
C( B h.
I \ I \
I \ I \
I \ I \
I \ I \
I \ I \
I \
-.
I \
I \
I \ I \
I \
I \
,.
100 I 30% \
I \ I \
: ....'f~ '\ i. ...........~ ~
I
~
.....•
.•· o •.....
~
...._.._.._.
1 e 12Yo •.. ,. 1
1
rl
................. :.~ ... ..
Ill I •" ... ._.._ I ;
c ~-··
I •
"'0
>.
1-
0 6 12 18 24 30 36 42 48
Hours Elapsed
FIGURE 3. Regulation of the gene encoding the enzyme, tyrosine aminotransferase, is typical
of mammalian genes that are induced by dietary protein. Panel A: Acquisition of the capacity
to synthesize this enzyme and its mRNA occurs in liver during the latter third of gestation.
Enzyme activity is indicated by the open circles. At the time of birth (arrow), synthesis begins
at a high rate in support of gluconeogenesis, and the capacity to respond to diet and hormones
begins. Symbols indicate the following treatments: injection of fetuses with cyclic AMP (closed
circles) of hydrocortisone (open squares); injection of neonates with hydrocortisone (closed
squares). Panel B: The quantity of enzyme in adult liver undergoes a diurnal cycle because
mRNA synthesis responds to feeding and the level of protein in the diet. Tissue from animals
that are fed a diet containing 12% protein contains very little TyrAT, and the diurnal variation
is small (open circles). The variation becomes much larger when dietary protein makes up 30%
(closed circles) or 60% (triangles) of the diet by weight. Diurnal rhythms of this magnitude
require that the enzyme and its mRNA are both unstable. as predicted by the kinetic theory
discussed in Chapter I. The figure is a composite drawn from data reported in References. 90· 9 '
366 Nutrition and Gene Expression
~ 6
>-.::' A. Serine Dehydratase
_.c
u >. 5
<-o0
.a 4
CD
:: E
~ Cl 3
-gO
>-0 Casein hydrolyzate
.c!:
CD !II 2
c=c: Casein hydrolyzate
::::)
CD
plus glucose
...c:
CD
~ ~
(/)
0 6 12 1B 24
Time (Hours)
120
CD B. Ornithine Aminotransferase
!II
...cu
-
CD
!II
100
-... -...
c:
cu ~BO
0 c:
-
c: 00 60
E
<
-
0
'#. 40
CD
c:
-...
.c
c:
0
20
0
0 5 10 15 20 25
Concentration (mM)
FIGURE 4. Glucose interferes with the ability of protein to increase the synthesis of several
enzymes of amino acid catabolism in liver. (A) Serine dehydratase is induced by dietary protein
or by hydrolyzed casein, administered by gastric incubation to intact rats at the times indicated
by arrows (closed circles). If I g of glucose is included along with the casein, the induction is
prevented (open circles). (B) Carbohydrate repression of ornithine aminotransferase can be
observed in isolated cells. Hepatocytes were prepared from rats fed a low protein diet and cultured
in the presence of hexoses with insulin added to the culture medium. After 28 h, fresh medium
containing the hexoses and 0.1 rnM dibutyryl cyclic AMP was added, and samples were taken
for assay at 52 h. (Data replotted from Peraino et al. 96 and Merrill and Pitot 101 .)
Chan and Hargrove 367
A second prominent feature of the gene for TyrAT in adult liver is that
it generates enzyme in a diurnal rhythm that is driven by the feeding cycle.
Shown in Figure 4B are results of allowing rats access to diets of different
protein content (12, 30, or 60% by weight) under an inverted light/dark
cycle. 93 The enzyme level begins to rise at the onset of darkness after feeding
is initiated, but begins to decline soon after food is removed. This type of
circadian rhythm is a prominent feature of short-lived gene products that
respond to nutrients. In the case ofTyrAT, the immediate stimulus that causes
the increased rate of synthesis is not totally clear; it is likely to involve changes
in levels of insulin, glucagon, and adrenal steroids in the blood plasma in
addition to elevated amino acid content. Recent evidence suggests that the
nutrient-related induction of TyrAT that occurs during feeding is a result of
altered production of mRNA, as judged by quantitative Northern analysis. 94
The low amplitude of the rhythm observed when animals are fed low protein
diets may indicate that carbohydrates decrease the rate at which this enzyme
is synthesized; this effect is noted for a variety of enzymes that initiate
degradation of amino acids. 7
-10 kb -8 kb -6 kb -4 kb -2 kb 0
Region IV III II I
.ill .IIIII II
Dietary Protein * + + *
Dietary Carbohydrate * +
Circadian Variation +
Active in Kidney +
FIGURE 5. DNA elements that respond to dietary protein and that promote circadian variation
in mRNA synthesis have been identified in the promoter for the gene encoding serine dehydratase
by testing for their sensitivity to digestion by DNAse I. DNA that was prepared from rats fed
diets containing elevated casein was readily digested by the nuclease at sites that mapped to
about -3200 and -3800 nucleotides 5' to the start site of transcription (regions II and III).
These sites were not sensitive to nuclease digestion in DNA isolated from animals fed a protein-
free diet, but a site located at positions - 100 to - 200 was more sensitive to digestion (region
I). Sensitivity of DNA in regions I and II varied in circadian fashion, with region II being more
sensitive in the nocturnal feeding period. The figure is based on data reported by Ogawa et al ' 2
VI. CONCLUSIONS
REFERENCES
I. Harper, A. E., Effect of variations in protein intake on enzymes of amino acid metab-
olism, Can. J. Biochem. Physiol., 43, 1589, 1965.
2. Fine, L. G., The role of nutrition in hypertrophy of renal tissue, Kidney Int., 32 (Suppl.
22), S2, 1987.
3. Szepesi, B. and Freedland, R. A., Dietary effects on rat liver enzymes in meal-fed
rats, J. Nutr., 96, 382. 1968.
4. Peters, J. C. and Harper, A. E., Adaptation of rats to diets containing different levels
of protein: effects on food intake, plasma and brain amino acid concentrations and brain
neurotransmitter metabolism, J. Nutr .. 115, 382, 1985.
5. Munro, H. N. and Crim, M. C., The proteins and amino acids, in Modern Nutrition
in Health and Disease, Shils, M. E. and Young, V. R., Eds .. 7th ed., Lea and Febiger,
Philadelphia, 1988, I.
6. Young, V. R. and Marchini, J. S., Mechanisms and nutritional significance of metabolic
responses to altered intakes of protein and amino acids. with reference to nutritional
adaptation in humans, Am. J. Clin. Nutr .. 51. 270, 1990.
7. Soling, H., Kaplan, J., Erbstoeszer, M., and Pitot, H. C., The role of hormones in
glucose repression in rat liver, Adv. Enzyme Regul., 7, 171, 1969.
8. Goldberg, M. L., The glucose effect: carbohydrate repression of enzyme induction.
RNA synthesis, and glucocorticoid activity-a role for cyclic AMP and cyclic GMP.
Life Sci .. 17, 1747, 1975.
9. Teppermann, H. M., Teppermann, J., Pownall, J. D., and Branch, A., On the
response of hepatic glucose-6-phosphate dehydrogenase activity to changes in diet com-
position and food intake pattern, Adv. Enzyme Regul., I. 121. 1963.
10. Harper, A. E., Diet and plasma amino acids, Am. J. Clin. Nutr .. 21. 358, 1968.
II. Ashley, D. V., Barclay, D. V., Chauffard, F. A., Moennoz, D., and Leathwood,
P. D., Plasma amino acid responses in humans to evening meals of differing nutritional
composition, Am. J. Clin. Nutr., 36, 143, 1982.
12. Eisenstein, A. B., Srack, 1., Gallo-Torres, H., Georgiadis, and Miller, 0. N., In-
creased glucagon secretion in protein-fed rats: lack of a relationship to plasma amino
acids, Am. J. Physiol., 236, E20, 1979.
13. Felig, P., Wahren, J., and Hendler, R., Influence of oral glucose ingestion on splanch-
nic glucose and gluconeogenic substrate metabolism in man, Diabetes, 24, 468, 1975.
Chan and Hargrove 371
14. Dole, V. P., James, A. T., Webb, P. W., Rizack, M. A., and Sturman, M. F., The
fatty acid patterns of plasma lipids during alimentary lipemia, J. Clin. Invest., 38, 1544,
1959.
15. Klahr, S., Effects of protein intake on the progression of renal disease, Annu. Rev. Nutr.,
9, 87, 1989.
16. Farwell, D. C., Miguez, J, B., and Herbst, E. J,, Ornithine decarboxylase and po-
lyamines in liver and kidneys of rats on cyclical regimens of protein-free and protein-
containing diets, Biochem. J., 168, 49, 1977.
17. Jakobsson, B., Celsi, G., Lindblad, B., and Aperia, A., Influence of different protein
intake on renal growth in young rats, Acta Paediatr. Scand., 76, 293, 1987.
18. Wada, L., Don, B. R., and Schambelan, M., Hormonal mediators of amino acid-
induced glomerular hyperfiltration in humans, Am. J. Physiol., 260, F787, 1991.
19. King, A., Troy, J,, Anderson, S., Neuringer, J,, Gunning, M., and Brenner, B. M.,
Nitric oxide: a potential mediator of renal hyperemia and hyperfiltration, J. Am. Soc.
Nephrol., !, 1271, 1991.
20. Rosenberg, M. E., Chmielewski, D., and Hostetter, T. H., Effect of dietary protein
on rat renin and angiotensinogen gene expression, J. Clin. Invest., 85, 1144, 1990.
21. Kaysen, G. A., Rosenthal, C., and Hutchinson, F. N., GFR increases before renal
mass or ODC activity increase in rats fed high protein diets, Kidney Int., 36, 441, 1989.
22. Sens, D. A., Levine, J, H., and Buse, M.G., Stimulation of hepatic and renal ornithine
decarboxylase activity by selected amino acids, Metabolism, 32, 787, 1983.
23. Morris, S. M., Jr., Moncman, C. L., Holub, J, S., and Hod, Y., Nutritional and
hormonal regulation of mRNA abundance for arginine biosynthetic enzymes in kidney,
Arch. Biochem. Biophys., 273, 230, 1989.
24. Efrat, S., Surana, M., and Fleischer, N., Glucose induces insulin gene transcription
in a murine pancreatic J3-cellline, J. Bioi. Chern., 266, 11141, 1991.
25. Ferris, C. F., Neurotensin, in Handbook of Physiology, Section 6, The Gastrointestinal
System, Vol. II, Neural and Endocrine Biology, Schultz, S. G., Makhlouf, G. M., and
Rauner, B. B., Eds., Waverly Press, Inc., Baltimore, MD, 1989, 559.
26. Taylor, I. L., Pancreatic polypeptide family: pancreatic polypeptide, neuropeptide Y,
and peptide YY, Op. cit., 1989, 475.
27. Brown, J, C., Buchan, A.M. J,, Mcintosh, C. H. S., and Pederson, R. A., Gastric
inhibitory polypeptide, Op. cit., 1989, 403.
28. Rehfeld, J, F., Cholecystokinin, Op. cit., 1989, 337.
29. Mayhew, D. A., Wright, P. H., and Ashmore, J., Regulation of insulin secretion,
Pharmacal. Rev., 21, 183, 1969.
30. Martin, C. R., Endocrine Physiology, Oxford University Press, Oxford, 1985, 159.
31. Unger, R. H., Dobbs, R. E., and Orci, L., Insulin, glucagon, and somatostatin secretion
in the regulation of metabolism, Annu. Rev. Physiol., 40, 307, 1978.
32. Edozien, J. C., Niehaus, N., Mar, M.-H., Makoui, T., and Switzer, B. S., Diet-
hormone interrelationships in the rat, J. Nutr., 108, 1767, 1978.
33. Rorsman, P., Ashcroft, F. M., and Berggen, P.-O., Regulation of glucagon release
from pancreatic A-cells, Biochem. Pharmacal., 41, 1783, 1991.
34. Castellino, P., Giordano, C., Perina, A., and DeFronzo, R. A., Effects of plasma
amino acid and hormone levels on renal hemodynamics in humans, Am. J. Physiol., 255,
F444, 1988.
35. Pecile, A. and Muller, E. E., Control of growth hormone secretion, in Neuroendocri-
nology, Vol. I, Martini, L. and Ganong, W., Eds., Academic Press, New York, 1966,
537.
36. Matzen, L. E., Andersen, B. B., Jensen, B. G., Gjessing, H. J,, Sindrup, S. H.,
and Kvetny, J,, Different short-term effect of protein and carbohydrate intake on TSH,
growth hormone, (GH), insulin, C-peptide, and glucagon in humans, Scand. J. Clin.
Lab. Invest., 50, 801, 1990.
372 Nutrition and Gene Expression
37. Alvarez, C. V., Mallo, F., Burguera, B., Cacicedo, L., Dieguez, C., and Casanueva,
F. F., Evidence for a direct pituitary inhibition by free fatty acids of in vivo growth
hormone responses to growth hormone-releasing hormone in the rat, Neuroendocrinology,
53, 185, 1991.
38. Vigas, M., Tatar, P., Jurcovicova, J,, and Jezlova, D., Glucoreceptors located in
different areas mediate the hypoglycemia-induced release of growth hormone, prolactin,
and adrenocorticotropin in man, Neuroendocrinology, 51, 365, 1990.
39. Munro, H. N., Steele, M. H., and Hutchison, W. C., Action of dietary proteins and
amino acids on the rat adrenal gland, Br. J. Nutr., 19, 137, 1965.
40. Rabolli, D. and Martin, R. J,, Effects of diet composition on serum levels of insulin,
thyroxine, triiodothyronine, growth hormone, and corticosterone in rats, J. Nutr., 107,
1068, 1977.
41. Slag, M. F., Ahmed, M., Gannon, M. C., and Nuttall, F. Q., Meal stimulation of
cortisol secretion: a protein induced effect, Metabolism. 30, 1104, 1981.
42. Aizawa, T., Yasuda, N., and Greer, M.A., Hypoglycemia stimulates ACTH secretion
through a direct effect on the basal hypothalamus, Metabolism, 30, 996, 1981.
43. Jepson, M. M., Bates, P. C., and Millward, D. J,, The role of insulin and thyroid
hormones in the regulation of muscle growth and protein turnover in response to dietary
protein in the rat, Br. J. Nutr., 59, 397, 1988.
44. Danforth, E., Jr. and Burger, A. G., The impact of nutrition on thyroid hormone
physiology and action, Annu. Rev. Nutr., 9, 201, 1989.
45. Gardner, J, D. and Jensen, R. T., Receptors for gut peptides and other secretagogues
on pancreatic acinar cells, in The Gastrointestinal System, Handbook of Physiology,
Section 6, Vol. 2, Schultz, S. G., Makhlouf, G. M., and Rauner, B. M., Eds., American
Physiological Society, 1989, 171.
46. Rosselin, G., Liver receptors for regulatory peptides, Op. cit., 245.
47. Sanchez, V., Governa, R., and Calvo, J. K., Glycogenolytic effect of vasoactive
intestinal peptide in the rat in vivo, Experientia, 47, 625, 1991.
48. Becker, D. J,, The endocrine responses to protein calorie malnutrition, Annu. Rev. Nutr.,
3, 187, 1983.
49. Goodman, M. N., Larsen, P. R., Kaplan, M. M., Aoki, T. T., Young, V. R., and
Ruderman, N. B., Starvation in the rat. II. Effect of age and obesity on protein sparing
and fuel metabolism in the rat, Am.]. Physiol., 239, E277, 1980.
50. Xie, Q.-W., Experimental studies on changes of neuroendocrine functions during star-
vation and refeeding, Neuroendocrinology, 53 (Suppl. 1), 52, 1991.
51. Thomas, G. B., Mercer, J, E., Karalis, T., Rao, A., Cummins, J, T., and Clarke,
I. J., Effect of restricted feeding on the concentrations of growth hormone, gonadotropins,
and prolactin in plasma, and on the amounts of messenger ribonucleic acid for GH,
gonadotropin subunits, and PRL in the pituitary glands of adult ovariectomized ewes,
Endocrinology, 126, 1361, 1990.
52. Sakaguchi, T., Takahashi, M., and Bray, G. A., Diurnal changes in sympathetic
activity, J. Clin. Invest., 82, 282, 1988.
53. Jarrousse, C., Lardeux, B., Bourdel, G., Girard-Globa, A., and Rosselin, G., Portal
insulin and glucagon in rats fed proteins as a meal: immediate variations and circadian
modulations, J. Nutr., 110, 1764, 1980.
54. Tiedgen, M. and Seitz, H. J., Dietary control of circadian variations in serum insulin,
glucagon, and hepatic cyclic AMP, J. Nutr., 110, 876, 1980.
55. Peret, J., Foustock, S., Chanez, M., Bois-Joyeux, B., and Assani, R., Plasma glu-
cagon and insulin concentrations and hepatic phosphoenolpyruvate carboxyk:inase and
pyruvate kinase activities during adaptation of rats to a high protein diet, J. Nutr., Ill,
1173, 1981.
56. Kuhara, T., Ikeda, S., Ohneda, A., and Sasaki, Y., Effects of intravenous infusion
of 17 amino acids on the secretion ofGH, glucagon, and insulin in sheep, Am. J. Physiol.,
260, E21, 1991.
Chan and Hargrove 373
57. Hugues, J.-N., Enjalbert, A., Burger, A., Voirol, M.-J., Sebaoun, J,, and Epelbaum,
J., Sensitivity of thyrotropin (TSH) secretion to 3,5 .3 '-triiodothyronine and TSH-releasing
hormone in rat during starvation, Endocrinology. 119, 253, 1986.
58. Blake, N. G., Eckland, D. J, A., Foster, 0. J, F., and Lightman, S. L., Inhibition
of hypothalamic thryrotropin-releasing hormone messenger ribonucleic acid during food
deprivation, Endocrinology, 129, 2714, 1991.
59. Straus, D. S. and Takemoto, C. D., Effect of fasting on insulin-like growth factor-!
(IGF-1) and growth hormone receptor mRNA levels and IGF-1 gene transcription in rat
liver, Mol. Endocrinol., 4, 91, 1990.
60. Thissen, J, P., Triest, S., Underwood, L. E., Maes, M., and Ketelslegers, J, M.,
Divergent responses of serum insulin-like growth factor-! and liver growth hormone
receptors to exogenous GH in protein-restricted rats, Endocrinology, 126, 908, 1990.
61. Plotsky, P. M. and Vale, W. W., Patterns of growth hormone-releasing factor and
somatostatin secretion into the hypophyseal-portal circulation of the rat. Science, 230,
461' 1986.
62. Bruno, J, F., Olchovsky, D., White, J, D., Leidy, J, W., Song, J,, and Berelowicz,
M., Influence of food deprivation in the rat on hypothalamic expression of growth
hormone-releasing factor and somatostatin, Endocrinology, 127, 2111, 1990.
63. Bruno, J. F., Song, J,, and Berelowitz, M., Regulation of rat hypothalamic prepro-
growth hormone-releasing factor messenger ribonucleic acid by dietary protein, Endo-
crinology, 129, 1226, 1991.
64. Glaeser, B. S., Maher, T. J,, and Wortman, R. J,, Changes in brain levels of acidic,
basic, and neutral amino acids after consumption of single meals containing various
proportions of protein, J. Neurochem., 41, 1016, 1983.
65. Knopf, R. F., Conn, J. W., Fajans, S. S., Floyd, J. C., Guntsche, E. M., and Rull,
J. A., Plasma growth hormone response to intravenous amino acids, Am. J. Endocrinol.,
26, 1140, 1965.
66. Felig, P., Progress in endocrinology and metabolism. The glucose-alanine cycle, Me-
tabolism, 22, 179, 1973.
67. Kayne, F. J, and Price, N.C., Amino acid effector binding to rabbit muscle pyruvate
kinase, Arch. Biochem. Biophys., 159, 292, 1973.
68. Dong, F. M. and Freedland, R. A., Effects of alanine on gluconeogenesis in isolated
rat hepatocytes, J. Nutr., 110, 2341, 1980.
69. Perez-Sala, D., Parrila, R., and Ayuso, M. S., Key role of L-alanine in the control
of hepatic protein synthesis, Biochem. J., 241, 491, 1987.
70. Poso, A. R. and Mortimore, G. E., Requirement for alanine in the amino acid control
of hepatic protein degradation, Proc. Nat/. Acad. Sci. U.S.A., 81, 2039, 1984.
71. Muller, W. A., Faloona, G. R., and Unger, R. H., The effect of alanine on glucagon
secretion, J. Clin. Invest., 50, 2215, 1971.
72. Walser, M., Role of branched-chain ketoacids in protein metabolism, Kidney Int., 38,
595, 1990.
73. MacDonald, M. J,, McKenzie, D. I., Kaysen, J, H., Walker, T. M., Moran, S.M.,
Fabien, L. A., and Towle, H. C., Glucose regulates leucine-induced insulin release
and the expression of the branched chain ketoacid dehydrogenase E Ia subunit gene in
pancreatic islets, J. Bioi. Chern., 266, 1335, 1991.
74. Garthwaite, J, and Garthwaite, G., Mechanisms of excitatory amino acid neurotoxicity
in rat brain slices, Adv. Exp. Bioi. Med., 268, 505, 1990.
75. Pestana, A., Dietary and hormonal control of enzymes of amino acid catabolism in liver,
Eur. J. Biochem., 11, 400, 1969.
76. Lardy, H. A. and Merryfield, M. L., Ferroactivator and the regulation of gluconeo-
genesis, Curr. Top. Cell. Reg., 18, 243, 1981.
374 Nutrition and Gene Expression
96. Peraino, C., Lamar, C., Jr., and Pitot, H. C., Studies on the induction and repression
of enzymes in rat liver. IV. Effects of cortisone and phenobarbital, J. Bioi. Chem., 241,
2944, 1966.
97. Anderson, H. L., Benevenga, N. J., and Harper, A. E., Associations among food
and protein intake, serine dehydratase, and plasma amino acids, Am. J. Physiol., 214,
1008, 1968.
98. Jost, J, P., Khairallah, E. A., and Pitot, H. C., Studies on the induction and repression
of enzymes in rat liver. Regulation of the rate of synthesis and degradation of serine
dehydratase by dietary amino acids and glucose, J. Bioi. Chem., 243, 3057, 1968.
99. Noda, C., Yakiyama, M., Nakamura, T., and lchihara, A., Requirements of both
glucocorticoids and glucagon as co-inducers for activation of transcription of the serine
dehydratase gene in cultured rat hepatocytes, J. Bioi. Chem., 263, 14764, 1988.
100. Sudilovsky, 0., Pestana, A., Hinderaker, P. H., and Pitot, H. C., Cyclic adenosine
3' ,5'-monophosphate during glucose repression in rat liver, Science, 174, 142, 1971.
101. Merrill, M. J, and Pitot, H. C., Inhibition of cyclic AMP-dependent induction of
ornithine aminotransferase by simple carbohydrates in cultured hepatocytes, Arch. Biochem.
Biophys., 259, 250, 1987.
102. Towle, H. C., Thompson, K. S., Liu, J., and Shih, H.-M., Carbohydrate metabolism
regulates gene transcription in liver: identification of response elements and nuclear
factors, FASEB J., 6, A402, 1992.
103. Castro, E. C., Nutrient effects on DNA and chromatin structure, Annu. Rev. Nutr., 7,
407, 1987.
Chapter 17
TABLE OF CONTENTS
I. Introduction ...................................................... 378
0-8493-6961-4/93/$0 ()()+$.50
© 1993 by CRC Pre!'.s, Inc. 377
378 Nutrition and Gene Expression
I. INTRODUCTION
4
A TRANSCRIPTION RATE
3
~
0 6 12 18 24 48 72
I.J..J
(J)
<1:
I.J..J
a:
u
z:
a_.
0
LL.
0 6 12 18
CALBINOIN-D28K
5 PROTEIN
4
0 6 12 18 24 72
TIME AFTER 1,25(0Hl 2D3 INJECTION
FIGURE 1. Analysis of rat renal calbindin-D 28 k transcription, mRNA, and protein levels in
kidney after 1,25(0H),D, treatment. (A) Calbindin-D gene transcription at various times after
1,25(0H),D3 administration (200 ng/100 g body weight to vitamin D-deficient rats). The tran-
scription rate was measured using the nuclear transcription assay46 and expressed as fold increase
compared to control. The results shown are the mean of four different experiments; (B) The
time course of increase in calbindin-D 28 k mRNA levels following 1,25(0H)2 D, administration.
Northern blots of poly(A +)RNA isolated from kidneys of vitamin D-deficient rats at various
times after single injection of 1,25(0H) 20 3 were quantitated by densitometric analysis. 44 The
signal intensities were adjusted for variations in J3-actin densities and the fold increase in mRNA
levels compared to control was calculated. The values shown are the mean of three separate
Northern hybridization experiments: (C) The time course of calbindin-D 28 k accumulation after
1,25(0H)2 0 3 treatment. The levels of calbindin-D at various times were measured by immu-
nobinding assay 44 and expressed as fold increase compared to control. Each value represents the
mean of three separate groups of rats.
Gill and Christakos 381
A B
AGE (Wks)
2 3 4 5
CALBINOIN- OgK mRNA
,i
I I
I I
I I
I I
CALBINOIN- OgK I I
I
~I I
1
>- I
..... I
Vi I
.........z .I
VDR I
~
-' I
<[ I
z I
'"'Vi I
....> I
;::: I
<[
I
....0::-' I
I
I
I
I
I
IS S rRNA I
I VDR mRNA
I
I
I
/
ft
/
0
0 2 3 4 5
AGE (Wks)
FIGURE 2. Developmental changes in intestinal calbindin-D 9, mRNA and VDR mRNA as-
sessed by Northern blot hybridization. (A) Rat intestinal poly(A +)RNA (5 j.Lg per lane) was
fractionated through a formaldehyde-agarose gel. transferred onto a nylon membrane, and hy-
bridized with calbindin-D9,, VDR, and 18S rRNA probes: (B) densitometric quantitation of
Northern analysis. The results represent the mean :!: SE of three separate Northern hybridization
analyses from different groups of rats. The data were normalized based on results obtained upon
rehybridization with 18S rRNA eDNA. (From Huang, et al., J. Bioi. Chern .. 264, 17454, 1989.
With permission.)
epithelial mesenchymal tooth germ of erupting rat incisor, indicated the pres-
ence of both calbindin-D 9 k and calbindin-D 28 k mRNA in this tissue. 10 The
mRNA for both calbindins was nearly undectectable in vitamin D deficient
rats but was upregulated by I ,25(0H) 2 D 3 injection. These findings, although
suggestive of a direct role of I ,25(0H)zD 3 regulated calbindin gene expression
in the mineralization process, are still preliminary. Further studies are needed
to verify the vitamin D dependence of calbindin in chondrocytes and ame-
loblasts.
0 0 0
200
A B
2001-
c
2001-
."'
;z
::>
::t:
400 f-
."'
;z
::>
::t:
4001-
><
u
'r
u
400
600 ~
6001- 600 f-
800 1- \ \
8001- 800f-
1000 I I I I I I I I I I I I
1000 1000
0 250 500 750 1000 1250 0 250 500 750 1000 1250 0 250 500 750 1000 1250
MOUSE CHICK MOUSE
FIGURE 3. Dot matrix analysis showing the homologies between human (- 840/ +!54),
mouse ( -1075/ + 154), and chick (- 850/ + 154) calbindin-D 28 k gene promoter regions. Each
dot indicates a minimum of 17 base pair matches out of 20. The coding regions are in the lower
right comer.
FIGURE 4. Sequence homology between the VDRE of the rat osteocalcin gene and sequence
- 198/169 of the mouse calbindin-D 28 , promoter.
REFERENCES
I. DeLuca, H. F., The vitamin D story: a collaborative effort of basic science and clinical
medicine, FASEB J., 2, 224, 1988.
2. Henry, H. L. and Norman, A. W., Vitamin D: metabolism and biological action, Annu.
Rev. Nutr., 4, 493, 1984.
3. Christakos, S., Gabrielides, C., and Rhoten, W. B., Vitamin D-dependent calcium
binding proteins: chemistry, distribution, functional considerations and molecular biology,
Endocrine Rev., 10, 3, 1989.
4. Feher, J, J,, Facilitated calcium diffusion by intestinal calcium binding protein, Am. J.
Physiol., 244, C303, 1983.
5. Bronner, F., Pansu, D., and Stein, W. D., An analysis of intestinal calcium-transport
across the rat intestine, Am. J. Physiol., 250, G561, 1986.
6. Christakos, S. and Norman, A. W., A vitamin D-dependent calcium binding protein
in bone, Science, 202, 70, 1978.
7. Balmain, N., Brehier, A., Cuisinier-Gieizes, P., and Mathieu, H., Evidence for the
presence of calbindin-D 28 , (CaBP28•) in the tibial growth cartilage of rats, Cell Tissue
Res., 245, 331, 1986.
8. Zhou, X. Y., Dempster, D. W., Marion, S. L., Pike, J, W., Haussler, M. R., and
Clemens, T. L., Bone vitamin D-dependent calcium binding protein is localized in
chondrocytes of growth plate cartilage, Calcif. Tissue Int., 38, 244, 1986.
9. Elms, T. N. and Taylor, A. N., Calbindin-D 28 k localization in rat molars during oden-
togenesis, J. Dent. Res., 66, 1431, 1987.
10. Berdal, A., Dupret, J, M., Pike, J, W., and Mathieu, H., Vitamin D controlled
expression of calbindin genes in teeth, Abstracts, 8th Workship on Vitamin D, 1991, 206.
II. Morrisey, R., Bucci, T., Empson, R., and Lutkin, E., Calcium binding protein: its
cellular localization in jejunum, kidney and pancreas, Proc. Soc. Exp. Bioi. Med., 149,
56, 1975.
Gill and Christakos 387
12. Christakos, S., Friedlander, E. J., Frandsen, B. R., and Norman, A. W., Devel-
opment and application of a radioimmunoassay for chick intestinal calcium binding protein
and tissue distribution, Endocrinology, 104, 1495, 1979.
13. Sonnenberg, J., Pansini, A. R., and Christakos, S., Vitamin D-dependent rat renal
calcium binding protein: development of a radioimmunoassay tissue distribution and
immunological identification, Endocrinology, 115, 640, 1984.
14. Roth, J., Bonner-Weir, S., Norman, A. W., and Orci, L., Immunocytochemistry of
vitamin D-dependent calcium binding protein in chick pancreas: exclusive localization in
~cells, Endocrinology, Ill, 2216, 1982.
15. Rhoten, W. B. and Christakos, S., Vitamin D dependent calcium binding protein is
present in mammalian~ cells, Diabetes, 32, 130A, 1983.
16. Corradino, R. A., Wasserman, R. H., Pubolos, M. H., and Chang, S. 1., Vitamin
0 3 induction of a calcium binding protein in the uterus of the laying hen, Arch. Biochem.
Biophys., !25, 378, 1968.
17. Delorme, A. C., Dana, J. L., Acker, M.G., Ripoche, M.A., and Mathieu, H., In
rat uterus, 17~ estradiol stimulates a calcium binding protein similar to duodenal vitamin
D dependent calcium binding protein, Endocrinology, 113, 1340, 1983.
18. Bruns, M. E., Overpeck, J. G., Smith, G. C., Hirsch, G. N., Mills, S. E., and
Bruns, D. E., Vitamin D-dependent calcium binding protein in rat uterus: differential
effects of estrogen, tamoxifen, progesterone and pregnancy on accumulation and cellular
localization, Endocrinology, 122, 2371, 1988.
19. Delorme, A. C., Cassier, P., Geny, B., and Mathieu, H., Immunocytochemical lo-
calization of vitamin D-dependent calcium binding protein in the yolk sac of the rat,
Placenta, 4, 263, 1983.
20. Bruns, M. E., Kleeman, E., and Bruns, D. E., Vitamin D-dependent calcium binding
protein in mouse yolk sac: biochemical and immunochemical properties and response to
I ,25-dihydroxycholecalciferol, J. Bioi. Chem., 261, 7485, 1986.
21. Taylor, A. N., Chick brain calcium binding protein: response to cholecalciferol and some
developmental aspects, J. Nutr., 107, 480, 1977.
22. Jande, S. S., Maler, L., and Lawson, D. E. M., Immunohistochemical mapping of
vitamin D dependent calcium binding protein in brain, Nature, 294, 765, 1981.
23. Roth, J., Baeten, D., Norman, A. W., and Garda-Segura, L. M., Specific neurons
in chick central nervous system stain with an antibody against intestinal vitamin D-
dependent calcium binding protein, Brain Res., 222, 452, 1981.
24. Feldman, S. C. and Christakos, S., Vitamin D-dependent calcium binding protein in
rat brain: biochemical and immunocytochemical characterization, Endocrinology, 112,
290, 1983.
25. Celio, M., Calbindin-D 28k and parvalbumin in the rat nervous system, J. Neuroscience,
35, 375, 1990.
26. Parmentier, M., Ghysens, M., Rysen, F., Lawson, D. E. M., Pasteels, J. L., and
Pochet, R., Calbindin in vertebrate classes: immunohistochemical localization and West-
em blot analysis, Gen. Comp. Endocrinol., 65, 399, 1987.
27. Christakos, S., Malkowitz, L., Sori, A., Sperduto, A., and Feldman, S.C., Calcium
binding protein in squid brain: biochemical similarity to the 28,000 M, vitamin D de-
pendent calcium binding protein, J. Neurochem., 49, 1427, 1987.
28. Christakos, S. and Norman, A. W., Vitamin D dependent calcium binding protein and
its relation to 1,25-dihydroxyvitamin D receptor localization and concentration, in Cal-
cium Binding Proteins and Calcium Function, Siegel, F. et al., Eds., Elsevier, New
York, 1980, 371.
29. Thorens, B., Roth, j., Norman, A. W., Perrelet, A., and Orci, L., Immunocyto-
chemical localization of vitamin D-dependent calcium binding protein in chick duodenum,
J. Cell Bioi., 94, 201, 1982.
388 Nutrition and Gene Expression
30. Taylor, A. N., Gleason, W. A., and Lankford, G. L., Vitamin D dependent calcium
binding. Immunocytochemical localization of the rat intestinal protein, J. Histochem.
Cytochem., 32, 153, 1984.
31. Christakos, S., Brunette, M. G., and Norman, A. W., Localization of immunoreactive
vitamin D-dependent calcium binding protein in chick nephron, Endocrinology, 109, 322,
1981.
32. Roth, J., Thorens, B., Hunziker, W., Norman, A. W., and Orci, L., Vitamin D-
dependent calcium binding protein: immunocytochemical localization in chick kidney.
Science, 214, 197, 1981.
33. Rhoten, W. B. and Christakos, S., Immunocytochemical localization of vitamin D-
dependent calcium binding protein in mammalian nephron, Endocrinology, 109, 981,
1981.
34. Roth, J., Brown, D., Norman, A. W., and Orci, L., Localization of the vitamin D-
dependent calcium binding protein in mammalian kidney, Am. J. Physiol., 243, F243,
1982.
35. Desplan, C., Thomasset, M., and Moukhatar, M. S., Synthesis, molecular cloning
and restriction analysis of DNA complementary to vitamin D dependent calcium binding
protein mRNA from rat intestine, J. Bioi. Chern., 258, 2762, 1983.
36. Hunziker, W., Siebert, P., King, M., Strucki, P., Dugaiczk, A., and Norman, A. W.,
Molecular cloning of a vitamin D-dependent calcium binding protein mRNA sequence
from chick intestine, Proc. Nat/. Acad. Sci. U.S.A., 80, 4228, 1983.
37. Hunziker, W., The 28kDa vitamin D dependent calcium binding protein is a six domain
structure, Proc. Nat/. Acad. Sci. U.S.A., 83, 7578, 1986.
38. Wood, T. L., Kobayashi, Y., Frantz, G., Varghese, S., Christakos, S., and Tobin,
A. J., Calcium binding protein (calbindin-D 28k): expression of calbindin-D28k RNAs in
rodent kidney and brain, DNA, 7, 585, 1988.
39. Hunziker, W. and Schrickel, S., Rat brain calbindin-D28 k: six domain structure and
extensive amino acid homology with chicken calbindin-D 28. , Mol. Endocrinol., 2, 465,
1988.
40. Perret, C., Desplan, C., Brehier, A., and Thomasset, M., Characterization of rat 9-
kDa cholecalcin (CaBP) messenger RNA using a complementary DNA, Eur. J. Biochem.,
148, 61, 1983.
41. Theofan, G. and Norman, A., Effects of a amanitin and cycloheximide on 1,25-
dihydroxyvitamin D, dependent calbindin-D 28k and its mRNA in vitamin D3 replete chick
intestine, J. Bioi. Chern., 261, 7311, 1986.
42. Theofan, G., Nguyen, A. P., and Norman, A. W., Regulation of calbindin-D 28k gene
expression by I ,25-dihydroxyvitamin D3 is correlated to receptor occupancy, J. Bioi.
Chern., 261, 16943, 1986.
43. Dupret, J., Brun, P., Perret, C., Lomri, N., Thomasset, M., and Cuisinier-Gieizes,
P., Transcriptional and posttranscriptional regulation of vitamin D-dependent calcium
binding protein in rat duodenum by I ,25-dihydroxycholecalciferol, J. Bioi. Chern., 262,
16553, 1987.
44. Varghese, S., Lee, S., Huang, Y.-C., and Christakos, S., Analysis of rat vitamin D-
dependent calbindin-D28k gene expression, J. Bioi. Chern., 263, 9776, 1988.
45. Huang, Y.-C. and Christakos, S., Modulation of rat calbindin-D,.. gene expression by
1,25-dihydroxyvitamin D3 and dietary alteration, Mol. Endocrinol., 2, 928, 1988.
46. Varghese, S., Deaven, L. L., Huang, Y. C., Gill, R., Iacopino, A. M., and Chris-
takos, S., Transcriptional regulation and chromosomal assignment of the mammalian
calbindin-D28k gene, Mol. Endocrinol., 3, 495, 1989.
47. Huang, Y.-C., Lee, S., Stolz, R., Gabrielides, C., Pansini-Porta, A., Bruns, M. E.,
Bruns, D., Mifflin, T., Pike, J, W., and Christakos, S., Effect of hormones and
development on the expression on the rat I ,25-dihydroxyvitamin D, receptor gene and
comparison to calbindin gene expression, J. Bioi. Chem., 264, 17454, 1989.
Gill and Christakos 389
48. Rhoten, W. B. and Christakos, S., Cellular gene expression for calbindin-D 28 k in mouse
kidney, Anat. Rec., 227, 145, 1990.
49. Iacopino, A. M. and Christakos, S., Reduction of neuronal calcium binding protein
(calbindin-D2,.) gene expression in aging and neurodegenerative diseases, Proc. Nat/.
Acad Sci. U.S.A., 87, 4078, 1990.
50. Iacopino, A. M. and Christakos, S., Corticosterone regulates calbindin-D 28 • mRNA
and protein levels in rat hippocampus, J. Bioi. Chern., 265, 10177, 1990.
51. Iacopino, A. M. and Christakos, S., Calcium binding protein (calbindin-D2,.) gene
expression in the developing and aging mouse cerebellum, Mol. Brain Res., 8, 283,
1990.
52. Li, H. and Christakos, S., Differential regulation by 1,25 dihydroxyvitamin D3 of
calbindin-D9 • and calbindin-D2,. gene expression in mouse kidney, Endocrinology, 128,
2844, 1991.
53. Enomoto, H., Hendy, G. N., and Clemens, T. L., Regulation of calbindin-D,.. (CaBP)
mRNA in chick kidney cells: critical importance of posttranscriptional mechanisms and
calcium ion concentration, J. Bone Min. Res., 6 (Suppl. 1), abstract 390, Sl83, 1991.
54. Meyer, Galligan, M., Jones, G., Komm, B. S., and Haussler, M. R., 1 ,25(0H)zD 3
dependent regulation of calbindin-D 2,. mRNA apparently requires the synthesis of an
uncharacterized protein in chick duodenal organ culture, J Bone Min. Res., 6 (Suppl. 1),
abstract 406, Sl85, 1991.
55. Kimberg, D. V., Baerg, R. D., Gershone, E., and Graudusius, R. T., Effect of
cortisone treatment on the active transport of calcium by the small intestine, J. Clin.
Invest., 50, 1309, 1971.
56. Hall, A. K., Bishop, J. E., and Norman, A. W., Inhibitory and stimulatory effects of
dexamethasone and 1,25dihydroxyvitamin D3 on chick intestinal calhindin-D28• and its
mRNA, Mol. Cell. Endocrinol., 51, 25, 1987.
57. Corradino, R. A. and Fullmer, C. S., Positive cotranscriptional regulation of intestinal
calbindin-D 2,. gene expression by 1,25dihydroxyvitamin D3 and glucocorticoids, Endo-
crinology, 128, 944, 1991.
58. Navikis, R. J., Katzenellenbogen, B. S., and Nalbandov, A. V., Effects of the sex
steroids and vitamin D 3 on calcium binding protein in the chick shell gland, Bioi. Reprod.,
21, 1153, 1979.
59. Opperman, L. A., Sandes, T. j., Mills, S. E., Christakos, S., Bruns, D. E., and
Bruns, M. E., The control of calbindin-D9k and calbindin-D28• in mouse uterus by
1,25(0H),D3 and estrogen, J. Bone Min. Res., 5 (Suppl. 2), S264, 1990.
60. Hall, A. K., Wang, Y.-Z., Gill, R. K., and Christakos, S., Modulation of calbindin-
D2,.-like gene expression by retinoic acid in neuronal cells overexpressing a retinoic acid
receptor, J. Bone Min. Res., 6 (Suppl. 1), Sl84, 1991.
61. Kim, H. T. and Chen, T. L., 1,25Dihydroxyvitamin D3 interaction with dexamethasone
and retinoic acid: effects on procollagen messenger ribonucleic acid levels in rat osteoblast
like cells, Mol. Endocrinol., 3, 97, 1989.
62. Schute, R., Umesono, K., Mangelsdorf, D. J., Bolado, J,, Pike, J. W., and Evans,
R. M., Jun-fos and receptors for vitamin A and D recognize a common response element
in the human osteocalcin gene, Cell, 61, 497, 1990.
63. Umesono, K. and Evans, R. M., Determinants of target gene specificity for steroid/
thyroid hormone receptor, Cell, 57, 1139, 1990.
64. Mader, S., Kuman, U., de Verneuil, H., and Chambon, P., Three amino acids of
the esterogen receptor are essential to its ability to distinguish an estrogen from gluco-
corticoid response element, Nature, 338, 271, 1989.
65. Danielsen, M., Hinck, L., and Ringold, A.M., Two amino acids within the knuckle
of the first zinc finger specify DNA response element activation by glucocorticoid re-
ceptor, Cell, 57, 1131, 1989.
390 Nutrition and Gene Expression
66. Umesono, K., Murakami, K. K., Thompson, C., and Evans, R. M., Direct repeats
as selective response elements for the thyroid hormone, retinoic acid and vitamin D,
receptors, Cell, 65, 1255, 1991.
67. Stumpf, W. E. and O'Brien, L. P., 1,25(0H), Vitamin D, sites of action in the brain,
Histochemistry, 87, 393, 1987.
68. Stumpf, W. E. and Bidman, H.-J., Sites and mechanisms of action of vitamin Din
the nervous system, Abstracts, Eighth Workshop on Vitamin D, 23, 1991, 23.
69. Mattson, M. P., Rychlik, B., Chu, C., and Christakos, S., Evidence for calcium
reducing and excitoprotective roles for calcium binding protein, calbindin-D"' in cultured
hippocampal neurons, Neuron, 6, 41, 1991.
70. Christakos, S. and Norman, A. W., Evidence for a specific high affinity binding protein
for 1,25 dihydroxyvitamin D3 in chick kidney and pancreas, Biochem. Biophys. Res.
Commun., 89, 56, 1979.
71. Christakos, S. and Norman, A. W., Biochemical characterization of 1,25dihydroxyvitamin
D, receptors in chick pancreas and kidney cytosol, Endocrinology, 108, 140, 1981.
72. Clark, S. A., Stumpf, W. E., Sar, M., DeLuca, H. F., and Tanaka, Y., Target cells
for 1,25dihydroxyvitamin D3 in rat pancreas, Cell Tissue Res., 109, 515, 1980.
73. Parmentier, M., DeViglder, J. J. M., Muri, E., Szisirer, C., Islam, M. Q., Guerts
Van Kessel, A., Lawson, D. E. M., and Vassart, G., The human calbindin 27 kDa
gene: structural organization of the 5' and 3' region, chromosomal assignment and re-
striction fragment length polymorphism, Genomics, 4, 309, 1989.
74. Gill, R. K. and Christakos, S., Isolation of genomic clones harboring sequences ho-
mologous to mammalian calbindin-DZKk and their characterization, J. Bone Mineral Res.,
4, 1989, S291.
75. Minghetti, P. P., Cancela, L., Fujisawa, Y., Theofan, G., and Norman, A. W.,
Molecular structure of the chicken vitamin-D induced calbindin-D28k gene reveals eleven
exons. six Cal+ binding domains and numerous promoter regulatory elements, Mol.
Endocrinol .. 2, 355. I988.
76. Wilson, P. W., Rogers, J., Harding, M., Pohl, V., Pattyn, G., and Lawson, D. E. M.,
Structure of chick chromosomal gene for calbindin and calretinin, J. Mol. Biol.. 200,
615, 1988.
77. Ferrari, S., Drusian, E., Baltini, R., and Fregni, M., Nucleotide sequence of the
promoter region of the gene encoding chicken calbindin-DZKk• Nucleic Acid Res., 16,
353, 1988.
78. Perret, C., Lomri, N., Gouhier, N., Auffrey, C., and Thomasset, M., The rat vitamin-
D dependent calcium binding protein (9-KDa CaBP) gene: complete nucleotide sequence
and structural organization, Eur. J. Biochem., 172, 43, 1988.
79. Gill, R. and Christakos, S., Identification of sequence elements in the mouse calbindin-
D28k gene which confer basal activation and hormonal inducible response, Abstracts,
Eighth Workshop on Vitamin D, 1991, 81.
80. Markose, E. R., Stein, J. L., Stein, G. S., and Lian, J, B., Vitamin D-mediated
modifications in protein-DNA interaction at two promoter elements of the osteocalcin
gene, Proc. Nat/. Acad. Sci. U.S.A.. 87, 1701, 1990.
81. Demay, M. B., Gerardi, J. M., DeLuca, H. F., and Kronenberg, H. M., DNA
sequence in the rat osteocalcin gene that bind the I ,25 dihydroxyvitamin D3 receptor and
confer responsiveness to I ,25-dihydroxyvitamin D,, Proc. Nat!. Acad. Sci. U.S.A., 87,
369, 1990.
Chapter 18
TABLE OF CONTENTS
I. Introduction ...................................................... 392
I. INTRODUCTION
SKC, uv
LIVER
I PTH
t
SECRETION L -_ _ . BONE ----....J
t
HYPOCALCEMIA
I RESORPTION
\e
CALCITONIN
FIGURE 2. Morphology of normal rat diploid osteoblast cultures. (A through E) Light mi-
croscopy of cultured osteoblasts isolated from 21-d fetal calvaria by sequential collagenase
digestion 5° (A and B) Cells labeled with 'H thymidine show greater than 95% of the cells are
proliferating after plating (A, day 5). The cells multilayer, fom1ing nodules (B, day 12), and
the onset of expression of the osteoblast phenotype is indicated by alkaline phosphatase staining
of cells where proliferation has ceased in the center of the nodule (C. day 18 alkaline phosphatase
histochemistry). Every cell has ceased to proliferate and greater than 90% of the cells are alkaline
phosphatase positive (0, day 28). It is within the multilayered nodule that mineralization is
initiated; von Kossa silver staining shows mineralized nodules throughout the dish. A through
E, photographed at 100 x. (F) Scanning electron micrograph of a nodule on day 28 at 20 x,
with mineralized matri!( enveloping osteoblasts. (G and H): Transmission electron microscopy
(bars are 5 and 2.5 microns). (G) shows orthogonal organization of the collagen matrix in thick
layers between the osteoblasts in vitro. (H) Higher magnification of the collagen bundles shows
mineral deposition within the collagen fibrils and absence of intracellular calcification.
MATRIX
PROLIFERATION MATURATION MINERALIZATION
z cRl3
0 H4 AP CP ([; CA+2
iii 100
en
w
a:
a. 80
X
w
..J
<(
60
:!!!
x
<( 40
:!!!
0~ 20
0
0 7 14 21 28 35
DAYS IN CULTURE
(AP-1 SITES)
e
PHENOTYPE SUPPRESSION-,~--------,
ALK 0:: I
PHOS (£)
(£)
FIGURE 3. Temporal expression of cell growth and osteoblast phenotype related genes during
the development of in vitro formed bone-like tissue by normal diploid rat osteoblasts. Top panel:
isolated primary cells were initially cultured in MEM with 10% FCS, then after confluence, in
BGJb medium supplemented with 10% FCS, 50 fLglml ascorbic acid and 10 mM 13-glycerol
phosphate. Cellular RNA was isolated at the times indicated (3, 5, 7, 10, 12, 16, 21, 28, and
35 d) during the differentiation time course and assayed for the steady-state levels of various
transcripts by Northern blot analysis. The resulting blots were quantitated by scanning densi-
tometry and the results plotted relative to the maximal expression of each transcript. Three
periods of gene expression are represented: (A) Genes characterizing the proliferation period
include H4 histone (which reflects DNA synthesis), c-myc and c-fos, plus genes expressed for
formation of the type I collagen, fibronectin (FN), and transforming growth factor-13 (TGFI3);
(B) alkaline phosphatase (AP) is expressed postproliferatively and associated with extracellular
matrix maturation; (C) genes induced to high levels with onset of extracellular matrix mineral-
ization; represented are osteopontin (OP) and osteocalcin (OC). Calcium (Ca2 +) accumulation
is indicated. This temporal pattern defines three periods, proliferation, matrix maturation, and
mineralization reflecting a developmental sequence of osteoblast maturation. Lower panel: the
interrelationship and feedback signals between the periods are indicated. AP-I activity in pro-
liferating osteoblast, that is, fos-jun protein binding to AP-I sites, suppresses activity of post-
proliferative expressed genes.
398 Nutrition and Gene Expression
with expression of cell cycle (e.g., histone) and cell growth (e.g., c-myc, c-
fos and AP-1 activity) regulated genes. It is fundamental to development of
the bone cell phenotype that several genes associated with formation of the
extracellular matrix (type I collagen, fibronectin, and TGF~) are actively
expressed in this proliferation period. Immediately following the downreg-
ulation of proliferation, as reflected by the decline in DNA synthesis eH-
thymidine incorporation) and histone gene expression, the expression of al-
kaline phosphatase (enzyme activity and mRNA) increases greater than ten-
fold. At this time, a differentiation-specific histone gene is expressed. 54 •55
During this period (from 12 to 18 d) the extracellular matrix undergoes a
series of modifications in composition and organization that renders it com-
petent for mineralization. During this matrix maturation phase, every cell has
become alkaline phosphatase positive as the cultures progress into the mi-
neralization stage. In heavily mineralized cultures, cellular levels of alkaline
phosphatase mRNA decline.
With the onset of extracellular matrix mineralization, other bone-specific
genes are induced. For example, osteopontin or SPPI, a 60-kDa calcium
binding protein enriched in 0-phosphoserine, 56 and osteocalcin, a -y-carbox-
yglutamate-containing protein, 57•56 are upregulated in concert with the accu-
mulation of mineral. Notably, osteopontin is expressed during the period of
active proliferation (at 25% of maximal levels), consistent with its high level
of expression following oncogene transformation or phorbal ester treatments
of cells. 59 Osteopontin mRNA is decreased postproliferatively and then ex-
hibits an induction at the onset of mineralization achieving peak levels of
expression during the mineralization period (days 16 to 20). In contrast to
osteopontin, the vitamin K-dependent protein, 60 osteocalcin, is expressed only
postproliferatively with the onset of nodule formation. 51 This 5. 7-kDa calcium
binding protein is characterized by 3 -y-carboxyglutamic acid residues (Gla),
binds tightly to hydroxyapatite, and is maximally expressed during active
mineralization of the extracellular matrix (ECM) in vivo51 and in vitro. 51 A
related vitamin K-dependent protein in bone, matrix Gla protein (MGP), 61 is
characterized by the presence of 5 Gla residues in a 10-kDa molecule. MGP
is present in proliferating cells and is increased in relation to accumulation
of collagen, but levels of expression are not correlated with the mineralization
phase. 62 Several manipulations of the culture system51 demonstrate that in-
duction of high levels of osteocalcin and osteopontin mRNA are dependent
upon formation of a mineralized extracellular matrix whereas MGP levels are
sustained independent of mineralization. 62 Both osteocalcin and osteopontin
are expressed in mature osteoblasts within the nodules 63 and the proteins are
localized to the mineralized bone extracellular matrix. 64 Osteocalcin has been
shown to contribute to regulation of the mineral phase in bone both in vitro
as a potential inhibitor of mineral nucleation 65 and in vivo as a bone matrix
signal that promotes osteoclast differentiation and activation. 16 •66 •67 Osteo-
pontin has been localized to the region of osteoclast contact. 66 More recently,
Lian and Stein 399
MATRIX
MINERALIZATION
PROLIFERATION MATURATION
120 400
100 H4 AP OP
300
n.
:::!!:
:::> 75 80
:::!!: 200
x
c(
50
II I•
40 100
:::!!:
:.!!
0
25
0 0 I
0
7 14 21 28 35 14 21 7 14 21 28 35
7 28 35
400
100 CXll
900 ~
300
oc
:::!!:
:::> 75
:::!!: 600
200
xc( 50
:::!!: 300 100
25
:.!!
0
0 0
I• I• II I• 0
7 14 21 28 35 7 14 21 28 35
7 14 21 28 35
DAYS DAYS DAYS
FIGURE 4. Examples of vitamin D-regulated genes during the three periods of osteoblast
phenotype development. Solid bars are controls and open bars are cells treated 48 h prior to
harvest with oo-•
M) I ,25(0H) 2D 3 . Samples during the time course were hybridized together
for quantitation of the control and hormone-treated samples as percent maximal expression for
each gene. Note the pleiotropic effects on proliferation related genes. Histone (H4) and type I
collagen (COLL) are inhibited early (day 8 through 16). then stimulated in mature osteoblasts
in a mineralized matrix (day 28 through 35); alkaline phosphatase (AP) is also stimulated.
MATRIX
MATURATION
OA YS AFTER PLA T1 NG
observed related to extent of mineralized matrix that was formed at the time
of vitamin D exposure (Figure 5, lower panel). Continuous presence of hor-
mone blocked further maturation of the cultures (e.g., growth and mineral-
ization of nodules were inhibited). It has been observed that chronic exposure
of rat osteosarcoma cells (Ros 17/2.8) to I ,25(0H) 2 D 3 can result in altered
phenotypic expression. After 5 d, osteocalcin mRNA was decreased and MGP
expression, which is not normally detected in this Ros line, was induced. 61
One factor contributing to the variability of response of the osteoblast to
I ,25(0H) 2 D3 is potentially related to changes in the receptor levels of vitamin
D. Receptor level is regulated by proliferative status of the cell, presence of
glucocorticoids, and homologous upregulation of its receptor, as shown in
the Ros 17/2.8 rat osteosarcoma osteoblast-like cellline. 29 •95 However, it can
be seen in Figure 4 that genes expressed both during proliferation and dif-
ferentiation (e.g., OP and MGP) are regulated by the hormone throughout
the developmental sequences. Thus, receptor availability is not rate limiting.
Calcium is yet another contributing factor to the regulation of the vitamin D
receptor in osteoblast-like cells. This was suggested by the report96 that the
upregulation of the vitamin D receptors in UMR 106 cells is inhibited by two
different calcium channel blockers, nitrendipine and verapamil. Such obser-
vations raise the possibility that osteoblasts within a matrix that is undergoing
extracellular mineralization, may show variable levels of vitamin D respon-
siveness if the receptor level is closely regulated by the extracellular calcium
concentration.
In addition to the direct modulation of osteoblast gene activity that results
from interaction of the vitamin D receptor complex with specific gene se-
quences, new evidence has accumulated to indicate several nongenomic ef-
fects. Changes in cellular calcium occur in response to 1,25(0H) 2 D 3 in a
matter of seconds and these have been classified as "rapid" or nongenomic
effects. 1-a-25(0HhD3 rapidly increases cytosolic calcium and alters mem-
brane phospholipid metabolism in hepatocytes, 97 cultured osteoblasts, 3 .4·98
chondrocytes 7 and isolated enterocytes. 4 The rapid transmembrane influx of
extracellular calcium after stimulation by 1,25(0H) 2 D 3 is mediated largely
by the opening of voltage-gated calcium channels. 3 •99 The rapid, "nongen-
omic'' effects of vitamin D include prolonged opening of the voltage-sensitive
calcium channels present in the plasma membrane, 99 phosphoinositide break-
down,4 altered enzymatic activity in matrix vesicles, 7 and calcium transport
in chick intestine. 100 A fundamental question raised by these observations is
whether the responses are mediated by a different type of receptor for
1,25(0H) 2 D3 4·101 and whether there are interactions with the steroid-receptor
signaling pathway.
Thus, many aspects of osteoblast metbolism are regulated by vitamin D.
Several factors, however, cloud an understanding of the precise effects of
vitamin D on the osteoblast in relation to bone formation and resorption.
First, other than an understanding of collagen as a bone matrix structural
404 Nutrition and Gene Expression
TABLE 1
Sequence Comparison of Known Vitamin D-Responsive Elements in
the Promoters of Vitamin D-Regulated Genes
API
-512 -483
Human osteocalcin 5'GGTGACTCA cc GGGTGA ACG GGGGCA TT 3'
API
-470 -443
Rat osteocalcin 5'TGCCCTGCA CT GGGTGA ATG AGGACA TT 3'
-761 -741
Mouse osteopontin 5' AC AA GGTTCA CGA GGTTCA CG 3'
- 198 -180
Mouse calbindin-D28k 5' GGGGGA TGTG AGGAGA 3'
-489 -475
Rat calbindin-D9, 5' GGGTCT CGG AAGCCC 3'
.. ·r-----1·.
AP-1
.
GCACTGGGTGAA TGAGGACA TT ACTGACCGCT
AP- 1
r------1
AP-1
r--1
ATGACCCCCM TT AGTCCTGGCAG
GRE GRE
..----, r;---1
GilTIITMMGGT IITTGCAGAACAGACMGTCCC
c:QtS~\C"~
-1100 -464 -437 -99 -76 -33 -1 50
mRNA
'
FIGURE 6. Rat osteocalcin gene promoter indicating sequences for physiological regulatory
elements. Structural organization of the rat osteocalcin gene. (A) Sequences of those regulatory
elements in the proximal promoter that have been defined and partially characterized. These
include the vitamin D responsive element (VDRE); the osteocalcin box (OC box), which is a
primary proximal transcription regulatory element containing the CCAAT motif as a central core;
and the glucocorticoid response elements (GLUC) in the rat gene 173 or overlapping the TATA
motif as defined in the human gene. 121 Within the OC box and VDRE elements are also found
active AP-1 sites which bind the oncogene-encoded Fos/Jun protein complex. The solid circles
above or below G residues indicate vitamin D receptor protein-DNA interactions defined at single
nucleotide resolution within the vitamin D responsive element. 94 (B) With reference to the OC
box and TATA promoter elements, intron and exon organization and location of the pre-, pro-
peptide, and mature osteocalcin forms are illustrated.
.D.D
~~ 0
<<
II
(.)
II
a: a.. + a: a..
PROBE:VDRE PROBE:VDRE
ROS 17/2.8 ROB
FIGURE 7. Characterization of vitamin D-mediated enhancement of protein-DNA interactions
at the rat OC gene VDRE in normal osteoblasts and Ros 17/2.8 cells. Gel mobility shift assays
using nuclear proteins from untreated (C) or w-• M vitamin D-treated (D) confluent (day 8)
Ros 17/2.8 cells or mature (day 32) normal diploid osteoblasts show enhancement of protein-
DNA interactions with a "P-labeled rat osteocalcin VDRE oligonucleotide DNA probe following
treatment of the cells with vitamin D receptor (VDR) binding is indicated by arrows. Monoclonal
antibodies against the porcine VDR which detect only the porcine VDR (P-Ab) or which cross-
react with the rat VDR (R-Ab) were preincubated with the nuclear proteins for 20 min prior to
DNA probe addition. 124 These antibodies demonstrate that the vitamin D enhanced protein-DNA
interactions observed in both cell types are dependent on the binding of the vitamin D receptor.
Note that two VDR-DNA complexes are formed in the osteosarcoma cells and only the fast-
mobility complex is detectable in differentiated ROB cells expressing osteocalcin.
412 Nutrition and Gene Expression
T G C C C T G C A C T G G G T G A A T GA G G A C A T T A C T G A C C
A C G G G AC G T G A C C C A C T T A C T C C T G T A A T G AC T G G
HUMAN
G G T G A C T C A C C G G G T G A AC G G G G G C A T T G C G A G G C
C C A C T G A G T G GC C C A C T T G C C C C C G T A A C G C T C C G
0
z :z~
.0
<(
.0
<( ....,....,o
:::> :::> _J
,, 0
"-J
I
+ a..
I
a: ++
Cl) z Cl) Cl) I
0:::>0 Oa...
u......, u.. u.. <(
Recombinant
Fos/Jun
Extracts from Proteins
Vitamin D
Treated Cells
FIGURE 8- Vitamin D receptor and Fos-Jun protein complex binding to the human VDRE
sequences. Nuclear extracts from 48-h vitamin D-treated Ros cells were used in gel mobility
shift assays (left panel) with an oligonucleotide probe to the human VDRE as indicated by the
dotted underline in the top panel sequence. The vitamin D receptor complex is blocked by
preincubation with antibody recognizing the rat receptor (R-AB) but not the pig-specific antibody
(9P-AB), provided by H. DeLuca, Madison, WI. The probe used for these protein-DNA inter-
actions has the classic AP-I consensus deleted (solid underline) but retains the AP-I site ho-
mologous to the rat VDRE (bold letters) that resides within the core of this regulatory element.
The right panel demonstrates that this is an active AP-I site by binding of only the Fos-Jun
protein complex which can be competed by the AP-1 oligonucleotide.
414 Nutrition and Gene Expression
I. Release of the Fos-Jun complex from the AP-I sites to permit sequences
to be available for occupancy by the vitamin D receptor complex and/
or by tissue-specific osteocalcin box transcription factors;
2. Modifications of the Fos-Jun complex that facilitate binding of acti-
vation related factors;
3. Association with other DNA binding proteins or protein-protein inter-
actions.
With respect to the latter possibility, binding of the Fos-Jun complex may
play a dual positive and negative role in the regulation of transcription. The
Fos-Jun complex may suppress osteocalcin gene transcription when prolif-
eration is ongoing by directly or indirectly modulating sequence-specific in-
teractions at the vitamin D receptor binding domain. Then postproliferatively,
the Fos-Jun complex may facilitate vitamin D receptor binding to support the
sequential upregulation of osteocalcin and other vitamin D responsive genes.
Such regulation could be accomplished by different fos-jun protein complexes.
Both fos and jun are members of a family of related proteins, and a recent
study indictes differential stimulation of c-jun and jun-B by I ,25(0H) 2 D 3 in
MC3T3-EI osteoblastic cells. 133
Further experimental results are necessary to determine whether the or-
ganization of steroid receptor binding domains and AP-I sites within pro-
moters of genes that are hormone responsive can provide a general mechanism
Lian and Stein 415
TABLE 2
Examples of Human Diseases Related to
Disturbances in the Vitamin D Endocrine System
Anticonvulsant therapy
Cirrhosis
Diabetes
Drug-induced metabolism
Glucocorticoid antagonism
Hyperparathyroidism
Hypoparathyroidism
Leukemia
Malabsorption
Osteomalacia
Osteoporosis
Psoriasis
Renal disease
Sarcoidosis
Steatorrhea
doses of phosphate and 1,25(0H) 2D3 have allowed for fairly normal growth
of these children. 139 In addition to these inborn errors of vitamin D metabolism
are those disorders involving a tissue-selective resistance to I ,25(0H) 2 D3-
but here there is, as yet, no understanding of the cellular basis which relates
to a direct effect of vitamin D actions either at the genomic or nongenomic
levels. For example, elevated 1,25(0H) 2D3 levels occur in human and rat
osteopetrosis, a hereditary disorder having a spectrum of osteoclast abnor-
malities.140 The hormone level may be elevated to maintain normal serum
calcium in the absence of active bone resorption. Alterations in some vitamin
D-regulated proteins (osteocalcin, osteopontin, collagen, alkaline phospha-
tase) indicate perturbations in bone responsiveness. 141
Vitamin D (1 ,25(0H) 2D 3) is being considered for the treatment of osteo-
porosis, 142 a growing problem. 143 Type I postmenopausal osteoporosis occurs
predominantly in women within 20 years after the menopause and is related
to estrogen deficiency. It is characterized by accelerated bone loss which leads
to suppressed PTH secretion and a trend toward low serum I ,25(0H) 2 D
concentrations and inefficient calcium absorption. 144. 146 In clinical trials in
which calcium intake was limited and dosages of 1,25(0H)zD 3 of 0.5 J.Lg/d
were employed, hypercalcemia and hypercalcuria occurred uncommonly. 147
Short-term rather than continuous treatment preferentially stimulates osteo-
blast function but not osteoclast activity. 148 Type II is the age related osteo-
porosis resulting in defects in the vitamin D endocrine system that includes
primary failure by the kidney and defective 25(0H)D1a-hydroxylase activ-
ity, 149 a decrease in vitamin D receptor concentration in intestinal mucosa and
possibly in bone cells, and, in some elderly patients, vitamin D deficiency.
With age there is decreased intestinal absorption of calcium, and this is a
major determinant in the etiology of the disease. 144
Although some investigators have documented reduced levels of
1,25(0H)zD3 in osteoporotic subjects when compared to age matched controls,
several studies document vitamin D levels in the normal range. 150 Thus, the
value of calcitriol treatment for established postmenopausal osteoporosis re-
mains controversial. Findings of several major clinical trials that have used
calcitriol in the treatment of osteoporosis are conflicting. There are studies,
e.g., Aloia et al. 151 and Caniggia et al., 152 that indicate calcitriol treatment
reduces bone loss in women with postmenopausal osteoporosis by increasing
calcium absorption and reducing bone resorption. Gallagher et al. 153 has re-
cently demonstrated that the treatment of postmenopausal osteoporotic women
with calcitriol for 2 years was associated with an increase in spine density
and total body calcium when compared to placebo. In the recent Tilyard
studyl 76 (n = 600), a 50% reduction of spinal deformation compared to
calcium-treated controls was observed. However, the Falch et al. 154 and the
Ott and Chestnut 155 studies indicate that calcitriol is not effective in the
treatment of established postmenopausal osteoporosis. The different obser-
vations may relate to the patient population and/or perhaps the stage of the
disease.
418 Nutrition and Gene Expression
TABLE 3
Stimulation of Osteocalcin (OC) and
Matrix Gla Protein (MGP) mRNA by
Vitamin D Analogs in Osteoblasts
Ratio of Vitamin
D/controt•
oc MGP
VI. SUMMARY
REFERENCES
I. Naveh-Many, T., Marx, R., Keshet, E., Pike, J. W., and Silver, J., Regulation of
I ,25-dihydroxyvitamin 03 receptor gene expression by I ,25-dihydroxyvitamin 03 in the
parathyroid in vivo, J. Clin. Invest., 86, 1968, 1990.
2. Ozono, K., Sone, T., and Pike, J. W., The genomic mechanism of action of 1,25-
dihydroxyvitamin 0 3 , J. Bone Min. Res., 6, 1021, 1991.
3. Civitelli, R., Kim, Y. S., Gunsten, S. L., Fujimori, A., Huskey, M., Avioli, L. V.,
and Hruska, K. A., Nongenomic activation of the calcium message system by vitamin
0 metabolites in osteoblast-like cells, Endocrinology, 127, 2253, 1990.
4. Lieberherr, M., Grosse, B., Duchambon, P., and Drueke, T., A functional cell surface
type receptor is required for the early action of 1,25-dihydroxyvitamin 0 3 on the phos-
phoinositide metabolism in rat enterocytes, J. Bioi. Chern., 264, 20403, 1989.
5. Baran, D. T., Sorensen, A. M., Owen, T., Shalhoub, V., Stein, G., and Lian, J.,
lo.,25-0ihydroxyvitamin 0 3 rapidly increases cytosolic calcium in clonal rat osteosarcoma
cells lacking the vitamin 0 receptor, J. Bone Min. Res., 6, 1269, 1991.
6. Harmond, M. F., Thomasset, M., Rouais, F., and Ducassou, D., In vitro stimulation
of articular chondrocyte differentiated function by 1,25-dihydroxycholecalciferal or 24,R,25-
dihydroxycholecalciferol, J. Cell. Physiol., 119, 359, 1984.
7. Boyan, B. D., Schwartz, 1., Carnes, D. L., and Ramirez, V., The effects of vitamin
0 metabolites on the plasma and matrix vesicle membranes of growth and resting cartilage
cells in vitro, Endocrinology, 122, 2851, 1988.
8. Gerstenfeld, L. C., Kelly, C. M., von Deck, M., and Lian, J. B., Effect of 1,25-
dihydroxyvitamin D, on induction of chondrocyte maturation in culture: extracellular
matrix gene expression and morphology, Endocrinology, 126, 1599, 1990.
9. Owen, T. A., Aronow, M.A., Barone, L. M., Bettencourt, B., Stein, G., and Lian,
J. B., Pleiotropic effects of vitamin Don osteoblast gene expression are related to the
proliferative and differentiated state of the bone cell phenotype: dependency upon basal
levels of gene expression, duration of exposure and bone matrix competency in normal
rat osteoblast cultures, Endocrinology, 128, 1496, 1991.
10. Soda, T., Shinki, T., and Takahashi, N., The role of vitamin Din bone and intestinal
cell differentiation, Annu. Rev. Nutr., 10, 195, 1990.
11. DeLuca, H. F., The vitamin D story: a collaborative effort of basic science and clinical
medicine, FASEB J., 2, 224, 1988.
12. Minghetti, P. P. and Norman, A. W., 1,25(0H) 2-vitamin D3 receptors: gene regulation
and genetic circuitry, FASEB J., 2, 3043, 1988.
13. Stern, P. H., Vitamin 0 and bone, Kidney Int., 29, Sl7, 1990.
14. Marx, S. J. and Barsony, J., Tissue-selective 1,25-dihydroxyvitamin D3 resistance:
novel applications of calciferols, J. Bone Min. Res., 3, 481, 1988.
15. Toverud, S. U., Hammarstrom, L. E., and Kristoffersen, U. M., Quantitative studies
on acid phosphatase in developing rat bones and teeth during hypervitaminosis D, Arch.
Oral Bioi., 20, 175, 1975.
16. Lian, J. B., Carnes, D. L., and Glimcher, M. j., Bone and serum concentrations of
osteocalcin as a function of 1,25 (OH) 2 D3 circulating levels in rats with bone disorders,
Endocrinology, 120, 2123, 1987.
17. Marie, P. J., Hott, M., and Garba, M.-T., Contrasting effects of 1,25-dihydroxyvi-
tamin D3 on boRe matrix and mineral appositional rates in the mouse, Metabolism, 34,
777, 1985.
18. Weisbrode, S. E., Capen, C. C., and Norman, A. W., Ultrastructural evaluation of
the effect of 1,25-dihydroxyvitamin D3 on bone of thyroparathyroidectomized rats fed a
low-calcium diet, Am. J. Pathol., 92, 459, 1978.
19. Hock, J. M., Gunnes-Hey, M., Poser, J., Olson, H., Bell, N.H., and Raisz, L. G.,
Stimulation of underminera1ized matrix formation by 1,25 dihydroxyvitamin D, in long
bones of rats, Calcif. Tissue Int., 28, 79, 1986.
Lian and Stein 421
20. Gallagher, J. A., Beneton, M., Harvey, L., and Lawson, D. E. M., Response of
rachitic rat bones to I ,25-dihydroxyvitamin D,: biphasic effects on mineralization and
lack of effect on bone resorption, Endocrinology, 119, 1603, 1986.
21. Marie, P. J, and Travers, R., Continuous infusion of I ,25-dihydroxyvitamin D3 stim-
ulates bone turnover in the normal young mouse, Calcif. Tissue Int., 35, 418, 1983.
22. Underwood, J, L. and DeLuca, H. F., Vitamin D is not directly necessary for bone
growth and mineralization, Am. J. Physiol., 246, E493, 1984.
23. Holtrop, M. E., Cox, K. A., Carnes, D. L., and Holick, M. F., Effects of serum
calcium and phosphorus on skeletal mineralization in vitamin D-deficient rats, Am. J.
Physiol., 251, E234, 1986.
24. Clark, S. A., Boass, A., and Toverud, S. U., Effects of high dietary contents of calcium
and phosphorus on mineral metabolism and growth of vitamin D-deficient suckling and
weaned rats, Bone Min., 2, 257, 1987.
25. Kollenkirchen, U., Walters, M. R., and Fox, J,, Plasma Ca influences vitamin D
metabolite levels as rats develop vitamin D deficiency, Am. J. Physiol., 260, E447, 1991.
26. Wronski, T. J,, Halloran, B. P., Bikle, D. D., Globus, R. K., and Morey-Holton,
E. R., Chronic administration of I ,25-dihydroxyvitamin D,: increased bone but imparied
mineralization, Endocrinology, 119, 2580, 1986.
27. Raisz, L. G., Trummel, C. L., Holick, M. F., and DeLuca, H. F., 1,25-dihydrox-
ycholecalciferol, a potent stimulator of bone resorption in tissue culture, Science, 175,
768, 1972.
28. Tanaka, Y. and DeLuca, H. F., Bone mineral mobilization activity of I ,25-dihydroxy-
cholecalciferol, a metabolite of vitamin D., Arch. Biochem. Biophys., 146, 574, 1971.
29. Chen, T. L., Cohn, C. M., Morey-Holton, E., and Feldman, D., lo:,25-Dihydroxy-
vitamin D 3 receptors in cultured rat osteoblast-like cells, J. Bioi. Chem., 258, 4350,
1983.
30. Chen, T. L., Li, J, M., van Ye, T., Cone, C. M., and Feldman, D., Hormonal
responses to 1,25-dihydroxyvitamin D3 in cultured mouse osteoblast-like cells-modulation
by changes in receptor level, J. Cell. Physiol., 126,21, 1986.
31. Rodan, G. A. and Martin, T. J,, Role of osteoblasts in hormonal control of bone
resorption-a hypothesis, Ca/cif. Tissue Int., 33, 349, 1981.
32. McSheehy, P. M. J. and Chambers, T. J., 1,25-Dihydroxyvitamin D 3 stimulates rat
osteoblastic cells to release a soluble factor that increases osteoclastic bone resorption,
J. Clin. Invest., 80,425. 1987.
33. Abe, E., Miyaura, C., Tanaka, H., Shina, Y., Kuribayashi, T., Suda, S., Nishii,
Y., DeLuca, H. F., and Suda, T., lo:,25-Dihydroxyvitamin D, promotes fusion of
mouse alveolar macrophages both by a direct mechanism and by a spleen cell-mediated
indirect mechanism, Proc. Nat!. Acad. Sci. U.S.A., 80, 5583, 1983.
34. Roodman, G. D., Ibbotson, K. J., MacDonald, B. R., Kuehl, T. J., and Mundy,
G. R., I ,25-Dihydroxyvitamin D, causes formation of multinucleated cells with several
osteoclast characteristics in cultures of primate marrow, Proc. Nat!. Acad. Sci. U.S.A.,
82, 8213, 1985.
35. Narbaitz, R., Stumpf, W. E., Sar, M., Huang, S., and DeLuca, H. F., Autoradio-
graphic localization of target cells for lo:,25-dihydroxyvitamin D, in bones from fetal
rats, Calcif. Tissue Int., 35, 177, 1983.
36. Owen, M. and Friedenstein, A. J., Stromal stem cells: marrow-derived osteogenic
precursors, in Cell and Molecular Biology of Vertebrate Hard Tissues, Evered, D. and
Harnett, S., Eds., John Wiley & Sons, Chichester, 1988, 42.
37. Benayhahu, D., Kletter, Y., Zipori, D., and Weintraub, S., Bone marrow-derived
stromal cell line expressing osteoblastic phenotype in vitro and osteogenic capacity in
vivo, J. Cell. Physiol., 140, I, 1989.
422 Nutrition and Gene Expression
38. Bernier, S. M., Desjardina, J., Sullivan, A. K., and Gollzman, D., Establishment
of an osseous cell line from fetal rat calvaria using an immunocytolytic method of cell
selection: characterization of the cell line and of derived clones, J. Cell. Physiol., 145,
274, 1990.
39. Grigoriadis, A. E., Heersche, J, N. M., and Aubin, J, E., Differentiation of muscle,
fat, cartilage, and bone from progenitor cells present in a bone-derived clonal cell pop-
ulation: effect of dexamethasone, J. Cell Bioi., 106, 2139, 1988.
40. Chen, T. L., Cone, C. M., Morey-Hollon, E., and Feldman, D., Glucocorticoid
regulation of I ,25(0H) 2D 3-vitamin D, receptors in cultured mouse bone cells, J. Bioi.
Chern., 257, 13564, 1982.
41. Peck, W. A., Birge, S. J., and Fedak, S. A., Bone cells: biochemical and biological
studies after enzymatic isolation, Science, 146, 1476, 1964.
42. Wong, G. L., Luben, R. A., and Cohn, D. V., 1,25-Dihydroxycholecalciferol and
parathormone: effects on osteoclast-like and osteoblast-like cells, Science, 197, 663, 1977.
43. Beresford, J, N., Gallagher, J, A., and Russell, R. G. G., 1,25-Dihydroxyvitamin
D, and human bone-derived cells in vitro: effects on alkaline phosphatase, Type I collagen
and proliferation, Endocrinology, 119, 1176, 1986.
44. Robey, P. G. and Termine, L. J., Human bone cells in ~·itro, Calc. Tissue Int., 37,
453, 1985.
45. Bellows, C. G., Aubin, J. E., Heersche, H. N. M., and Antosz, M. E., Mineralized
bone nodules formed in vitro from enzymatically released rat calvaria cell populations,
Calcif. Tissue Int., 38, 143, 1986.
46. Bhargava, U., Bar-Lev, M., Bellows, C. G., and Aubin, J, E., Ultrastructural analysis
of bone nodules formed in vitro by isolated fetal rat calvaria cells, Bone, 9, 155, 1988.
47. Ecarot-Charrier, B., Glorieux, F. H., van der Rest, M., and Pereira, G., Osteoblasts
isolated from mouse calvaria initiate matrix mineralization in culture, J. Cell Bioi., 96,
639, 1983.
48. Gerstenfeld, L. C., Chipman, S. D., Glowacki, J,, and Lian, J, B., Expression of
differentiated function by mineralizing cultures of chicken osteoblasts, Dev. Bioi., 122,
49, 1987.
49. Ibaraki, K., Termine, J, D., Whitson, S. W., and Young, M. F., Bone matrix mRNA
expression in differentiating fetal bovine osteoblasts, J. Bone Miner. Res., 7, 743, 1992.
50. Aronow, M. A., Gerstenfeld, L. C., Owen, T. A., Tassinari, M. S., Stein, G. S.,
and Lian, J, B., Factors that promote progressive development of the osteoblast phe-
notype in cultured fetal rat calvaria cells, J. Cell Physiol., 143, 213, 1990.
51. Owen, T. A., Aronow, M., Shalhoub, V., Barone, L. M., Wilming, L., Tassinari,
M. S., Kennedy, M. B., Pockwinse, S., Lian, J. B., and Stein, G. S., Progressive
development of the rat osteoblast phenotype in vitro: reciprocal relationships in expression
of genes associated with osteoblast proliferation and differentiation during formation of
the bone extracellular matrix, J. Cell. Physiol., 143, 420, 1990.
52. Gerstenfeld, L. C., Chipman, S.D., Kelly, C. M., Hodgens, K. J,, Lee, D. D., and
Landis, W. J., Collagen expression, ultrastructural assembly, and mineralization in
cultures of chicken embryo osteoblasts, J. Cell Bioi., 106, 979, 1988.
53. Stein, G. S., Lian, J. B., and Owen, T. A., Relationship of cell growth to the regulation
of tissue-specific gene expression during osteoblast differentiation, FASEB J., 4, 3111,
1990.
54. Collart, D., Ramsey-Ewing, A., Bortell, R., Lian, J., Stein, J., and Stein, G.,
Isolation and characterization of a eDNA from a human histone H2B gene which is
reciprocally expressed in relation to replication-dependent H2B histone genes during HL60
cell differentiation, Biochemistry, 30, 1610, 1991.
Lian and Stein 423
55. Shalhoub, V., Gerstenfeld, L. C., Lian, J. B., Stein, J. L., and Stein, G. S., Down-
regulation of cell growth and cell cycle regulated genes during chick osteoblast differ-
entiation with the reciprocal expression of histone gene variants, Biochemistry, 28, 5318,
1989.
56. Oldberg, A., Franzen, A., and Heinegard, D., Cloning and sequence analysis of rat
bone sialoprotein (osteopontin) eDNA reveals an Arg-Gly-Asp cell-binding sequence,
Proc. Nat!. Acad. Sci. U.S.A., 83, 8819, 1986.
57. Hauschka, P. V., Lian, J. B., Cole, D. E. C., and Gundberg, C. M., Osteocalcin
and matrix Gla protein: vitamin K-dependent proteins in bone, Physiol. Rev., 69, 990,
1989.
58. Price, P. A., Role of vitamin K-dependent proteins in bone metabolism, Annu. Rev.
Nutr., 8, 565, 1988.
59. Craig, A. M., Smith, J. H., and Denhardt, D. T., Osteopontin, a transformation-
associated cell adhesion phosphoprotein, is induced by 12-0-tetradecanoylphorbol 13-
acetate in mouse epidermis, J. Bioi. Chern., 264, 9682, 1989.
60. Lian, J. B. and Friedman, P. A., The vitamin K-dependent synthesis of gamma-
carboxyglutamic acid by bone microsomes, J. Bioi. Chern., 253, 6623, 1978.
61. Fraser, J.D. and Price, P. A., Induction of matrix Glaprotein synthesis during prolonged
I ,25-dihydroxyvitamin D, treatment of osteosarcoma cells, Calcif. Tissue Int., 46, 270,
1990.
62. Barone, L. M., Owen, T. A., Tassinari, M. S., Bortell, R., Stein, G. S., and Lian,
J. B., Developmental expression and hormonal regulation of the rat matrix Gla protein
(MGP) gene in chondrogenesis and osteogenesis, J. Cell. Biochem., 46, 000-000, 1991.
63. Pockwinse, S., Wilming, L., Conlon, D., Stein, G. S., and Lian, J. B., Expression
of cell growth and bone specific genes at single cell resolution during development of
bone tissue-like organization in primary osteoblast cultures, submitted.
64. Nagata, T., Bellows, C. G., Kasugai, S., Butler, W. T., and Sodek, J., Biosynthesis
of bone proteins [SPP-1 (secreted phosphoprotein-!, osteopontin), BSP (bone sialoprotein)
and SPARC (osteonectin)] in association with mineralized-tissue formation by fetal rat
calvarial cells in culture, Biochem. J., 274, 513, 1991.
65. Boskey, A. L., Wians, F. H., Jr., and Hauschka, P. V., The effect of osteocalcin on
in vitro lipid-induced hydroxyapatite formation and seeded hydroxyapatite growth, Calc.
Tissue Int., 37, 57, 1985.
66. Lian, J. B., Tassinari, M., and Glowacki, J., Resorption of implanted bone prepared
from normal and warfarin-treated rats, J. Clin. Invest., 73, 1223, 1984.
67. Glowacki, J. and Lian, J., Impaired recruitment and differentiation of osteoclast pro-
genitors by osteocalcin-depleted bone implants, Cell Differ., 21, 247, 1987.
68. Reinbolt, F. P., Hultenby, K., Oldberg, A., and Heinegard, D., Osteopontin-a
possible anchor of osteoclasts to bone, Proc. Nat!. Acad. Sci. U.S.A., 87, 4473, 1990.
69. Miyauchi, A., Alvarez, J., Greenfield, E. M., Teti, A., Grano, M., Colucci, S.,
Zambonin-Zallone, A .. , Ross, F. P., Teitelbaum, S. L., Cheresh, D., and Hruska,
K. A., Recognition of osteopontin and related peptides by an etvf3 3 integrin stimulates
immediate cell signals in osteoclasts, J. Bioi. Chern., 266, 20369, 1991.
70. Weinreb, M., Shinar, D., and Rodan, G. A., Different pattern of alkaline phosphatase,
osteopontin, and osteocalcin expression in developing rat bone visualized by in situ
hybridization, J. Bone Min. Res., 5, 831, 1990.
71. Sandberg, M., Autio-Harmainen, H., and Vuorio, E., Localization of the expression
of types I, Ill, and IV collagen, TGF-131 and c-fos genes in developing human calvarial
bones, Dev. Bioi., 130, 324, 1988.
72. Yoon, K., Buenaga, R., and Rodan, G. A., Tissue specificity and developmental
expression of osteopontin, Biochem. Biophys. Res. Comm., 148, 1129, 1987.
424 Nutrition and Gene Expression
73. Scharla, S. H., Strong, D. D., Mohan, S., Baylink, D. J., and Linkhart, T. A.,
1,25-Dihydroxyvitamin D, differentially regulates the production of insulin-like growth
factor I (IGF-1) and IGF-binding protein-4 in mouse osteoblasts, Endocrinology. 129,
3139, 1991.
74. Kurose, H., Yamaoka, K., Okada, S., Nakajima, S., and Seino, Y., 1,25-Dihy-
droxyvitamin D, [1,25-(0H) 2D 3] increases insulin-like growth factor I (IGF-I) receptors
in clonal osteoblastic cells. Study on interaction of IGF-I and 1,25-(0H),D,, Endocri-
nology. 126, 2088, 1990.
75. Petkovich, P.M., Wrana, J, L., Grigoriadis, A. E., Heersche, J. N. M., and Sodek,
J., 1,25-Dihydroxyvitamin D3 increases epidermal growth factor receptors and trans-
forming growth factor 13-like activity in a bone-derived cell line, J. Bioi. Chern., 262,
13424, 1987.
76. Gronowicz, G., Egan, J. J., and Rodan, G. A., The effect of 1,25-dihydroxyvitamin
D, on the cytoskeleton of rat calvaria and rat osteosarcoma (ROS 17/2. 8) osteoblastic
cells, J. Bone Min. Res., I, 441, 1986.
77. Lomri, A. and Marie, P. J., Changes in cytoskeletal proteins in response to parathyroid
hormone and I ,25-dihydroxyvitamin D in human osteoblastic cells, Bone Min., 10, I,
1990.
78. Hong, M. H., Jin, C. H., Toshiyuki, S., Ishimi, Y., Abe, E., and Suda, T., Tran-
scriptional regulation of the production of the third component of complement (C3) by
lcx,25-dihydroxyvitamin D 3 in mouse marrow-derived stromal cells (ST2) and primary
osteoblastic cells, Endocrinology, 129, 2774, 1991.
79. Shull, S., Tracy, R. P., and Mann, K. G., Identification of a vitamin D-responsive
protein on the surface of human osteosarcoma cells, Proc. Nat/. Acad. Sci. U.S.A., 86,
5405, 1989.
80. Majeska, R. J, and Rodan, G. A., The effect of 1,25(0H),D3 on alkaline phosphatase
in osteoblastic osteosarcoma cells, J. Bioi. Chern., 257, 3362, 1982.
81. Mulkins, M.A., Manolagas, S.C., Deftos, L. H., and Sussman, H. H., Dihydroxy-
vitamin D3 increases bone alkaline phosphatase enzyme levels in human osteogenic sar-
coma cells, J. Bioi. Chern., 258, 6219, 1983.
82. Murray, E. J., Murray, S. S., and Manolagas, S. C., Two-dimensional gel autora-
diographic analyses of the effects of 1.25-dihydroxycholecalciferol on protein synthesis
in clonal rat osteosarcoma cells, Endocrinology, 126, 2679, 1990.
83. Harrison, J. R., Petersen, D. N., Lichtler, A. C., Mador, A. T., Rowe, D. W., and
Kream, B. E., I ,25-Dihydroxyvitamin D3 inhibits transcription of type I collagen genes
in the rat osteosarcoma cell line ROS 17/2.8, Endocrinology, 125, 327, 1989.
84. Noda, M., Vogel, R. L., Craig, A. M., Prahl, J., DeLuca, H. F., and Denhardt,
D. T., Identification of a DNA sequence responsible for binding of the I ,25-dihydroxy-
vitamin D 3 receptor and I ,25-dihydroxyvitamin D, enhancement of mouse secreted phos-
phoprotein I (Spp-1 or osteopontin) gene expression, Proc. Nat/. Acad. Sci. U.S.A.,
87, 9995, 1990.
85. Kerner, S. A., Scott, R. A., and Pike, J, W., Sequence elements in the human osteo-
calcin gene confer basal activation and inducible response to hormonal vitamin D 3 , Proc.
Nat/. Acad. Sci. U.S.A., 86, 4455, 1989.
86. Lian, J. B., Stewart, C., Puchacz, E., Mackowiak, S., Shalhoub, V., Collart, D.,
Zambetti, G., and Stein, G., Structure of the rat osteocalcin gene and regulation of
vitamin D-dependent expression, Proc. Nat/. Acad. Sci. U.S.A., 86, 1143, 1989.
87. Demay, M. B., Gerardi, J. M., DeLuca, H. F., and Kronenberg, H. M., DNA
sequences in the rat osteocalcin gene that bind the I ,25-dihydroxyvitamin D, receptor
and confer responsive to I ,25-dihydroxyvitamin D,, Proc. Nat/. Acad. Sci. U.S.A., 87,
369, 1990.
88. Spiess, Y. H., Price, P. A., Deftos, J, L., and Manolagas, S.C., Phenotype-associated
changes in the effects of I ,25-dihydroxyvitamin D3 on alkaline phosphatase and bone
GLA-protein of rat osteoblastic cells, Endocrinology, 118, 1340, 1986.
Lian and Stein 425
89. Kim, Y. S., Birge, S. J,, Avioli, L. V., and Miller, R., Cell density-dependent vitamin
D effects on calcium accumulation in rat osteogenic sarcoma cells (ROS 17/2), Calcif.
Tissue Int., 41, 218, 1987.
90. Canalis, E. and Lian, J, B., I ,25-dihydroxyvitamin 0 3 effects on collagen and DNA
synthesis in periosteum and periosteum-free calvaria, Bone, 6, 457, 1985.
91. Franceschi, R. T. and Young, J,, Regulation of alkaline phosphatase by I ,25-dihy-
droxyvitamin D3 and ascorbic acid in bone-derived cells, J. Bone Min. Res., 5, 1157,
1990.
92. Kyeyune-Nyombi, E., Lao, W. K.-H., Baylink, D. J., and Strong, D. D., Stimulation
of cellular alkaline phosphatase activity and its messenger RNA level in a human osteo-
sarcoma cell line by I ,25-dihydroxyvitamin D3 , Arch. Biochem. Biophys., 275, 363,
1989.
93. Canalis, E., Effect of hormones and growth factors on alkaline phosphatase activity and
collagen synthesis in cultured rat calvariae, Metabolism, 32, 14, 1983.
94. Markose, E. R., Stein, J, L., Stein, G. S., and Lian, J, B., Vitamin D-mediated
modifications in protein-DNA interactions at two promoter elements of the osteocalcin
gene, Proc. Nat/. Acad. Sci. U.S.A., 87, 1701, 1990.
95. Pan, L. and Price, P. A., Ligand-dependent regulation of the I ,25-dihydroxyvitamin
0 3 receptor in rat osteosarcoma cells, J. Bioi. Chern., 262, 4670, 1987.
96. van Leeuwen, J, P. T. M., Birkenhager, J, C., Boorman, C. J,, Schilte, J, P., and
Pols, H. A. P., Functional involvement of calcium in the homologous up-regulation of
the 1,25-dihydroxyvitamin D, receptor in osteoblast-like cells, FEBS, 270, 165, 1990.
97. Baran, D. T., Sorensen, A. M., Honeyman, T. W., Ray, R., and Holick, M. F.,
la,25-dihydroxyvitamin D 3-induced increments in hepatocyte cytosolic calcium and ly-
sophosphatidylinositol: inhibition by pertussis toxin and la,25-dihydroxyvitamin D,, J.
Bone Min. Res., 5, 517, 1990.
98. Leiberherr, M., Effects of vitamin D3 metabolites on cytosolic free calcium in confluent
mouse osteoblasts, J. Bioi. Chern., 262, 13168, 1987.
99. Caffrey, J, M. and Farach-Carson, M. C., Vitamin D, metabolites modulate dihy-
dropyridine-sensitive calcium currents in clonal rat osteosarcoma cells, J. Bioi. Chern.,
264, 20265, 1989.
100. Nemere, I. and Norman, A. W., Rapid action of 1,25-dihydroxyvitamin D, on calcium
transport in perfused chick duodenum: effect of inhibitors, J. Bone Min. Res., 2, 99,
1987.
101. Farach-Carson, M. C., Sergeev, I., and Norman, A. W., Nongenomic actions of
I ,25-dihydroxyvitamin D, in rat osteosarcoma cells: structure-function studies using li-
gand analogs, Endocrinology, 129, 1876, 1991.
102. Gill, R. K. and Christakos, S., Identification of sequence elements in the mouse
calbindin-D28 k gene which confer basal activation and hormone inducible response, in
Vitamin D Gene Regulation Structure-Function Analysis and Clinical Application, Nor-
man, A. W., Bouillon, R., and Thomasset, M., Eds., Walter de Gruyter, Berlin, 1991,
36.
103. Darwish, H. M. and DeLuca, H. F., Identification of a I ,25-dihydroxyvitamin D,-
response element in the 5'-flanking region of the rat calbindin D-9k gene, Proc. Natl.
Acad. Sci. U.S.A., in press.
104. Morrison, N. A., Shine, J,, Fragonas, J.-C., Verkest, V., McMenemy, L., and
Eisman, J, A., 1,25-Dihydroxyvitamin D-responsive element and glucocorticoid repres-
sion in the osteocalcin gene, Science, 246, 1158, 1989.
105. Burger, E. H., Van Der Meer, J, W. M., Van De Gevel, J, S., Gribnau, J, C.
Thesingh, C. W., and Van Furth, R., In vitro formation of osteoclasis from long-term
culture of bone marrow mononuclear phagocytes, J. Exp. Med., 156, 1604, 1982.
106. Walker, D. G., Control of bone resorption by haematopoietic tissue. The induction and
reversal of congenital osteopetrosis in mice through use of bone marrow and splenic
transplants, J. Exp. Med., 142, 651, 1975.
426 Nutrition and Gene Expression
107. Bar-Shavit, Z., Teitelbaum, S. L., Reitsma, P., Hall, A., Pegg, L. E., Trial, J,, and
Kahn, A., Induction of monocytic differentiation and bone resorption by I ,25-dihy-
droxyvitamin 0 3 , Proc. Nat/. Acad. Sci. U.S.A., 80, 5907, 1983.
108. Holtrop, M. E. and Raisz, L. G., Comparison of the effects of I ,25-dihydroxychole-
calciferol, prostaglandin E2 and osteoclast-activating factor with parathyroid hormone on
the ultrastructure of osteoclasis in cultured long bones of fetal rats, Calc. Tissue Int.,
29, 201, 1979.
109. Takahashi, N., Akatsu, T., Sasaki, T., Yamaguchi, A., Kodama, H., Matin, T. J,,
and Soda, T., Osteoblastic cells are involved in osteoclast formation, Endocrinology,
123, 2600, 1988.
110. Udagawa, N., Takahashi, N., Akatsu, T., Sasaki, T., Yamaguchi, A., Kodama, H.,
Matin, T. J,, and Soda, T., The bone marrow-derived stromal cell lines MC3T3-G2/
PA6 and ST2 support osteoclast-like cell differentiation in co-culture with mouse spleen
cells, Endocrinology, 125, 1805, 1989.
Ill. Franceschi, R. T., Romano, P.R., and Park, K.-Y., Regulation of type I collagen
synthesis by I ,25-dihydroxyvitamin 0 3 in human osteosarcoma cells, J. Bioi. Chern.,
263, 18938, 1988.
112. Lian, J, B., Couttes, M. C., and Canalis, E., Studies of hormonal regulation of
osteocalcin synthesis in cultured fetal rat calvariae, J. Bioi. Chern., 60, 8706, 1985.
113. Aronow, M.A., Owen, T. A., Stein, G. S., and Lian, J. B., Estrogen inhibition of
osteocalcin mRNA expression occurs in cultured rat osteoblasts only after formation of
a mineralized extracellular matrix, J. Bone Min. Res., 5, S273, 1990.
114. Turner, R. T., Colvard, D. S., and Spelsberg, T. C., Estrogen inhibition of periosteal
bone formation in rat long bones: down-regulation of gene expression for bone matrix
proteins, Endocrinology, 127, 1346, 1990.
115. Schepmoes, G., Breen, E., Owen, T. A., Aronow, M. A., Stein, G. S., and Lian,
J, B., Influence of dexamethasone on the vitamin D-mediated regulation of osteocalcin
gene expression, J. Cell. Biochern., 47, 184, 1991.
116. Schiile, R., Umesono, K., Mangelsdorf, D. J., Bolado, J,, Pike, J, W., and Evans,
R. M., Jun-Fos and receptors for vitamins A and D recognize a common response element
in the human osteocalcin gene, Cell, 61, 497, 1990.
117. Nishimoto, S. K., Salka, C., and Nimni, M. E., Retinoic acid and glucocorticoids
enhance the effect of I ,25-dihydroxyvitamin 0 3 on bone 'Y-carboxyglutamic acid protein
synthesis by rat osteosarcoma cells, J. Bone Min. Res., 2, 571, 1987.
118. Noda, M., Transcriptional regulation of osteocalcin production by transforming growth
factor-[3 in rat osteoblast-like cells, Endocrinology, 124, 612, 1989.
119. Noda, M., Yoon, K., and Rodan, G. A., Cyclic AMP-mediated stabilization ofosteo-
calcin mRNA in rat osteoblast-like cells treated with parathyroid hormone, J. Bioi. Chern.,
263, 18574, 1988.
120. Theofan, G. and Price, P. A., Bone Gla protein messenger ribonucleic acid is regulated
by both 1,25-dihydroxyvitamin 0 3 and 3',5'-cyclic adenosine monophosphate in rat os-
teosarcoma cells, Mol. Endocrinol., 3, 36, 1989.
121. Stromstedt, P. E., Poellinger, L., Gustaffson, J, A., and Cralstedt-Duke, J,, The
glucocorticoid receptor binds to a sequence overlapping the TATA box of the human
osteocalcin promoter: a potential mechanism for negative regulation, Mol. and Cell Bioi.,
II, 3379, 1991.
122. Nanes, M.S., Rubin, J,, Titus, L., Hendy, G. N. and Catherwood, B. D., Interferon-
'/ inhibits I ,25-dihydroxyvitamin 0 3-stimulated synthesis of bone Gla protein in rat os-
teosarcoma cells by a pretranslational mechanism, Endocrinology, 127, 588, 1990.
123. Stein, G. S., Lian, J. B., Dworetzky, S. I., Owen, T. A., Bortell, R., Bidwell, J, P.,
and van Wijnen, A. J,, Regulation of transcription-factor activity during growth and
differentiation: involvement of the nuclear matrix in concentration and localization of
promoter binding proteins, J. Cell. Biochern., 47, 300, 1991.
Lian and Stein 427
124. Bortell, R., Owen, T. A., van Wijnen, A. J., Bidwell, J, P., Gavazzo, P., Breen,
E., DeLuca, H., Stein, G. S., and Lian, J, B., Vitamin D responsive protein/DNA
interactions at multiple promoter regulatory elements contribute to the level of osteocalcin
gene expression, Proc. Natl. Acad. Sci. U.S.A., 1992.
125. Terpening, C. M., Haussler, C. A., Jurutka, P. W., Galligan, M.A., Komm, B. S.,
and Haussler, M. R., The vitamin D-responsive element in the rat bone Gla protein
gene is an imperfect direct repeat that cooperates with other cis-elements in 1,25-dihy-
droxyvitamin D,-mediated transcriptional activation, Mol. Endocrinol., 5, 373, 1991.
126. Schiile, R., Rangarajan, P., Kliewer, S., Ransome, L. J,, Bolado, J,, Yang, N.,
Verma, I. M., and Evans, R. M., Functional antagonism between oncoprotein c-Jun
and the glucocorticoid receptor, Cell, 62, 1217, 1990b.
127. Owen, T. A., Bortell, R., Yocum, S. A., Smock, S. L., Zhang, M., Abate, C.,
Shalhoub, V., Aronin, N., Wright, K. L., van Wijnen, A. J,, Stein, J, L., Curran,
T., Lian, J. B., and Stein, G. S., Coordinate occupancy of AP-1 sites in the vitamin
D responsive and CCAAT box elements by fos-jun in the osteocalcin gene: a model for
phenotype suppression of transcription, Proc. Natl. Acad. Sci. U.S.A., 87, 9990, 1990.
128. Ozono, K., Liao, J., Kerner, S. A., Scott, R. A., and Pike, J, W., The vitamin D-
responsive element in the human osteocalcin gene, J. Bioi. Chern., 265, 21881, 1990.
129. Ross, T. K., Moss, V. E., Prahl, J. M., and DeLuca, H. F., A nuclear protein essential
for binding of rat 1,25-dihydroxyvitamin D3 receptor to its response elements, Proc.
Natl. Acad. Sci. U.S.A., in press.
130. Brown, T. A. and DeLuca, H. F., Phosphorylation of the I ,25-dihydroxyvitamin D3
receptor. A primary event in I ,25-dihydroxyvitamin D3 action, J. Bioi. Chern., 265,
10025, 1990.
131. Hsieh, J.-C., Jurutka, P. W., Galligan, M.A., Terpening, C. M., Haussler, C. A.,
Samuels, D. S., Shimizu, Y., Shimizu, N., and Haussler, M. R., Human vitamin D
receptor is selectively phosphorylated by protein kinase C on serine 51, a residue crucial
to its trans-activation function, Proc. Nat{. Acad. Sci. U.S.A., 88, 9315, 1991.
132. Lian, J, B., Stein, G. S., Bortell, R., and Owen, T. A., Phenotype suppression: a
postulated molecular mechanism for mediating the relationship of proliferation and dif-
ferentiation by fos/jun interactions at AP-I sites in steroid responsive promoter elements
of tissue-specific genes, J. Cell. Biochem., 45, 9, 1991.
133. Candeliere, G. A., Prud'homme, J,, and St-Arnaud, R., Differential stimulation of
fos and jun family members by calcitriol in osteoblastic cells, Mol. Endocrinol., 12,
1780, 1991.
134. Diamond, M. I., Miner, J, N., Yoshinaga, S. K., and Yamamoto, K. R., Transcription
factor interactions: selectors of positive or negative regulation from a single DNA element,
Science, 249, 1266, 1990.
135. Lian, J, B. and Stein, G. S., Transcriptional control of vitamin D regulated proteins,
J. Cell. Biochem., 49, 1992.
136. Feldman, D. and Malloy, P. J., Hereditary 1,25-dihydroxyvitamin D resistant rickets:
molecular basis and implications for the role of I ,25(0H),D 3 in normal physiology, Mol.
Cell. Endocrinol., 72, C57, 1990.
137. Marx, S. J,, Liberman, U. A., Eil, C., Gamblin, G. T., Degrange, D. A., and
Balsan, S., Hereditary resistance to I ,25-dihydroxyvitamin D3 suppresses parathyroid
hormone secretion from bovine parathyroid cells in tissue culture, Endocrinology, 117,
2214, 1984.
138. Liberman, U. and Marx, S. J,, Vitamin D dependent rickets, in Primer on Metabolic
Bone Diseases and Disorders of Mineral Metabolism, Fauvus, M. J., Ed., American
Society of Bone and Mineral Research, Kelseyville, CA, 1990, 178.
139. Glorieux, F. H., Rickets, the continuing challenge, New Engl. J. Med., 325, 1875,
1991.
428 Nutrition and Gene Expression
140. Marks, S. J., Jr., Osteopetrosis-multiple pathways for the interruption of osteoclast
functions, Appl. Pathol., 5, 172, 1987.
141. Shalhoub, V., Jackson, M. E., Lian, J. B., Stein, G. S., and Marks, S. C., Jr.,
Gene expression during skeletal development in three osteopetrotic rat mutations: evidence
for osteoblast abnormalities, J. Bioi. Chern., 266, 9847, 1991.
142. DeLuca, H. F., Newly discovered actions of 1,25-dihydroxyvitamin 0 3 and osteoporosis:
basis and treatment, Proc. Soc. Exp. Bioi. Med., 191, 1989.
143. Avioli, L. V., Significance of osteoporosis: a growing international health care problem,
Calcif. Tissue Int., 49, S5, 1991.
144. Eastell, R., Yergey, A. L., Vieira, N. E., Cedel, S. L., Kumar, R., and Riggs,
B. L., Interrelationship among vitamin D metabolism, true calcium absorption, parathy-
roid function, and age in women: evidence of an age-related intestinal resistance to I ,25-
dihydroxyvitamin A action, J. Bone Min. Res., 6, 125, 1991.
145. Franz, K. B., Abnormalities in parathyroid hormone secretion and I ,25-dihydroxyvi-
tamin 0 3 formation in women with osteoporosis, New Engl. J. Med., 320, 1697, 1989.
146. Silverberg, S. J., Shane, E., de Ia Cruz, L., Segre, G. V., Clemens, T. L., and
Bilezikian, J. P., Abnormalities in parathyroid hormone secretion and l ,25-dihydroxy-
vitamin 0 3 formation in women with osteoporosis, New Engl. J. Med., 320, 277, 1989.
147. Gallagher, J. C., Jerpbak, C. M., Lee, W. S. S., Johnson, K. A., DeLuca, H. F.,
and Riggs, B. L., 1,25-dihydroxyvitamin 0 3 : short- and long-term effects on bone and
calcium metabolism in patients with postmenopausal osteoporosis, Proc. Nat!. Acad. Sci.
U.S.A., 79, 3325, 1982.
148. Geusens, P., Vanderschueren, D., Verstraeten, A., Dequeker, J., Devos, P., and
Bouillon, R., Short-term course of I ,25(0H) 2 D, stimulates osteoblasts but not osteoclasis
in osteoporosis and osteoarthritis, Calcif. Tissue Int., 49, 168, 1991.
149. Tsai, K. S., Heath, H., Kumar, R., and Riggs, B. L., Impaired vitamin D metabolism
with aging in women. Possible role in pathogenesis of senile osteoporosis, J. Clin. Invest.,
73, 1668, 1984.
150. Gallagher, J. C. and Riggs, B. L., Action of 1,25-dihydroxyvitamin 0 3 on calcium
balance and bone turnover and its effect on vertebral fracture rate, Metabolism, 39, 30,
1990.
151. Aloia, J. F., Vaswani, A., Yeh, J. K., Ellis, K., Yasumura, S., and Cohn, S. H.,
Calcitriol in the treatment of postmenopausal osteoporosis, Am. J. Med., 84, 401, 1988.
152. Caniggia, A., Nuti, R., Galli, M., Lore, F., Turchetti, V., and Righi, G. A., Effect
of a long-term treatment with I ,25-dihydroxyvitamin 0 3 on osteocalcin in postmenopausal
osteoporosis, Calcif. Tissue Int., 38, 328, 1986.
153. Gallagher, J. C., Riggs, B. L., Recker, R. R., and Goldgar, D., The effect of calcitriol
on patients with postmenopausal osteoporosis with special reference to fracture frequency,
Proc. Soc. Exp. Bioi. Med., 191, 287, 1989.
154. Falch, J. A., Odegaard, 0. R., Finnanger, A.M., and Matheson, I., Postmenopausal
osteoporosis: no effect of three years treatment with 1,25-dihydroxycholecalciferol, Acta
Med. Scand., 221, 199, 1987.
155. Ott, S.M. and Chestnut, C. H., III, Calcitriol treatment is not effective in postmeno-
pausal osteoporosis, Ann. Int. Med., 110, 267, 1989.
156. Manolagas, S.C., Hustmyer, F. G., and Yu, Z.-P., Immunomodulating properties of
1,25-dihydroxyvitamin 0 3 , Kidney Int., 38, S9, 1990.
157. Lemire, J. M. and Archer, D. C., 1,25-dihydroxyvitamin D, prevents the in vivo
induction of murine experimental autoimmune encephalomyelitis, J. Clin. Invest., 87,
1103, 1991.
158. Kragballe, K., Gjertsen, B. T., De Hoop, D., Karlsmark, T., van de Kerkbof, P. C.,
Larko, 0., Nieboer, C., Roed-Peterson, J., Strand, A., and Tikjb, G., Double-blind,
right/left comparison of calcipotriol (MC903) and betamethasone valerate in treatment of
psoriasis vulgaris, Lancet, 337, 193, 1991.
Lian and Stein 429
!59. Okano, M., I a!pha,25-(0H) 2D 3 use on psoriasis and ichthyosis, Int. J. Dermatol., 30,
62, 1991.
160. Huckins, D., Felson, D. T., and Holick, M., Treatment of psoriatic arthritis with oral
1,25-dihydroxyvitamin 0 3 : a pilot study, Arthritis Rheum.. 33:1723-1727.
161. Ostrem, V. K., Tanaka, Y., Prahl, J., DeLuca, H. F., and Ikekawa, N., 24- and 26-
homo-1 ,25-dihydroxyvitarnin 0 3 : preferential activity in inducing differentiation of human
leukemic cells HL-60 in vitro, Proc. Nat/. Acad. Sci. U.S.A., 84, 2610, !987.
162. Paulson, S. K., Perlman, K., DeLuca, H. F., and Stern, P. H., 24- and 26-homo-
1,25-dihydroxyvitamin 0 3 analogs: potencies on in vitro bone resorption differ from those
reported for cell differentiation, J. Bone Min. Res., 5, 201, 1990.
163. Zhou, J.-Y., Norman, A. W., Lubbert, M., Collins, E. D., Uskokovik, M. R., and
Koeffler, H. P., Novel vitamin D analogs that modulate leukemic cell growth and
differentiation with little effect on either intestinal calcium absorption or bone calcium
mobilization, Blood, 74, 82, 1989.
164. Binderup, L. and Bramm, E., Effects of a novel vitamin D analog MC903 on cell
proliferation and differentiation in vitro and on calcium metabolism in vivo, Biochem.
Pharmacal., 37, 889, 1988.
165. Bouillon, R., Allewaert, K., Tan, B. K., van Baelen, H., de Clercq, P., and Vandewalle,
M., Biological activity of I ,25-dihydroxyvitamin D analogs: influence of relative affin-
ities for DBP and vitamin D receptor, in Vitamin D Gene Regulation Structure-Function
Analysis and Clinical Application, Norman, A. W., Bouillon, R., and Thomasset, M.,
Eds., Walter de Gruyter, Berlin, 1991, 131.
166. Evans, D. B., Thavarajah, M., Uskokovic, M. R., and Kanis, J. A., Increased potency
of I ,25-dihydroxyvitamin 0 3 on human osteoblast-like cells following structural side-
chain modification, Bone, II, 439, 1990.
167. Marie, P. J. Coones, D., Hott, M., and Miravet, L., Comparative effects of a novel
vitamin D analogue MC-903 and I ,25-dihydroxyvitamin 0 3 on alkaline phosphatase
activity, osteocalcin and DNA synthesis by human osteoblastic cells in culture, Bone,
II, 171, !990.
168. Valaja, T., Mahonen, A., Pirskanen, A., and Maenpaa, P. H., Affinity of 22-oxa-
1,25(0H) 2D3 for I ,25-dihydroxyvitarnin D receptor and its effects on the synthesis of
osteocalcin in human osteosarcoma cells, Biochem. Biophys. Res. Commun .. 169, 629,
1990.
169. Pernalete, N., Mori, T., Nishii, Y., Slatopolsky, E., and Brown, A. J., The activity
of22-oxacalcitriol in osteoblast-like (ROS 17/2.8) cells, Endocrinology, 129,778, 1991.
170. Tanaka, Y. and DeLuca, H. F., 26,26,26,27,27,27 -hexafluoro-1 ,25-dihydroxyvitamin
0 3 : a highly potent, long-lasting analog of 1,25-dihydroxyvitamin 0 3 , Arch. Biochem.
Biophys., 229, 348, 1984.
171. Kiriyama, T., Sumiaki, 0., Ejima, E., Kurihara, N., Hakeda, Y., Ito, N., Izumi,
M., Kumegawa, M., and Nagataki, S., Effect of a highly potent fluoro analog of I ,25-
dihydroxyvitamin 0 3 on human bone-derived cells, Endocrinology, 128, 81, 1991.
172. lnaba, M., Okuno, S., Inoue, A., Nishizawa, Y., Morii, H., and DeLuca, H. F.,
DNA binding property of vitamin 0 3 receptors associated with 26,26,26,27,27,27-hex-
afluoro-I,25(0H)2D3, Arch. Biochem. Biophys.. 268, 35, 1989.
173. Heinrichs, A., Stein, J., Lian, J. B. and Stein, G. S., unpublished observations.
174. Lian, J. B. and Stein, G. S., unpublished observations.
175. Lian, J. B., Stein, G. S., and Uskokovic, M., unpublished observation.
176. Tilyard, M. W., Spears, G. F. S., Thomson, J., and Dovey, S., Treatment of post-
menopausal osteoporosis with calcitriol or calcium, N. Engl. J. Med., 326, 357, 1992.
Chapter 19
TABLE OF CONTENTS
0·8493·6961-4/93/$0.00 +$.50
© 1993 by CRC Press, Inc. 431
432 Nutrition and Gene Expression
I. INTRODUCTION
Retinol and retinoic acid have profound effects on a wide range of phys-
iological and biochemical processes. 1-11 For example, the aldehyde derivative
of retinol (11-cis-retinal) functions in vision. 5 Retinol is converted to retinoic
acid (RA) in multiple tissues, 2-4 and RA has profound effects on pattern
formation during early development, skeletal growth, cellular growth, and
differentiation. 3.4. 6-11 The diverse effects of RA are due to its regulation of
the abundance and/or activity of specific proteins. As shown in Table 1, these
proteins include hormones and growth factors, 12-14 cell surface and nuclear
receptors, 15 -17·19 ·22 -26 and nuclear trans-acting factors, 18 ·20 ·21 specific en-
zymes/8·27-33 plasma proteins, 38 ·39 ·42 and proteins involved in cellular archi-
tecture and other cellular functions. 34-36 .4°·43 -47
RA regulates cellular functions by binding to intracellular retinoic acid
receptors (RR). 48 -57 Two distinct families of RR have been identified, i.e.,
RAR and RXR, each with multiple isoforms. The RAR family consists of
RARa and RAR[3, which are found in many tissues, and RAR')', which occurs
predominantly in skin and lung. 53 The RXR family consists of an a, [3, and
')' isoforms and RXR')' is found mainly in liver, lung, and muscle. 54 Even
though there is low sequence homology between RARs and RXRs, each binds
RA and DNA. Tissues like liver and white adipose tissue express multiple
forms of the RRs. 4H,4 9·55 ·56 The physiological significance of multiple receptor
isoform expression in tissues has not been explained.
Both RARs and RXRs are structurally similar to steroid and thyroid
hormone receptors (TR). 48 -54 Both function as ligand-activated transcription
factors in a manner similar to the regulation of gene expression described for
steroid and thyroid hormones. 51 RRs contain Zn-finger protein sequence mo-
tifs which function in receptor binding to DNA, at retinoic acid response
elements (RARE). The carboxyl terminal end of the molecule functions in
ligand binding. In contrast to steroid receptors, RR do not interact with heat
shock proteins, like HSP90, 57 proteins which prevent steroid receptor binding
to DNA. These observations suggest that RR may function more like TRs.
If analogous to TRs, RRs may be associated with the nuclear compartment
in the absence of ligand. RA binding to nuclear receptors sets in motion a
sequence of events that culminate in a change in transcription of the cis-
linked gene. Such transcriptional changes lead to corresponding changes in
mRNA coding for specific proteins and eventual changes in cell function.
The DNA targets for RR binding, i.e., RARE, have been the subject of
intense investigation. Umesono et al. 58 first reported that the thyroid hormone
response element (TRE) located upstream from the rat pituitary growth hor-
mone (GH) gene was a target for RAR binding and action. The GH TRE
used in these studies was palindromic and represented a modified version of
the TRE identified in the GH gene. The notion that RAR might utilize TREs
was reinforced by the finding that RA treatment of cultured pituitary cells
Jump et al. 433
TABLE 1
Cellular Proteins Regulated by Retinoic Acid
Induced (I) or
Protein Cellitissue repressed (R) Ref.
Hormones/Growth Factors
Enzymes
Other Proteins
TABLE 2
T 3 Binding Capacity in Rat Liver,
3T3-F442A, and 3T3-Ll Adipocytes
erbA alpha 2 -.
TR Beta 1 _.
FIGURE I. T, receptor isofonn analysis in mouse liver, white adipose tissue, 3T3-F442A,
and 3T3-LI adipocytes. Total RNA (25 f.Lg) from mouse liver, epididymal fat (WAT), 3T3-
F442A adipocytes (F442A), and 3T3-LI adipocytes (LI) was electrophoretically separated on
I% agarose-2.2 M formaldehyde gels and transferred to nitrocellulose. 33 Northern blots were
hybridized with eDNA inserts derived from rc-erbAa or rc-erbAI3 plasmids (generously provided
by H. Towle, Univ. of Minnesota). The rc-erbAa hybridized to two mRNAs: TRal, 5 kb;
c-erbA-2, 2.6 kb, while the rc-erbAI3 probe hybridizes to I mRNA of 6.2 kb 87 Duplicate
samples were electrophoresed for WAT, F442A, and Ll adipocytes. Rat liver ISS and 28S
ribosomal RNAs served as standards and were visualized by ethidium bromide staining.
Ll cells were also cloned from mouse embryonic fibroblast. 79 We have found
that following a 72 h treatment of Ll adipocytes with T 3 , mRNA 514 was
induced fivefold. Analysis of T 3 binding capacity in these cells showed that
TR were expressed at 70% of the level observed in rat liver (Table 2). Thus,
different adipocyte cell lines display different T 3 regulatory profiles.
Molecular cloning studies have shown that at least three ligand-binding
isoforms of TR, i.e., c-erbAai (TRal), c-erbAJ31 (TRJ31), and c-erbAJ32
(TRJ32), are expressed in various rodent tissues. 87 - 92 While TRJ32 is pituitary
specific, TRal and TRJ31 are expressed in several tissues. 87•88 In addition,
three variants of these proteins (i.e., c-erba2, c-erba3, and rev-erbAa) have
been described which do not bind T 3 . 88 -93 Northern analysis was used to
examine the TR isoforms expressed in mouse liver and WAT, and 3T3-F442A
and 3T3-Ll adipocytes (Figure 1). A rat liver c-erbAa eDNA hybridized to
two mRNAs in liver, WAT, and both adipocyte cell lines. The 5-kb mRNA
codes for TRal, while the 2.5-kb mRNA codes for c-erbAa2. The hybrid-
ization of rc-erbAa to liver RNA is weak when compared to the hybridization
signal in WAT, F442A, and Ll. TRJ31 (6.2 kb) was expressed in both liver
and WAT, but not in either adipocyte cell lines. The relative abundance of
TRa I, c-erbAa2, and TRJ31 in liver and Ll cells agrees favorably with
previous reports. 87
The differential T 3 responsiveness of F442A and Ll is due, at least in
part, to differences in T 3 binding capacity. Ifthese cells are "true" progenitors
to adipocytes in WAT, then the absence of TRJ31 expression in F442A and
Jump et al. 437
TABLE 3
Dexamethasone and Retinoic Acid Regulation of mRNA814 Levels in
3T3-F442A and 3T3-Ll Cells•
3T3-F442A 3T3-Ll
Treatment Units Fold Units Fold
' Adipocytes were exposed to I f.LM dexamethasone, retinoic acid, or a combination of treat-
ments for 72 h. 33 Control cultures were exposed to vehicle (0.1% ethanol) for 72 h. RNA
was extracted and analyzed for expression of mRNA 514 . Results are expressed as mean +
SD. Number of determinations ranged from 6 to 12 for each treatment.
Ll, and the low T 3 binding capacity in F442A cells, suggests that the T3
regulatory system in adipocyte precursors might be under developmental con-
trol. The F442A and Ll cells might represent early to intermediate stages in
development, and thus they may not be satisfactory models for T3 action.
RAR Alpha
RAR Beta
FIGURE 2. Retinoic acid receptor isofonn analysis in mouse liver, 3T3-F442A, and 3T3-Ll
cells. Total mRNA (25 flog) isolated from mouse liver (lane I) and 3T3-F442A preadipocytes
(lanes 2,3), 3T3-F442A adipocytes (lanes 4,5), 3T3-Ll preadipocytes (lanes 6, 7), and 3T3-Ll
adipocytes (lanes 8,9) was electrophoretically separated in I% agarose-2.2 M formaldehyde
gels." Following electrophoresis RNA was transferred to nitrocellulose and hybridized with
either 32 P-labeled inserts derived from pRARaO (RAR alpha) or pRAR[3 (RAR beta) plasmids.
RARa hybridizes to two mRNA of 2.8 and 3.8 kb, while RAR[3 hybridizes to two mRNAs of
3.2 and 3.6 kb." Rat liver 18S and 28S ribosomal RNAs served as standards and were visualized
by ethidium bromide staining.
RARa and RARI3 were expressed in rat white epididymal fat (not shown).
If the mRNA levels coding for these various RAR isoforms reflect the level
of active receptor in these cells, then RARa is the predominant RAR isoform
expressed in both F442A and Ll cells. In addition, RARI3 is upregulated by
RA in some cell types. 16 • 17 We found no evidence for an effect of RA on
RARI3 expression in either preadipocytes or adipocytes (not shown).
V. IDENTIFICATION OF PROSPECTIVE RA
AND GLUCOCORTICOID RESPONSIVE
CIS-ACTING ELEMENTS
514-2.1-CAT
...
lllllllllllllllllllllllllllllllllllllllllllllllll CAT I
-2110 +1'9
514-1 6-CAT
...
OJIIIIIII!liiiiiiUIIIIIIIIIIIIIII CAT I
-1588 +u - Dexamethasone
... CSJ Retinoic Acid
514-1 I-CAT Jllllllfflll\111111111 CA~
-1075 +19
514- 29-CAT
...
1111111 CAT I
-2'90 +U
075 290
514-PA-CAT
-1588 ~
FIGURE 3. Analysis of retinoic acid and glucocorticoid induction of chloramphenicol acetyltransferase (CAT) expression in various deletion mutations of the
Sl4 promoter. 3T3-F442A fibroblasts were stably transfected with plasmids containing various 5' deletions of the SJ4 promoter. The location of the TRE in
pSl4-4.3-CAT is between -2750 and -2550 bp. Stably transfected cells were differentiated to adipocytes and treated with I ].1M RA or DEX for 72 h. CAT
activity was assayed and results are expressed as fold induction, mean ± SD, N = 3 to 6.
t
.....
442 Nutrition and Gene Expression
100 - MMTV-CAT
90 ISSSI TK14M-CAT
80 CJ RSV -CAT
70 ~ S14 -PA-CAT
rn
·c:
+'
;:)
60
50
:f;.
·;; 40
:,J
()
< 30
~ 20
u
10
0 ~~
Veh Dex RA
is
Veh
i Dex RA
Precdipocytes Adipocytes
-1554
Reg ion 1
-1 530
-1 477
Reg io n 2
-1 457
FIGURE 5. DNase I footprint analysis of the S 14 enhancer region. A DNA fragment extending
from - 1380 to -1588 bp relative to the Sl4 gene transcription start site was isolated. The
noncoding strand was ·12 P-labeled with T 4 polynucleotide kinase and used for DNase I footprint
analysis. 103 In lanes labeled "D" , the 32 P-labeled DNA was treated with dimethyl sulfate to
generate a G-ladder. In lanes labeled " P" and "A", the 32 P-DNA was mixed with nuclear
proteins (30 JLg) extracted from Ll preadipocyte and Ll adipocyte nuclei , respectively. Samples
were digested with DNase I; the 32 P-DNA was extracted and electrophoresed on a 8% acrylamide
sequencing gel, dried , and exposed to X-ray film.
'- pre-mRNA
Promoter
TABLE 4
Changes in DNA-Protein Interaction within the S14
Tissue-Specific Enhancer during Adipocyte
Differentiation
' Location of protected regions relative to the 5' end of the S 14 gene.
ACKNOWLEDGMENTS
The authors wish to thank Drs. Maija Zile and Henry Bayley for many
helpful suggestions during the preparation of this manuscript. We also wish
to thank Dr. P. Chambon, B. Spiegelman, H. Towle, L. Kedes, H-S. Sui,
R. Koenig, and S. Conrad for the various cDNAs, expression vectors, and
adipocyte cell lines used in these studies. This research was supported by
grants from the National Institutes of Health (GM36851, DK43220), Amer-
ican Diabetes Association, The Upjohn Company, and Michigan State Uni-
versity.
REFERENCES
I. Wolbach, S. B., Effects of vitamin A deficiency and hypervitaminosis A in animals, in
The Vitamins, Vol. I, Sebrell, W. B., Jr. and Harris, R. S., Eds., Academic Press, New
York, 1954, 106.
2. Ganguly, J., Rao, M. R. S., Murthy, S. K., and Sarada, K., Systemic mode of action
of vitamin A, in Vitamins and Hormones, Vol. 38, Munson, P. L., Diczfalusy, E.,
Glover, J., and Olson, R. E., Eds., 1980, 1.
3. Roels, 0. A., Biochemical systems, in The Vitamins, 2nd ed., Vol. I, Sebrell, W. H.,
Jr. and Harris, R. S., Eds., Academic Press, New York, !967, 245.
4. Wolf, G., Multiple functions of Vitamin A, Physiol. Rev., 64, 873, 1984.
5. Wald, G., The visual function of the Vitamins A, Vitam. Horm., 18, 417, 1960.
6. Napoli, J. L., The biogenesis of retinoic acid: a physiologically significant promoter of
differentiation, in Chemistry and Biology of Synthetic Retinoids, Dawson, M. I. and
Okamura, W. H., Eds., CRC Press, Boca Raton, FL, 229, 1990.
7. Sani, B. B., Cellular retinoic acid binding proteins and action of retinoic acid, in Chem-
istry and Biology of Synthetic Retinoids, Dawson, M. I. and Okamura, W. H., Eds.,
CRC Press, Boca Raton, FL, 1990, 365.
8. Wolf, G., Recent progress in vitamin A research: nuclear retinoic acid receptors and
their interaction with gene elements, J. Nutr. Biochem., 1, 284, 1991.
9. Chytil, F., Retinoic acid: biochemistry and metabolism, J. Am. Acad. Dermatol .. IS,
741, 1986.
10. Melton, D. A., Pattern formation during animal development, Science, 252, 234, 1991.
11. Blomhoff, R., Green, M. H., Berg, T., and Norum, K. R., Transport and storage of
vitamin A, Science, 250, 399, 1990.
Jump eta!. 449
12. Morita, S., Fernadez-Mejia, C., and Melmed, S., Retinoic acid selectively stimulates
growth hormone secretion and messenger ribonucleic acid levels in rat pituitary cells,
Endocrinology, 124, 2052, 1989.
13. Bedo, G., Santisteban, P., and Aranda, A., Retinoic acid regulates growth hormone
gene expression, Nature (London), 339, 231, 1989.
14. Glick, A. B., Flanders, K. C., Danielpour, D., Yuspa, S. H., and Sporn, M. B.,
Retinoic acid induces transforming growth factor ~2 in cultured keratinocytes and mouse
epidermis, Cell Regul., I, 87, 1989.
15. Petkovich, P. M., Heersche, J. N. M., Tinker, D. 0., and Jones, G., Retinoic acid
stimulates I ,25-dihydroxyvitamin D, binding in rat osteosarcoma cells, J. Bioi. Chem.,
259, 8274, 1984.
16. Sucov, H. M., Murakami, K. K., and Evans, R. M., Characterization of an auto-
regulated response element in the mouse RAR~ gene, Proc. Nat/. Acad. Sci. U.S.A.,
87, 5392, 1990.
17. de The, H., Marchio, A., Tiollais, P., and Dejean, A., Differential expression and
ligand regulation of the RAR a and ~ genes, EMBO J., 8, 429, 1989.
18. Lafyatis, R., Kim, S-J., Angel, P., Roberts, A. B., Sporn, M. B., Karin, M., and
Wilder, R., lnterleukin-1 stimulates and all-trans retinoic acid inhibits collagenase gene
expression through its 5' activator protein-1-binding site, Mol. Endocrinol., 4, 973, 1990.
19. Clarke, C. L., Roman, S. D., Graham, J., Koga, M., and Sutherland, R. L.,
Progesterone receptors regulated by retinoic acid in human breast cancer cell line T-4 7D,
J. Bioi. Chem., 265, 12694, 1990.
20. Suva, L. J., Ernst, M., and Rodan, G. A., Retinoic acid induces zif268 early gene
expression in rat preosteoblastic cells, Mol. Cell. Bioi., 11, 2503, 1991.
21. Luscher, B., Mitchell, P., Williams, T., and Tjian, R., Regulation of transcription
factor AP-2 by the morphogen retinoic acid and second messengers, Genes Dev., 3,
1507, 1989.
22. Chakraborty, A. K., Orlo, S. J., and Pawelek, J. M., Stimulation of the receptor for
melanocyte stimulating hormone by retinoic acid, FEBS, 276, 205, 1989.
23. Side!, N., Taga, T., Hirano, T., Kishimoto, T., and Saxon, A., Retinoic acid induced
growth inhibition of a human myeloma cell line via down-regulation of IL-6 receptors,
J. Immunol., 146, 3809, 1991.
24. Sidell, N. and Ramsdell, F. R., Retinoic acid upregulates interleukin-2 receptors on
activated human thymocytes, Cell. Immunol., 115, 299, 1988.
25. Junquero, D., Modat, G., Coquelet, C., and Bonne, C., Retinoids enhance the number
ofEGF receptors in corneal endothelial cells, Exp. Eye Res., 51, 49, 1990.
26. Ubels, J. L., Iorfino, A., and O'Brien, W. J., Retinoic acid decreases the number of
EGF receptors in corneal epithelium and Chang conjunctival cells, Exp. Eye Res., 52,
763, 1991.
27. Chiocca, E. A., Davies, P. J, A., and Stein, J, P., The molecular basis of retinoic
acid action, J. Bioi. Chem., 263, 11584, 1988.
28. Imai, Y., Rodan, S. B., and Rodan, G. A., Effects of retinoic acid on alkaline phos-
phatase mRNA, catecholamine receptors and G-proteins in ROS 17/2.8 cells, Endocri-
nology, 122, 456, 1988.
29. Lucas, P. C., O'Brien, R. M., Mitchell, J, A., Davis, C. M., lmai, E., Forman,
B. M., Samuels, H. H., and Granner, D. K., A retinoic acid response element is part
of a pleiotropic domain in the PepCK gene, Proc. Nat/. Acad. Sci. U.S.A., 88, 2184,
1991.
30. Chan, S.D. H., Strewler, G. J., and Nissensa, R. A., Transcription activation of Gsa
gene expression by retinoic acid and parathyroid hormone-related protein in F9 terato-
carcinoma cells, J. Bioi. Chem., 265, 20081, 1990.
31. Duester, G., Shear, M. L., McBride, M.S., and Stewart, M. J., Retinoic acid response
element in human alcohol dehydrogenase ADH3: Implication for regulation of retinoic
acid synthesis, Mol. Cell. Bioi., 11, 1638, 1991.
450 Nutrition and Gene Expression
32. Darrow, L., Riches, R. j., Pecorino, L. T., and Strickland, S., Transcription factor
SPl is important for retinoic acid induced expression of tissue plasminogen activator gene
during F9 teratocarcinoma cell differentiation, Mol. Cell. Bioi., 10, 5883, 1990.
33. Lepar, G. j. and Jump, D. B., Retinoic acid and dexamethasone interact to regulate
S 14 gene transcription in 3T3-F442A adipocytes, Mol. Cell. Endocrinology, 77, 84, 65,
1992.
34. Strickland, S., Smith, K. K., and Marotti, K. K., Hormonal induction of differentiation
in teratocarcinoma stem cells: generation of parietal endoderm by retinoic acid and db-
cAMP, Cell, 21, 347, 1980.
35. Wang, S. Y. and Gudas, L. J,, Isolation of eDNA clones specific for collagen IV and
laminin from mouse teratocarcinoma, Proc. Nat/. Acad. Sci. U.S.A.. 80, 5880, 1983.
36. Vasios, G. W., Gold, J. D., Petkovich, M., Chambon, P., and Gudas, L. J,, A
retinoic acid response element is present in the 5' flanking region of the laminin 131 gene,
Proc. Nat!. Acad. Sci. U.S.A .. 86, 9099, 1989.
37. Wei, L-N., Blaner, W. S., Goodman, D. S., and Nguyen-Huu, C. M., Regulation of
cellular retinoid-binding proteins and their mRNAs during embryonal carcinoma cell
differentiation induced by retinoic acid, Mol. Endocrinol., 3, 454, 1989.
38. Munoz-Canoves, P., Vik, D. P., and Tack, B. F., Mapping a retinoic acid responsive
element in the promoter region of the complement factor H gene, J. Bioi. Chern., 265,
20065, 1990.
39. Antras, j., Lasnier, F., and Pairault, J,, Adipsin gene expression in 3T3-F442A
adipocytes is post-transcriptionally down-regulated by retinoic acid, J. Bioi. Chern .. 266,
1157, 1991.
40. Stellmach, V., Leask, A., and Fuchs, E., Retinoid mediated transcriptional regulation
of keratin genes in human epidermal and squamous carcinoma cells, Proc. Nat/. Acad.
Sci. U.S.A.. 88, 4582, 1991.
41. Hohl, D., Lichti, U., Breitkreutz, D., Steinert, P.M., and Roop, D. R., Transcription
of the human loricrin gene in vitro is induced by calcium and cell density and suppressed
by retinoic acid, J. Invest. Derrnatol., 96, 414, 1991.
42. Rottman, J. N., Widom, R. L., Nadal-Ginard, B., Mahdavi, V., and Karathanasis,
S. K., A retinoic acid response element in the apolipoprotein AI gene distinguishes
between two different retinoic acid pathways, Mol. Cell Bioi., II, 3814, 1991.
43. Scheibe, R. J., Ginty, D. D., and Wagner, J. A., Retinoic acid stimulates differentiation
of PC12 cell that are deficient in cAMP-dependent protein kinase A, J. Cell Bioi., 113,
1173, 1991.
44. Pacifici, M., Golden, E. B., Iwamoto, M., and Adams, S. L., Retinoic acid treatment
induces type X collagen gene expression in cultured chick chondrocytes, Exp. Cell Res.,
195, 38, 1991.
45. Glover, A., Oshima, R. G., and Adamson, E. D., Epithelial layer formation in dif-
ferentiation aggregates of F9 embryonal carcinoma cells, J. Cell Bioi., 96, 1690, 1983.
46. Linder, S. K., Krondahl, U., Sennerstrom, R., and Ringerty, N. R., Retinoic acid
induced differentiation of F9 embryonal carcinoma cells, Exp. Cell Res.. 132, 453, 1981.
47. Lehtonen, E., Lehto, V. P., Badley, R. A., and Virtanen, 1., Formation of vinculin
plaques precedes other cytoskeletal changes during retinoic acid induced teratocarcinoma
cells differentiation, Exp. Cell Res., 144, 191, 1983.
48. Petkovich, M., Brand, N.J., Krust, A., and Chambon, P., A human retinoic acid
receptor which belongs to the family of nuclear receptors, Nature (London), 330, 444,
1987.
49. Giguere, V., Ong, E. S., Segui, P., and Evans, R. M., Identification of a receptor for
the morphogen retinoic acid. Nature (London), 330, 624, 1987.
50. Brand, N., Petkovich, M., Krust, A., Chambon, P., de The, H., Marchio, A.,
Tiollais, P., and Dejean, A., Identification of a second human retinoic acid receptor,
Nature (London), 332, 850, 1988.
Jump et al. 451
51. Evans, R. M., The steroid and thyroid hormone receptor superfamily, Science, 240,
889, 1988.
52. Benbrook, D., Lenhardt, E., and Pfahl, M., A new retinoic acid receptor identified
from a hepatocellular carcinoma. Nature (London), 333, 669, 1988.
53. Zelent, A., Krust, A., Petkovich, M., Kastner, P., and Chambon, P., Cloning of
murine a and J3 retinoic acid receptors and a novel 'Y receptor predominantly expressed
in skin, Nature (London), 339, 714, 1989.
54. Mangelsdorf, D. J., Ong, E. S., Dyck, J, A., and Evans, R. M., Nuclear receptor
that identifies a novel retinoic acid response pathway, Nature (London), 345, 224, 1990.
55. DeLuca, L. M., Retinoids and their receptors in differentiation, embryogenesis and
differentiation, FASEB J., 5, 2924, 1991.
56. Haq, R. U. and Chytil, R., Expression of nuclear retinoic acid receptors in rat adipose
tissue, Biochem. Biophys. Res. Comm., 176, 1539, 1991.
57. Dalman, F. C., Sturzenbecker, L. J., Levin, A. A., Lucas, D. A., Perdew, G. W.,
Petkovich, M., Chambon, P., Grippo, J, F., and Pratt, W. B., Retinoic acid receptor
belongs to a subclass of nuclear receptors that do not form docking complexes with hsp90,
Biochemistry, 30, 5605, 1991.
58. Umesono, K., Giguere, V., Glass, C. K., Rosenfeld, M. G., and Evans, R. M.,
Retinoic acid and thyroid hormone induce gene expression through a common responsive
element, Nature (London), 336, 262, 1988.
59. Umesono, K., Murakami, K. K., Thompson, C. C., and Evans, R. M., Direct repeats
as selective response elements for the thyroid hormone, retinoic acid and vitamin 0 3
receptors, Cell, 65, 1255, 1991.
60. Mangelsdorf, D. J., Umesono, K., Kliewer, S. A., Borgmeyer, U., Ong, E. S., and
Evans, R. M., A direct repeat in the cellular retinol-binding protein type II gene confers
differential regulation by RXR and RAR, Cell, 66, 555, 1991.
61. Naar, A. M., Boutin, J.-M., Lipkin, S. M., Yu, V. C., Holloway, J. M., Glass,
C. K., and Rosenfeld, M.G., The orientation and spacing of core DNA-binding motifs
dictate selective transcriptional responses to three nuclear receptors, Cell, 65, 1267, 1991.
62. Glass, C. K., Lipkin, S. M., Devary, 0. V., and Rosenfeld, M. G., Positive and
negative regulation of gene transcription by a retinoic acid-thyroid hormone receptor
heterodimer, Cell, 59, 697, 1989.
63. Forman, B. M., Yang, C.-R., Au, M., Casanova, J., Ghysdael, J., and Samuels,
H. H., A domain containing leucine-zipper-like motifs mediate novel in vivo interaction
between the thyroid hormone and retinoic acid receptors, Mol. Endocrinol., 3, 1610,
1989.
64. Jump, D. B., Narayan, P., Towle, H. C., and Oppenheimer, J, H., Rapid effects of
triiodothyronine on hepatic gene expression, J. Bioi. Chem .. 259. 2789, 1984.
65. Jump, D. B. and Oppenheimer, J. H., High basal expression and 3.5. 3' -triiodothy-
ronine regulation of mRNA" 4 in lipogenic tissues. Endocrinology, 117, 2259, 1985.
66. Jump, D. B., Wong, N. C. W., and Oppenheimer, J, H., Chromatin structure and
methylation state of a thyroid hormone responsive gene in rat liver, J. Bioi. Chem., 262,
778, 1985.
67. Jump, D. B., Thyroid hormone regulation of rat liver Sl4 gene expression, in Gene
Regulation by Steroids Hormones IV, Roy. A. K. and Clarke. J. H., Eds., Springer-
Verlag, New York, 1989, 144.
68. Jump, D. B., Rapid induction of rat liver S14 gene transcription by thyroid hormone,
J. Bioi. Chem., 264, 4698, 1989.
69. Jump, D. B., Bell, A., and Santiago, V., Thyroid hormone and dietary carbohydrate
interact to regulate rat liver S 14 gene transcription and chromatin structure, J. Bioi.
Chem., 265, 3474, 1990.
70. Jump, D. B., Bell, A., Lepar, G., and Hu, D., Insulin rapidly induces rat liver S14
gene transcription, Mol. Endocrinol., 4, 1655, 1990.
452 Nutrition and Gene Expression
71. Oppenheimer, J, H., Schwartz, H. L., Mariash, C. N., Kinlaw, W. B., Wong,
N. C. W., and Freake, H. C., Advances in our understanding of thyroid hormone action
at the cellular level, Endocrine Rev., 8, 288, 1987.
72. Kinlaw, W. B., Ling, N.C., and Oppenheimer, J. H., Identification ofrat Sl4 protein
and comparison of its regulation with that of mRNA 514 employing synthetic peptide
antisera, J. Bioi. Chern., 264, 19779, 1989.
73. Lepar, G. J. and Jump, D. B., Hormonal regulation of the Sl4 gene in 3T3-F442A
cells, Mol. Endocrinol., 3, 1207, 1989.
74. Clarke, S. D., Armstrong, M. K., and Jump, D. B., Nutritional control of rat liver
fatty acid synthase and Sl4 mRNA abundance, J. Nutr., 120, 218, 1990.
75. Clarke, S. D., Armstrong, M. K., and Jump, D. B., Dietary polyunsaturated fats
uniquely suppress rat liver fatty acid synthase and Sl4 mRNA content, J. Nutr., 120,
225, 1990.
76. Blake, W. L. and Clarke, S.D., Suppression of rat hepatic fatty acid synthase and Sl4
gene transcription by dietary polyunsaturated fat, J. Nutr., 120, 1727, 1990.
77. Jump, D. B., Veit, A. M., Santiago, V. S., Lepar, G., and Herberholz, L., Tran-
scriptional activation of the rat liver S 14 gene during post-natal development, J. Bioi.
Chern., 263, 7254, 1988.
78. Goodridge, A. G., Dietary regulation of gene expression: enzymes involved in carbo-
hydrate and lipid metabolism, Annu. Rev. Nutr., 7, l 57, 1987.
79. Green, H. and Kehinde, 0., Subhnes of mouse 3T3 cells that accumulate lipid, Cell,
I, 113, 1974.
80. Spiegelman, B. M., Frank, M., and Green, H., Molecular cloning of mRNA from
3T3 adipocytes, J. Bioi. Chern., 258, 10083, 1983.
81. Bernlohr, D. A., BoJanowski, M.A., Kelly, T. J., Jr., and Lane, M.D., Evidence
for an increase in transcription of specific mRNA during differentiation of 3T3-Ll pre-
adipocytes, J. Bioi. Chern., 260, 5563, 1985.
82. Wise, L. S., Sui, H. S., and Rubin, C. S., Coordinate regulation of the biosynthesis
of ATP-citrate lyase and malic enzyme during adipocyte differentiation, J. Bioi. Chern.,
259, 4827, 1984.
83. Oppenheimer, J, H., Schwartz, H. L., and Surks, M. I., Tissue differences in the
concentration ofT, nuclear binding sites in the rat liver, kidney, pituitary, heart, brain,
spleen and testes, Endocrinology, 95. 897, 1974.
84. Buergi, U. and Abuehl, U., Nuclear triiodothyronine binding sites in rat adipocytes,
Metabolism, 33, 326, 1984.
85. Hausdorf, S., Clement, J., and Loos, U., Expression of Sl4 gene during differentiation
of 3T3-Ll cells, Horm. Metab. Res., 20, 723, 1988.
86. Moustaid, N. and Sui, H. S., Regulation of expression of the fatty acid synthase gene
in 3T3-Ll cells by differentiation and triiodothyronine, J. Bioi. Chern., 266, 18550,
1991.
87. Murray, M. B., Zilz, N., McCreary, N. L., MacDonald, M. J., and Towle, H. C.,
Isolation and characterization of rat eDNA clones for two distinct thyroid hormone re-
ceptors, J. Bioi. Chern., 263, 12770, 1988.
88. Hodin, R. A., Lazar, M.A., Wintman, B. I., Darling, D. S., Koenig, R. J,, Larsen,
P. R., Moore, D. D., and Chin, W. W., Identification of a thyroid hormone receptor
that is pituitary specific, Science, 244, 76, 1989.
89. lzumo, S. and Madhavi, V., Thyroid hormone receptor isoforms generated by alternative
splicing differentially activate myosin HC gene transcription, Nature (London), 334, 539,
1988.
90. Lazar, M. A., Hodin, R. A., Darling, D. S., and Chin, W. W., Identification of a
rat c-erbA-related protein which binds deoxyribonucleic acid, but does not bind thyroid
hormone, Mol. Endocrinol., 2, 893, 1988.
Jump et al. 453
91. Mitsuhashi, T., Tennyson, G. E., and Nikodem, V. M., Alternative splicing generates
messages encoding rat c-erbA proteins that do not bind thyroid hormone, Proc. Nat/.
Acad. Sci. U.S.A.. 85. 5804, 1988.
92. Koenig, R. J., Lazar, M. A., Hodin, R. A., Brent, G. A., Larsen, P. R., Chin,
W. W., and Moore, D. D., Inhibition of thyroid hormone action by a non-hormone
binding c-erbA protein generated by alternative mRNA splicing, Nature, 337, 659, 1989.
93. Cook, C. B. and Koenig, R. J., Expression of erbAo: and [3 mRNAs in regions of adult
rat brain, Mol. Cell. Endocrinol., 70. 13, 1990.
94. Kuri-Harcuch, W., Differentiation of 3T3-F442A cells into adipocytes is inhibited by
retinoic acid, Differentiation, 23, 164, 1982.
95. Castro-Munozledo, F., Marsch-Moreno, M., Beltran-Langarica, A., and Kuri-
Harcuch, W., Commitment of adipocyte differentiation in 3T3 cells is inhibited by
retinoic acid and the expression of lipogenic enzymes in modulated through cytoskeleton
stabilization, Differentiation, 36, 211, 1987.
96. Pairault, J. and Lasnier, F., Control of adipogenic differentiation of 3T3-F442A cells
by retinoic acid, dexamethasone and insulin: a topographic analysis, J. Cell Physiol.,
132, 279, 1987.
97. Stone, R. L. and Bernlohr, D. A., The molecular basis for inhibition of adipose
conversion of murine 3T3-Ll cells by retinoic acid, Differentiation, 45, 119, 1990.
98. Feigner, P. L., Gadek, T. R., Holm, M., Roman, R., Chan, H. W., Wenz, M.,
Northrop, J. P., Ringold, G. M., and Danielsen, M., Lipofection: a highly efficient,
lipid-mediated DNA-transfection procedure, Proc. Nat/. Acad. Sci. U.S.A., 84, 7413,
1987.
99. Gorman, C. M., Moffat, L. F., and Howard, B. H., Recombinant genomes which
express chloramphenicol acetyltransferase in mammalian cells, Mol. Cell Bioi., 2, 1044,
1982.
100. Zilz, N. D., Murray, M. B., and Towle, H. C., Identification of multiple thyroid
hormone response elements located far upstream from the rat S14 promoter, J. Bioi.
Chern., 265, 8136, 1990.
101. Chandler, V. L., Maler, B. A., and Yamamoto, K. R., DNA sequences bound spe-
cifically by glucocorticoid receptor in vitro render a heterologous promoter hormone
responsive in vivo, Cell, 33, 489, 1983.
102. Brent, G., Larsen, P.R., Harney, J. W., Koenig, R. J., and Moore, D. D., Functional
characterization of the rat growth hormone promoter elements required for induction by
thyroid hormone with and without a co-tranfected [3-type thyroid hormone receptor, J.
Bioi. Chern., 264, 178, 1989.
103. MacDougald, 0. A. and Jump, D. B., Identification of functional cis-acting elements
in the S14 promoter, Biochern. J., 280, 761, 1991.
104. Gross, D. S. and Garrard, W. T., Nuclease hypersensitive sites in chromatin, Annu.
Rev. Biochern., 57, 159, 1988.
105. Jacoby, D. B., Zilz, N.D., and Towle, H. C., Sequences within the 5' flanking region
of the S14 gene confer responsiveness to glucose in primary hepatocytes, J. Bioi. Chern.,
264, 17623, 1989.
106. Distel, R. J., Ro, H.-S., Rosen, B. S., Groves, D. L., and Spiegelman, B. M.,
Nucleoprotein complexes that regulate gene expression in adipocyte differentiation: direct
participation of c-fos, Cell, 49, 835, 1987.
107. Herrera, R. 0., Robinson, H. S., Xanthopoulos, K. G., and Spiegelman, B. M., A
direct role for c/EBP and AP-I binding site in gene expression linked to adipocyte
differentiation, Mol. Cell Bioi., 9, 5331, 1989.
108. Christy, R. J., Yang, V. W., Ntambi, J. M., Geiman, D. E., Landschulz, W. H.,
Friedman, A. D., Nakabeppu, Y., Kelly, T. J., and Lane, M. D., Differentiation-
induced gene expression in 3T3-Ll preadipocytes: CCAAT/enhancer binding protein
interacts with and activates the promoters of adipocyte-specific genes, Genes Dev., 3,
1323, 1989.
454 Nutrition and Gene Expression
109. Friedman, A. D., Landschulz, W. H., and McKnight, S. L., C/EBP activates the
promoter of the serum albumin gene in cultured hepatoma cells, Genes Dev., 3, 1314,
1989.
110. McKnight, S. L., Lane, M. D., and Gluecksohn-Waelsch, S., Is CCAAT/enhancer
binding protein a central regulatory of energy metabolism?, Genes Dev., 3, 2021, 1989.
Ill. Tang, G. and Russel, R. M., 13-cis-retinoic acid is an endogenous compound in human
serum, J. Lipids Res., 31, 175, 1990.
112. Cullum, M. E. and Zile, M. H., Metabolism of all-trans-retinoic acid and all-trans-
retinyl acetate. Demonstration of common physiological metabolites in rat small intestinal
muscosa and circulation, J. Bioi. Chern., 260, 10590, 1985.
Chapter 20
J. W. Suttie
TABLE OF CONTENTS
0-8493-6961-4/93/$0.00 +$.50
© 1993 by CRC Press. Inc. 455
456 Nutrition and Gene Expression
I. INTRODUCTION
Vitamin K was not discovered until the early 1930s when Dam 1 noted a
hemorrhagic syndrome in chicks fed a lipid-free diet. This response could be
cured by the addition of alfalfa meal to the diet or by the administration of
a lipid extract of green plants. By 1939, a series of investigations led by Dam
and his collaborators in Denmark, Almquist at Berkeley, and Doisy at St.
Louis University had established that the form of the vitamin which is found
in alfalfa, and now called vitamin K 1 or phylloquinone, was 2-methyl-3-
phytyl-1 ,4-naphthoquinone. A large number of bacteria, including numerous
faculative and obligatory anaerobes of the human lower bowel, synthesize a
series of vitamers with unsaturated multiprenyl side chains at the 3-position.
These were characterized soon after phylloquinone and were originally called
vitamin K 2 , but are now more correctly designated as menaquinones (Figure
1). This early history of the discovery and characterization of vitamin K has
been adequately reviewed. zA
The hemorrhagic condition that resulted from the dietary lack of vitamin
K was shown by Dam et al. 5 to be associated with a decrease in plasma
prothrombin activity, and, for some time, was thought to be due solely to a
lowered concentration of prothrombin (factor II). Later, as they were dis-
covered, it was shown that the synthesis of clotting factors VII, IX, and X,
which are involved in the activation of prothrombin to thrombin, was also
depressed in the deficient state. 4 These four plasma proteins were collectively
called the ''vitamin K-dependent clotting factors'', and for years were thought
to be the only proteins whose synthesis required the vitamin. It was not until
the 1970s, after the biochemical role of the vitamin had been established,
that two proteins now known to play an anticoagulant, rather than a procoa-
gulant, role in hemostasis, protein C6 , and protein S7 were discovered. The
general role of the vitamin K -dependent procoagulants and anticoagulants in
normal hemostasis is shown in Figure 2. Another vitamin K-dependent protein
(protein Z) of unknown function is also found in plasma. A limited number
of extrahepatic, vitamin K-dependent proteins, including osteocalcin or Bone
Gla Protein (BGP) and Matrix Gla Protein (MGP), have been identified. 8
However, efforts to understand the enzymology of the vitamin K-dependent
modification of proteins have concentrated on the activity expressed in hepatic
tissue.
Although vitamin K was known by the late 1930s to regulate the activity
of a few specific proteins, the lack of a general understanding of the mech-
anism of protein biosynthesis prevented serious experimental approaches to
the cellular and molecular mechanisms involved until the mid-1960s. By the
early 1940s, the 4-hydroxy-coumarins were identified as indirect anticoag-
ulants that functioned by antagonizing the action of vitamin K. 9 Studies
Suttie 457
c<r
0 0
0 0
Menadione Phylloquinone
0
Menaqui none-7
FIGURE 1. Biologically active forms of vitamin K. Phylloquinone, produced by plants, and
menaquinone-7 (MK-7), an example of the long chain menaquinone produced by bacteria, are
natural forms of the vitamin. Menadione is a synthetic compound which has biological activity
after it is alkylated to MK-4 in the liver.
! !
lla
x.
~~-TM n.
~-~~~·~\1'111"
_ ...//····---- .~
~··· inact.
VIlla
0
''
j
Thrombin
Fibrinogen - - - - - • Fibrin
FIGURE 2. The role of vitamin K-dependent proteins in blood coagulation. The classical
vitamin K-dependent clotting factors, II (prothrombin), VII, IX, and X. have procoagulant
hemostatic roles and circulate as inactive zymogens which are converted to active serine proteases
by specific peptide cleavage (e.g .• X to Xa). Protein C and proteinS are involved in the process
by which the rapid generation of thrombin from prothrombin is brought under control by inac-
tivation of accessory factors Va and VIlla of the hemostatic system. The other clotting factors
shown do not require vitamin K for their biosynthesis.
FIGURE 3. "(-Carboxyglutamic acid. The vitamin K-dependent proteins are typical glycopro-
teins that are unique in that they contain a number of 'Y-carboxyglutamyl (Gla) residues in a very
homologous (see Table I) amino terminal domain.
I Vit KH 2 Vit KO I
CH2
I
CH 2
I
)>-/'·
/
CHz
I
H-C-COOH
I
COOH 02 C0 2 COOH
FIGURE 4. The liver microsomal vitamin K-dependent carboxylase. The enzyme utilizes the
hydronaphthoquinone form of the vitamin (Vit KH 2 ) to drive the carboxylation of peptidyl bound
glutamyl residues to "(-carboxyglutamyl residues. The coproduct of the reaction is vitamin K
2,3-epoxide (Vit KO).
zyme, which have been reviewed in detail, 17- 21 demonstrated that the reaction
required 0 2 , C0 2 , and the reduced (hydronaphthoquinone) form of vitamin
K. These early studies established that the only source of energy needed to
drive this unique carboxylation reaction comes from the reoxidation of the
reduced vitamin.
Most studies of the enzyme hve utilized crude microsomes solubilized in
various detergents and short peptides containing Glu-Glu sequences as sub-
strates. The enzyme is usually studied at 25°C or lower, and most studies
have been carried out in very crude preparations. Reported measurements of
kinetic contents have been variable but have been in the range of 75 fLM for
reduced vitamin K, 2 mM for COzfHC03 - , and 100 fLM for 0 2.22 Most studies
have utilized short peptides such as Phe-Leu-Glu-Glu-Leu or Boc-Glu-Glu-
Leu-OMe as Glu site substrates. These have a Km of a few mM in contrast
to the low Km values measured for the natural protein substrates of the en-
zyme.22
There have been few attempts to understand the mechanism of action of
the enzyme by the use of steady-state kinetic measurements. It is difficult to
saturate the enzyme with the nonvariable substrates, but Uotila 23 has studied
the calf liver enzyme at a number of substrate concentrations. He concluded
that the carboxylase has a sequential reaction mechanism which includes a
quintemary complex of the enzyme with all of the four substrates. The data
are consistent with an ordered steady-state addition of the substrates to the
enzyme and suggest that C0 2 is either the first or last substrate to bind.
OH H+
c¢: R ~(-)
Glu Glu
C02
\.., Gla
~lOH
~
0
[ KH-OOH] (Qo R
H2 0 0
"r
HC-COOH
I
COOH
SH SH
t...,_,._,J
appears to be 41 the enzyme that is the site of the physiological action of the
4-hydroxycoumarins as anticoagulants. In crude in vitro systems, dithiothrei-
tol will also reduce vitamin K to vitamin KH 2 in a 4-hydroxycoumarin-
sensitive reaction. It is likely that this reduction is catalyzed by the same
enzyme that reduces vitamin KO to vitamin K, 42 but final proof of this
relationship will depend on purification of the two activities. Recent evidence
suggests that the physiological reductant for the enzyme(s) that catalyzes the
reduction of vitamin K to vitamin KH 2 may be thioredoxin. 43 •44
+I +10
F-11 Human Ala Asn Thr Phe Leu Gla Gla Val Arg Lys Gly Asn Leu Gla Arg
Bovine Ala Asn Lys Gly Phe Leu Gla Gla Val Arg Lys Gly Asn Leu Gla Arg
Chicken Ala Asn Lys Gly Phe Leu Gla Gla Met lie Lys Gly Asn Leu Gla Arg
Rat Ala Asn Ser Gly Phe Leu Gla Gla Leu Arg Lys Gly Asn Leu Gla A
F-X Human Ala Asn Ser Phe Leu Gla Gla Met Lys Lys Gly His Leu Gla Arg
F-IX Human Tyr Asn Ser Gly Lys Leu Gla Gla Phe Val Gin Gly Asn Leu Gla Arg
F-VII Human Ala Asn Ala Phe Leu Gla Gla Leu Arg Pro Gly Ser Leu Gla Arg
P-C Human Ala Asn Ser Phe Leu Gla Gla Leu Arg His Ser Ser Leu Gla Arg
P-S Human Ala Asn Ser Leu Leu Gla Gla Thr Lys Gin Gly Asn Leu Gla Arg
+20 +30
F-11 Human Gla Cys Val Gla Gla Thr Cy> Ser Tyr Gla Gla Ala Phe Gla Ala Leu
Bovine Gla Cys Leu Gla Gla Pro Cys Ser Arg Gla Gla Ala Phe Gla Ala Leu
Chicken Gla Cys Leu Gla Gla Thr Cys Asn Tyr Gla Gla Ala Phe Gla Ala Leu
Rat Gla Cys Val Gla Gla Gin Cys Ser Tyr Gla Gla Ala Phe Gla Ala Leu :;;..:
;;:::
F-X Human Gla Cys Met Gla Gla Thr Cys Ser Tyr Gla Gla Ala Arg Gla Val Phe
F-IX Human Gla Cys Me Gla Gla Lys Cys Ser Phe Gla Gla Ala Arg Gla Val Phe "~:
F-VII Human Gla Cys Lys Gla Gla Gin Cys Ser Phe Gla Gla Ala Arg Gla lie Phe ;::
$:l
P-C Human Gla Cys lie Gla Gla lie Cys Asp Phe Gla Gla Ala Lys Gla lie Phe ;::
P-S Human Gla Cys lie Gla Gla Leu Cys Asn Lys Gla Gla Ala Arg Gla Val Phe i:l...
+40 C'l
;::
F-11 Human Gla Ser Ser Thr Ala Asp Val Phe Trp Ala
I Lys Tyr "'
Bovine Gla Ser Leu Ser Ala hr Asp Ala Phe Trp Ala Lys Tyr "'~
Chicken Gla Ser Thr Val Asp hr Asp Ala Phe Trp ':::i
Rat Gla Ser Pro Gin Asp hr Asp Val Phe Lys Tyr ~
Trp Ala
I
F-X Human Gla Asp Ser Asp Lys hr Asn Gla Phe Trp Asn Lys Tyr """"c:;·
F-IX Human Gla Asn Thr Gla Arg hr Thr Gla Phe Trp Lys Gin Tyr ;::
~1
F-VII Human Lys Asp Ala Gla Lys Leu Ser Tyr ~
Arg 'Thr Phe Trp Ile
I s:::
P-C Human Gln Asn Val Asp Asp Thr Leu Ala Phe Trp Ser Lys His ::::
Gla Asn Asp Pro Asp Tyr Phe Tyr Pro Lys Tyr
;;;·
P-S Human Gla Thr
Note: References to the primary literature can be obtained in reviews46 A 9 with the exception of the rat sequence 111
which is identical to that of the mouse 111 Although the sequence of the Gla region of most of these proteins
was determined by peptide sequences, complete structures for most were obtained only when eDNA structures
were available.
.,..
0\
-...l
468 Nutrition and Gene Expression
prothromb'n l K K
FIGURE 7. Structural domains of the vitamin K-dependent plasma clotting factors. The cleav-
age site(s) that give rise to the active proteases are indicated by arrows. Abbreviations used:
y, y-carboxyglutamic acid-containing region; K, kringle; E, epidermal growth factor-like region.
The serine protease catalytic region is shown as a solid bar. The structures are drawn to scale,
and the 100-residue length is indicated. Adapted from Fung and MacGillivray.••
Note: The amino terminal residue of the mature form of the plasma protein is designated as
residue + I. The site of signal peptide cleavage for factor IX is between residues - 19
and - IS"I.I" and between -24 and - 25 for protein C. 77 The position of cleavage
has not been determined with certainty for all of the other factors.
....~
472 Nutrition and Gene Expression
interaction of enzyme and substrate at the Glu site. The ability of the free
propeptide to stimulate the carboxylase is not limited to weak binding high
Km substrates. Des-'Y-carboxy Bone Gla Protein (dBGP) is not a particularly
good carboxylase substrate, but in the presence of free propeptide it is effi-
ciently carboxylated91 with a Km of only 11 !J.M. These data indicating a role
of the propeptide in modulating the Glu binding site of the enzyme do not
suggest that the presumed docking function of the propeptide is not important.
A second protein originally found in bone, MGP, does not have an amino
terminal propeptide cleaved during protein processing. Rather, it has an in-
ternal sequence in the mature protein which is homologous to this region. 87
The Km of dMGP as a substrate is only 0.2 !J.M, and its carboxylation is not
stimulated, but is effectively inhibited by free propeptide, 91 presumably by
displacing it from the enzyme. These data make it clear that the propeptide
region of the few proteins that are substrates for the carboxylase serves a dual
role, it anchors or docks the substrate with the enzyme, and also alters the
affinity of the enzyme for the Glu residue which will be carboxylated.
Why both a docking and regulatory role for the propeptide should be
required for normal carboxylation is not readily apparent. The manner by
which the enzyme carboxylates its multisite substrates is not known, and it
is possible that some Glu (potential Gla) sites may have a high affinity for
the enzyme, or that it is an ordered series of events. The location of the Gla
residues in partially carboxylated forms of prothrombin92 suggests that the
most amino terminal Gla site is most readily carboxylated but that there may
also be high affinity sites elsewhere in the Glu region. The published studies
of the in vitro carboxylation of numerous peptides with multiple Glu sites
have not addressed the question of which of the possible sites are carboxylated.
This question is not easily addressed in a substrate with ten potential Gla
sites, but is more easily approached by studies of the carboxylation of dBGP
which has only three carboxylated Glu residues. Preliminary data from this
system93 suggests that the most carboxy terminal Glu is preferentially car-
boxylated, but the influence of the propeptide region on the pattern of car-
boxylation has not yet been determined.
Both rat (H-35) and human (HepG2) hepatoma cell lines 94. 96 have been
shown to secrete prothrombin and other vitamin K-dependent proteins. 95 •97
The general observation has been that warfarin treatment increases the intra-
cellular pool of prothrombin and decreases secretion into the culture medium,
while vitamin K administration increases secretion. It is not clear from the
available data if these responses are related in any way to changes in the rate
of expression of the prothrombin gene, or if they merely represent a response
to the altered ability of the cells to effectively carry out the 'Y-glutamyl
Suttie 475
carboxylation step. The H-35 rat hepatoma synthesis systems have also been
used 98 to demonstrate an increase of prothrombin synthesis upon the addition
of the amino terminal, fragment-! portion (residues 1 through 156) of pro-
thrombin to the media. Indirect evidence suggested that this response was
related to an effect on prothrombin gene expression, but no direct evidence
was presented, and the observation has not been repeated.
Plasma concentrations of vitamin K-dependent clotting factors are nor-
mally maintained in a narrow range, and there is some evidence for a humoral
factor that is involved in this control. Karpatkin and his co-workers 90 · 100 have
demonstrated that the intravenous injection of a small amount of plasma
obtained from an anticoagulant-treated rabbit with a low level of vitamin K-
dependent clotting factors into a normal rabbit will result in an increase in
the activities of these factors in the recipient rabbit. The humoral factor
responsible for this response has been called a coagulopoietin(s). This response
has also been observed in the rat, 101 and there are indications 101 . 103 that this
treatment increases biological activity of prothrombin more than prothrombin
antigen. It has been demonstrated 104 that this factor appears early during the
development of a deficiency state and that the activity is variable between
different animals. The chemical nature of the proposed coagulopoietin is not
known, and the current data do not clearly differentiate an effect on expression
of the vitamin K-dependent proteins, or of components of the carboxylase
system.
The concentration of undercarboxylated prothrombin in normal human
plasma is very low and increases substantially with oral anticoagulation or
vitamin K deficiency, and slightly in many patients with liver disease. 105 It
has been shown 106 · 107 that this protein is increased more dramatically in cases
of primary hepatocellular carcinoma than in other liver disorders, and its
presence can be used to screen for this disease. This increase is independent
of vitamin K status, and the molecular basis of the observation has been
studied in a rat model. 108 A series of Morris hepatoma tumors were trans-
planted into recipient Buffalo strain rats. Transplantation of some but not all
tumor lines resulted in increased levels of circulating undercarboxylated pro-
thrombin, and the tumor mass had low levels of carboxylase. The tumor mass
of those lines not secreting this marker had much higher levels of the car-
boxylase. The data suggest that the secretion of abnormal prothrombin by
hepatocellular tumors is the result of normal expression of the prothrombin
gene and a failure of the tumor to fully express the carboxylase gene.
These examples indicate that neither the role of vitamin K, if any, in
regulating gene expression, nor the role of other factors, which influence
gene expression on vitamin K metabolism, is yet understood. Progress in this
area should, however, be rapid. The vitamin K-dependent proteins themselves
have been cloned, and mRNA probes have been successfully used to follow
the regulation of prothrombin synthesis during fetal and neonatal develop-
ment. 109 • 110 The currently confused status of the possible regulation of clotting
476 Nutrition and Gene Expression
REFERENCES
1. Darn, H., Hemorrhages in chicks reared on artificial diets: a new deficiency disease,
Nature (London), 133, 909, 1934.
2. Suttie, J, W., Vitamin K, inHandbookofVitamins, 2nded., Machlin, L. J., Ed., Marcel
Dekker, New York, 1991.
3. Bentley, R. and Meganathan, R., Biosynthesis of vitamin K (menaquinone) in bacteria,
Microbial. Rev., 46, 241, 1982.
4. Suttie, J, W., Vitamin K, in The Fat-Soluble Vitamins, Diplock, A. T., Ed., William
Heinemann, London, 1985, 225.
5. Darn, H., Schonheyder, F., and Tage-Hansen, E., Studies on the mode of action of
vitamin K, Biochem. J., 30, 1075, 1936.
6. Stenflo, J,, A new vitamin K-dependent protein. Purification from bovine plasma and
preliminary characterization, J. Bioi. Chern., 251, 355, 1976.
7. Di Scipio, R. G., Herrnodson, M.A., Yates, S. G., and Davie, E. W., A comparison
of human prothrombin, factor IX (Christmas factor), factor X (Stuart factor), and protein
S, Biochemistry, 16, 698, 1977.
8. Price, P. A., Role of vitamin K-dependent proteins in bone metabolism, Annu. Rev.
Nutr., 8, 565, 1988.
9. Link, K. P., The discovery of dicumarol and its sequels, Circulation, 19, 97, 1959.
10. Olson, R. E., Vitamin K induced prothrombin formation: antagonism by actinomycin
D, Science, 145, 926, 1964.
11. Suttie, J. W., Metabolism and properties of a liver precursor to prothrombin, Vitam.
Horm., 32, 463, 1974.
12. Stenflo, J., Fernlund, P., Egan, W., and Roepstorff, P., Vitamin K dependent mod-
ifications of glutamic acid residues in prothrombin, Proc. Nat/. Acad. Sci. U.S.A., 71,
2730, 1974.
13. Nelsestuen, G. L., Zytkovicz, T. H., and Howard, J, B., The mode of action of
vitamin K. Identification of -y-carboxyglutamic acid as a component of prothrombin. J.
Bioi. Chern., 249, 6347, 1974.
14. Magnusson, S., Sottrup-Jensen, L., Petersen, T. E., Morris, H. R., and Dell, A.,
Primary structure of the vitamin K-dependent part of prothrombin, FEBS Lett., 44, 189,
1974.
15. Stenflo, J, and Suttie, J, W., Vitamin K-dependent formation of -y-carboxyglutamic
acid, Annu. Rev. Biochem., 46, 157, 1977.
16. Esrnon, C. T., Sadowski, J, A., and Suttie, J. W., A new carboxylation reaction. The
vitamin K-dependent incorporation of H14C03 into prothrombin, J. Bioi. Chern., 250,
4744, 1975.
17. Suttie, J, W., Mechanism of action of vitamin K: synthesis of Y-carboxyglutamic acid,
CRC Crit. Rev. Biochem., 8, 191, 1980.
18. Johnson, B. C., Post-translational carboxylation of preprothrombin, Mol. Cell Biochem.,
38, 77, 1981.
Suttie 477
19. Vermeer, C. and de Boer-van den Berg, M.A. G., Vitamin K-dependent carboxylase,
Haematologia, 18, 71, 1985.
20. Olson, R. E., The function and metabolism of vitamin K, Annu. Rev. Nutr., 4, 281,
1984.
21. Friedman, P. A. and Przysiecki, C. T., Vitamin K-dependent carboxylation, Int. J.
Biochem., 19, I, 1987.
22. Suttie, J, W., Vitamin K-dependent carboxylation of glutamyl residues in proteins,
BioFactors, 1, 55, 1988.
23. Uotila, L., Vitamin K-dependent carboxylase from calf liver: studies on the steady-state
kinetic mechanism, Arch. Biochem. Biophys., 264, 135, 1988.
24. Friedman, P. A., Shia, M.A., Gallop, P.M., and Griep, A. E., Vitamin K-dependent
-y-carbon-hydrogen bond cleavage and the non-mandatory concurrent carboxylation of
peptide bound glutamic acid residues, Proc. Nat/. Acad. Sci. U.S.A., 76, 3126, 1979.
25. Larson, A. E., Friedman, P. A., and Suttie, J. W., Vitamin K-dependent carboxylase:
stoichiometry of carboxylation and vitamin K 2,3-epoxide formation, J. Bioi. Chern.,
256, 11032, 1981.
26. McTigue, J. J. and Suttie, J. W., Vitamin K-dependent carboxylase: demonstration of
a vitamin K- and 0 2-dependent exchange of 'H from 3 H2 0 into glutamic acid residues,
J. Bioi. Chern., 258, 12129, 1983.
27. Anton, D. L. and Friedman, P. A., Fate of the activated -y-carbon-hydrogen bond in
the uncoupled vitamin K-dependent -y-glutamyl carboxylation reaction, J. Bioi. Chern.,
258, 14084, 1983.
28. Wood, G. M. and Suttie, J, W., Vitamin K-dependent carboxylase. Stoichiometry of
vitamin K epoxide formation, -y-carboxyglutamyl formation, and -y-glutamyl-3H cleavage,
J. Bioi. Chern., 263, 3234, 1988.
29. Dowd, P., Ham, S. W., and Geib, S. J,, Mechanism of action of vitamin K, J. Am.
Chern. Soc., 113, 7734, 1991.
30. Harbeck, M. C., Cheung, A. Y., and Suttie, J, W., Vitamin K-dependent carboxylase:
partial purification of the enzyme by antibody affinity techniques, Thrombosis Res.. 56,
317, 1989.
31. Hubbard, B. R., Ulrich, M. M. W., Jacobs, M., Vermeer, C., Walsh, C., Furie,
B., and Furie, B. C., Vitamin K-dependent carboxylase: affinity purification from bovine
liver by using a synthetic propeptide containing the -y-carboxylation recognition site,
Proc. Nat/. Acad. Sci. U.S.A., 86, 6893, 1989.
32. Wu, S-M., Morris, D. P., and Stafford, D. W., Identification and purification to near
homogeneity of the vitamin K-dependent carboxylase, Proc. Nat/. Acad. Sci. U.S.A.,
88, 2236, 1991.
33. Wu, S-M., Cheung, W-F., Frazier, D., and Stafford, D., Cloning and expression of
the eDNA for human -y-glutamyl carboxylase, Science, 254, 1634, 1991.
34. Berkner, K. L., Harbeck, M., Lingenfelter, S., Bailey, C., Sanders-Hinck, C. M.,
and Suttie, J, W., Purification and identification of the bovine liver-y-carboxylase, Proc.
Nat/. Acad. Sci. U.S.A., 89, 6242, 1992.
35. Kvalvaag, A. H., Tollersrud, 0. K., and Helgeland, L., A study on the intracellular
transport of prothrombin, albumin and transferrin in rat, Biochim. Biophys. Acta, 937,
319, 1988.
36. Wallin, R. and Martin, L. F., Early processing of prothrombin and factor X by the
vitamin K-dependent carboxylase, J. Bioi. Chern., 263, 9994, 1988.
37. Shah, D. V., Swanson, J, C., and Suttie, J, W., Abnormal prothrombin in the vitamin
K-deficient rat, Thrombosis Res., 35, 451, 1984.
38. Wallin, R., Gebhardt, 0., and Prydz, H., NAD(P)H dehydrogenase and its role in
the vitamin K (2-methyl-3-phytyl-1 ,4-naphthoquinone)-dependent carboxylation reaction,
Biochem. J., 169, 95, 1978.
478 Nutrition and Gene Expression
57. Griffin, J, H., Evatt, B., Zimmerman, T. S., Kleiss, A. J., and Wideman, C.,
Deficiency of protein C in congenital thrombotic disease, J. Clin. Invest., 68, 1370,
1981.
58. Clouse, L. H. and Comp, P. C., The regulation of hemostasis: the protein C system,
New Engl. J. Med., 314, 1298, 1986.
59. Esmon, C. T., The regulation of natural anticoagulant pathways, Science, 235, 1348,
1987.
60. Chung, K-S, Bezeaud, A., Goldsmith, J, C., McMillan, C. W., Menache, D., and
Roberts, H. R., Congenital deficiency of blood clotting factors II, VII, IX, and X,
Blood, 53, 776, 1979.
61. Pauli, R. M., Lian, J, B., Mosher, D. F., and Suttie, J, W., Association of congenital
deficiency of multiple vitamin K-dependent coagulation factors and the phenotype of the
warfarin embryopathy: clues to the mechanism of teratogenicity of coumarin derivatives,
Am. J. Hum. Genet., 41, 566, 1987.
62. Swanson, J, C. and Suttie, J, W., Prothrombin biosynthesis: characterization of pro-
cessing events in rat liver microsomes, Biochemistry, 24, 3890, 1985.
63. Soute, B. A. M., Vermeer, C., De Metz, M., Hemker, H. C., and Lijnen, H. R.,
In vitro prothrombin synthesis from a purified precursor protein. Ill. Preparation of an
acid-soluble substrate for vitamin K-dependent carboxylase by limited proteolysis of
bovine descarboxyprothrombin, Biochim. Biophys. Acta, 676, 101, 1981.
64. Shah, D. V., Swanson, J, C., and Suttie, J, W., Vitamin K-dependent carboxylase:
effect of detergent concentrations, vitamin K status, and added protein precursors on
activity, Arch. Biochem. Biophys., 222, 216, 1983.
65. Evans, M. R., Sung, M. W. P., and Esnouf, M. P., The early stages in the biosynthesis
of prothrombin, Biochem. Soc. Trans., 12, 1051, 1984.
66. Pan, L. C. and Price, P. A., The propeptide of rat bone -y-carboxyglutamic acid protein
shares homology with other vitamin K-dependent protein precursors, Proc. Nat/. Acad.
Sci. U.S.A., 82, 6109, 1985.
67. de Ia Salle, H., Altenburger, W., Elkaim, R., Dott, K., Dieterle, A., Drillien, R.,
Cazenave, J-P., Tolstoshev, P., and Lecocq, J-P., Active -y-carboxylated human factor
IX expressed using recombinant DNA techniques, Nature (London), 316, 268, 1985.
68. Anson, D. S., Austen, D. E. G., and Brownlee, G. G., Expression of active human
clotting factor IX from recombinant DNA clones in mammalian cells, Nature (London),
315, 683, 1985.
69. Busby, S., Kumar, A., Joseph, M., Halfpap, L., Insley, M., Berkner, K., Kurachi,
K., and Woodbury, R., Expression of active human factor IX in transfected cells, Nature
(London), 316, 271, 1985.
70. Kaufman, R. J., Wasley, L. C., Forie, B. C., Forie, B., and Shoemaker, C. B.,
Expression, purification, and characterization of recombinant -y-carboxylated factor IX
synthesized in Chinese hamster ovary cells, J. Bioi. Chern., 261, 9622, 1986.
71. Thim, L., Bjoern, S., Christensen, M., Nicolaisen, E. M., Lund-Hansen, T., Pedersen,
A. W., and Redner, U., Amino acid sequence and posttranslational modifications of
human factor VIla from plasma and transfected baby hamster kidney cells, Biochemistry,
27, 7785, 1988.
72. Yan, S. C. B., Grinnell, B. W., and Wold, F., Post-translational modifications of
proteins: some problems left to solve, Trends Biochem. Sci., 14, 264, 1989.
73. Brinkhous, K. M., Hedner, U., Garris, J. B., Diness, V., and Read, M. S., Effect
of recombinant factor VIla on the hemostatic defect in dogs with hemophilia A, hemophilia
B, and von Willebrand Disease, Proc. Nat/. Acad. Sci. U.S.A., 86, 1382, 1989.
74. Diness, V., Lund-Hansen, T., and Hedner, U., Effect of recombinant human FVIIA
on warfarin-induced bleeding in rats, Thrombosis Res., 59, 921, 1990.
75. Redner, U., Glazer, S., Pingel, K., Alberts, K. A., Blomback, M., Schulman, S.
and Johnsson, H., Successful use of recombinant factor VIla in patient with severe
haemophilia A during synovectomy, Lancet, 2, 1193, 1988.
480 Nutrition and Gene Expression
76. Macik, B. G., Hohneker, J., Roberts, H. R., and Griffin, A.M., Use of recombinant
activated factor VII for treatment of a retropharyngeal hemorrhage in a hemophilic patient
with a high titer inhibitor, Am. J. Hematol., 32, 232, 1989.
77. Foster, D. C., Rudinski, M. S., Schach, B. G., Berkner, K. L., Kumar, A. A.,
Hagen, F. S., Sprecher, C., Insley, M., and Davie, E. W., Propeptide of human protein
Cis necessary for gamma-carboxylation, Biochemistry, 26, 7003, 1987.
78. Jorgensen, M. J,, Cantor, A. B., Furie, B. C., Brown, C. L., Shoemaker, C. B.,
and Furie, B., Recognition site directing vitamin K-dependent -y-carboxylation residues
on the propeptide of factor IX, Cell, 48, 185, 1987.
79. Rabiet, M-J., Jorgensen, M. J,, Furie, B., and Furie, B. C., Effect of propeptide
mutations on post-translational processing of factor IX. Evidence that [3-hydroxylation
and-y-carboxylation are independent events, J. Bioi. Chem., 262, 14895, 1987.
80. Handford, P. A., Winship, P. R., and Brownlee, G. G., Protein engineering of the
propeptide of human factor IX, Protein Eng., 4, 319, 1991.
81. Bentley, A. K., Rees, D. J, G., Rizza, C., and Brownlee, G. G., Defective propeptide
processing of blood clotting factor IX caused by mutation of arginine to glutamine at
position -4, Cell, 45, 343, 1986.
82. Huber, P., Schmitz, T., Griffin, J,, Jacobs, M., Walsh, C., Furie, B., and Furie,
B. C., Identification of amino acids in the -y-carboxylation recognition site on the pro-
peptide of prothrombin, J. Bioi. Chem., 265, 12467, 1990.
83. Furie, B. and Furie, B. C., Molecular basis of vitamin K-dependent -y-carboxylation,
Blood, 75, 1753, 1990.
84. Ulrich, M. M. W., Furie, B., Jacobs, M. R., Vermeer, C., and Furie, B. C., Vitamin
K-dependent carboxylation. A synthetic peptide based upon the -y-carboxylation recog-
nition site sequence of the prothrombin propeptide is an active substrate for the carboxylase
in vitro, J. Bioi. Chem., 263, 9697, 1988.
85. Hubbard, B. R., Jacobs, M., Ulrich, M. M. W., Walsh, C., Furie, B., and Furie,
B. C., Vitamin K-dependent carboxylation. In vitro modification of synthetic peptides
containing the-y-carboxylation recognition site, J. Bioi. Chem., 264, 14145, 1989.
86. Wu, S-M., Soule, B. A. M., Vermeer, C., and Stafford, D. W., In vitro -y-carbox-
ylation of a 59-residue recombinant peptide including the propeptide and the -y-carbox-
yglutamic acid domain of coagulation factor IX. Effect of mutations near the propeptide
cleavage site, J. Bioi. Chem., 265, 13124, 1990.
87. Price, P. A., Fraser, J, D., and Metz-Virca, G., Molecular cloning of matrix Gla
protein: implications for substrate recognition by the vitamin K-dependent -y-carboxylase,
Proc. Nat!. Acad. Sci. U.S.A., 84, 8335, 1987.
88. Knobloch, J. E. and Suttie, J. W., Vitamin K-dependent carboxylase. Control of enzyme
activity by the "propeptide" region of factor X, J. Bioi. Chem., 262, 15334, 1987.
89. Cheung, A., Engelke, J, A., Sanders, C., and Suttie, J, W., Vitamin K-dependent
carboxylase: influence of the "propeptide" region on enzyme activity, Arch. Biochem.
Biophys., 274, 574, 1989.
90. Cheung, A., Suttie, J, W., and Bernatowicz, M., Vitamin K-dependent carboxylase:
structural requirements for propeptide activation, Biochim. Biophys. Acta, 1039, 90, 1990.
91. Engelke, J, A., Hale, J, E., Suttie, J, W., and Price, P. A., Vitamin K-dependent
carboxylase: utilization of decarboxylated bone Gla protein and matrix Gla protein as
substrates, Biochim. Biophys. Acta, 1078, 31, 1991.
92. Liska, D. J, and Suttie, J, W., Location of -y-carboxyglutamyl residues in partially
carboxylated prothrombin preparations, Biochemistry, 27, 8636, 1988.
93. Benton, M. E., Price, P. A., and Suttie, J, W., The processivity of the vitamin K-
dependent carboxylase, FASEB J., abstr., in press.
94. Munns, T. W., Johnston, M. F. M., Liszewski, M. K., and Olson, R. E., Vitamin
K-dependent synthesis and modification of precursor prothrombin in cultured H-35 hep-
atoma cells, Proc. Nat!. Acad. Sci. U.S.A., 73, 2803, 1976.
Suttie 481
95. Fair, D. S. and Bahnak, B. R., Human hepatoma cells secrete single chain factor X,
prothrombin, and antithrombin III, Blood, 64, 194, 1984.
96. Karpatkin, S., Finlay, T. H., Ballesteros, A. L., and Karpatkin, M., Effect of warfarin
on prothrombin synthesis and secretion in human HEP G2 cells, Blood, 70, 773, 1987.
97. Fair, D. S. and Marlar, R. A., Biosynthesis and secretion of factor VII, protein C,
protein S, and the protein C inhibitor from a human hepatoma cell line, Blood, 67, 64,
1986.
98. Graves, C. B., Munns, T. W., Carlisle, T. L., Grant, G. A., and Strauss, A. W.,
Induction of prothrombin synthesis by prothrombin fragments, Proc. Nat/. Acad. Sci.
U.S.A., 78, 4772, 1981.
99. Friedman, E. W., Karpatkin, M., and Karpatkin, S., Evidence suggesting the reg-
ulation of coagulation factor levels in rabbits by a transferable plasma agent, Blood, 48,
949, 1976.
I00. Karpatkin, M. and Karpatkin, S., Humoral factor that specifically regulates prothrom-
bin (factor II) levels (coagulopoietin-11), Proc. Nat/. Acad. Sci. U.S.A., 76, 491, 1979.
101. Shah, D. V., Nyari, L. J,, Swanson, J, C., and Suttie, J. W., Effect of a humoral
factor on plasma prothrombin and vitamin K-dependent liver carboxylase levels in the
rat, Thrombosis Res., 19, Ill, 1980.
102. Karpatkin, S., Chang, R-J., Pierce, W., and Karpatkin, M., Effect of coumadin-
induced coagulopoietin plasma on vitamin K-dependent carboxylation of liver micro-
somes, Br. J. Haematol., 55, 673, 1983.
103. Owens, M. R. and Cimino, C. D., A plasma factor enhances activity of vitamin K-
dependent coagulation proteins, Thromb. Haemostas., 50, 749, 1983.
I 04. Shah, D. V. and Suttie, J. W., Coagulopoietin activity in rat plasma: influence of degree
of hypoprothrombinemia and reproducibility of the response, Thromb. Res., 39, 43,
1985.
105. Blanchard, R. A., Forie, B. C., Jorgensen, M., Kruger, S. F., and Forie, B.,
Acquired vitamin K-dependent carboxylation deficiency in liver disease. New Engl. J.
Med., 305, 242, 1981.
106. Liebman, H. A., Forie, B. C., Tong, M. J., Blanchard, R. A., Lo, K-J., Lee, S-D.,
Coleman, M. S., and Forie, B., Des--y-carboxy (abnormal) prothrombin as a serum
marker of primary hepatocellular carcinoma. New Engl. J. Med., 310, 1427, 1984.
107. Naraki, T. and Watanabe, K., Quantitative measurement of PIVKA-II by the enzyme
immunoassay diagnostic kit Eitest Mono P-II, in Vitamin K-Dependent Proteins and Their
Metabolic Roles, Elsevier, New York, 1990, 83.
108. Shah, D. V., Engelke, J, A., and Suttie, J. W., Abnormal prothrombin in the plasma
of rats carrying hepatic tumors, Blood, 69. 850, 1987.
109. Kisker, C. T., Perlman, S., Bohlken, D., and Wicklund, B., Measurement of pro-
thrombin mRNA during gestation and early neonatal development. J. Lab. Clin. Med.,
112, 407, 1988.
110. Jamison, C. S. and Degen, S. J. F., Prenatal and postnatal expression of mRNA coding
for rat prothrombin, Biochim. Biophys. Acta, !088, 208, 1991.
Ill. Dihanich, M. and Monard, D., eDNA sequence of rat prothrombin, Nucleic Acids
Res., 18, 4251, 1990.
112. Degen, S. J, F., Shaefer, L. A., Jamison, C. S., Grant, S. G., FitzGibbon, J. J.,
Pai, J-A., Chapman, V. M., and Elliott, R. W., Characterization of the eDNA coding
for mouse prothrombin and localization of the gene on mouse chromosome 2. DNA Cell
Bioi., 9, 487, 1990.
113. Diuguid, D. L., Rabiet, M. J,, Forie, B. C., Liebman, H. A., and Forie, B., Molecular
basis of hemophilia B: a defective enzyme due to an unprocessed propeptide is caused
by a point mutation in the factor IX precursor, Proc. Nat/. Acad. Sci. U.S.A., 83, 5803,
1986.
Chapter 21
Richard I. Schwarz
TABLE OF CONTENTS
0-8493-6961-41931$0.00 + $.50
© 1993 by CRC Press, Inc. 483
484 Nutrition and Gene Expression
I. INTRODUCTION
But before focusing in more detail on the collagen pathway, one needs
a better overview of how ascorbate itself is regulated and how other functions
are affected by this molecule. While it is generally realized that humans,
other primates, and guinea pigs are incapable of making ascorbate because
of a mutation in a required enzyme (L-gulonolactone oxidase), 18 it is less well
known that most cells, even in a competent organism, lack the capacity to
make ascorbate as well. Instead, the vitamin is made in either the liver or
the kidney and transported to other tissues via the blood. 22 One consequence
is that the ascorbate concentration in each tissue varies depending on the
ability to actively transport ascorbate. In this regard ascorbate acts more like
a hormone than a vitamin. So it is not difficult to see why in a competent
organism, where most cells do not produce ascorbate, that a mutation that
deletes the synthetic capacity would have little effect, as long as the diet had
sufficient vitamin C. The reason behind this complex regulation is not known.
However, it may relate to the postulated role of ascorbate as a signaling
molecule between cell types. 28 ·36 ·8o
In addition to its hormone and signaling potential, one finds that ascorbate
has many critical biological activities in different tissues in the body. As-
corbate acts as a hydrophilic protector of biological molecules from oxidative
damage. Lipids in the blood are shielded from oxidation by the presence of
ascorbate. 21 •42 It also acts as a biological reducing agent with the ability to
reduce ferric to ferrous and cupric to cuprous. Other reducing agents like
NADH and glutathione are ineffective. This accounts for its role as a cofactor
Schwarz 485
FIGURE I. Stained frozen sections o f chick leg tendons from 16-d e mbryos (top) and from
a 4-month old chicken (bottom: 2.5 x lower magnification). Two critical changes occur over
thi s relatively short-time span as the o rgani sm goes from a s tage of rapid growth to attainment
of full size. First, the tissue goes from being hypercellular to being hypocellular. Second, the
level of collagen synthesis drops (see Reference 71). The consequence is that the capacity of
the tissue to produce collagen is dramat ically reduced.
488 Nutrition and Gene Expression
In hindsight, one could predict that the strongest ascorbate effect would
be seen in embryonic cells in a cell culture environment permissive for main-
taining differentiated function. Indeed, one finds that primary avian tendon
(PAT) cells from 16-d chick embryos grown in low serum medium increase
their procollagen production from ~ 10 to ~50% of total protein synthesis
on addition of ascorbate. 68 Other cells producing type I procollagen, where
a relatively high procollagen rate has been maintained in culture, also show
a strong ascorbate effect on collagen production. What has made this so
interesting is that there is only evidence for the direct action of ascorbate on
the posttranslational modification step of prolyl hydroxylation. The impli-
cation is that the general increase in the collagen pathway occurs because
most steps are tightly coupled allowing feedback regulation. 66 The strong
evidence for this type of control is presented below.
For 25 years, researchers have been adding ascorbate to cell cultures and
analyzing its effects on the collagen pathway. While many changes have been
observed, the most consistent effect has been that ascorbate causes cells to
maximally hydroxylate their proline residues-between 40 and 50% of the
prolines are hydroxylated in the collagen molecule. It had been suggested in
the 1950s based on the stability of the collagen helix in different species 16
and then shown in the 1970s based on resistance to protease digestion4 •27 that
hydroxylation of proline residues stabilizes the triple helical conformation, 19
a hallmark of the collagen molecule. It is now known that the enzyme, prolyl
hydroxylase, prefers the nonhelical form and the reaction is therefore self-
limiting as increasing hydroxylation triggers the change to a helical form.
The question can then be asked whether this conformation change has any
additional effects on the collagen pathway. zAo
From early studies using electron microscopy to visualize the changes
occurring during wound healing in the scorbutic guinea pig, there were in-
dications that collagen secretion was retarded. 63 This was deduced from swell-
ing inside cells of what was presumed to be unsecreted collagen. Further
evidence came from experiments where the hydroxylation reaction was in-
hibited by using the ferrous ion chelator, a,a'-dipyridyl. Ferrous ion is a
necessary cofactor in the hydroxylation reaction; so, removing iron from the
reaction would be equivalent to depriving the cells of ascorbate (the role of
ascorbate in the hydroxylation reaction is to keep the iron in the reduced
state). In these experiments, a,a'-dipyridyl depressed collagen secretion. 40
In cell culture with 3T3 and chick embryo fibroblasts, 5 5 Peterkofsky could
show that the presence of ascorbate stimulated the secretion of procollagen.
In this case, newly synthesized procollagen was tracked by using a short pulse
of radioactive proline. The fraction of labeled procollagen in the medium was
found to be greater when cells were treated with ascorbate.
Schwarz 489
100
"' "'
""
50
I
""
\
\
#: \
""'
\
Q3
() 20 \
\
c
"'
\
Ol \
c
·c: \
\
"iti
10 \
E
~ \
c \
(j)
Ol
\
~ \
0() 5 \
0
'\.
ct '"~
0 2 3 4 5 6 7
Hours
FIGURE 2. Secretion of 3 H-labeled procollagen from PAT cells during the chase period when
ascorbate was added only at the time of the chase (the open and closed triangles are independent
determinations). The short dashed line shows the secretion profile of cells always in the presence
of ascorbate and the long dashed line shows the secretion profile of cells in the absence of
ascorbate (for more details of these controls see Reference 69). After -1 h, the cells are able
to hydroxylate the already synthesized procollagen and secrete it at the fast rate.
shift procollagen secretion to the slow route. 34 The implication of this result
is that a step in the fast route, possibly a receptor, requires the triple helical
conformation of the procollagen molecule in order to proceed.
These pulse-chase experiments raised the question of what the positive
effect of ascorbate addition would be on the process of procollagen secretion
and whether it would be directly opposite to those obtained using the inhibitor,
Schwarz 491
a,a' -dipyridyl. In the experiments above, freshly isolated chick embryo ten-
don cells were used, and one cannot ask the question directly. In the chick
embryo, ascorbate is made in the kidney and liver, and freshly isolated tendon
cells are, therefore, already maximally stimulated with ascorbate. One way
to circumvent this problem is to grow the cells in culture without ascorbate
for more than 36 h. 65 When pulse-chase experiments were performed using
tendon cells in culture, the data obtained by ascorbate addition to scorbutic
cultures was completely compatible with the inhibitor studies. 69 With ascor-
bate, the cells show a biphasic secretion profile with half-lives of ~20 min
and~ 120 min. In confluent cultures almost all the procollagen ( ~90%) leaves
the cell by the fast route. This may mean that in freshly isolated cells, assayed
in suspension culture, that the round shape caused by being unattached to a
substratum may partially inhibit the fast route. In any case, scorbutic tendon
cells secrete all of their procollagen by the slow route. Because ascorbate is
actively and rapidly transported into the cell, one can gauge how quickly
secretion rates can change by addition of ascorbate only at the time of the
chase (Figure 2). In an hour or less the cells stopped secreting at the slow
rate and started secreting the already synthesized procollagen molecules at
the high rate. Because the prolyl hydroxylase enzyme is located in the ER, 13 • 14
this meant that the accumulated pool of procollagen must also be in the ER
and that the rate-controlling step in the secretion process is leaving the ER.
These experiments also showed that the underhydroxylated procollagen mol-
ecule shares the same pool and is capable of switching to the fast route if it
becomes hydroxylated and triple helical.
The reason why a triple helical molecule should be secreted six times
faster than its nonhelical counterpart is not understood but is critical to a
major action of ascorbate on the collagen pathway. The mechanism of pro-
collagen secretion has been debated for 25 years; yet it remains unclear
whether procollagen is secreted through the Golgi or more directly out of the
cell. Early experiments by Ross et a!. argued that the kinetics of labeling
during a pulse-chase experiment did not support transport through the Golgi. 63
Marchi and LeBlond tracked procollagen and other proteins using 3 H-proline
labeling in fibroblast cultures, and they presented evidence that the Golgi
was involved. 39 Antibody studies have localized procollagen in the Golgi.
Olsen et a!. showed by antibody staining that procollagen molecules could
be seen in the Golgi in tendon cells under conditions that sufficiently disrupted
the membranes and allowed antibody penetration. 43 -45 •53 On the other hand,
Pierce et a!. 59 using antibodies could not find type IV procollagen in the Golgi
of parietal yolk sack cells. Since EM immunostaining is subject to many
potential artifacts depending on the cell system and the antibody, definitive
conclusions are hard to draw using this technique alone. Moreover, the kinetic
data presented above would argue that procollagen can be secreted in more
than one way, making the interpretation of this earlier data even more difficult.
In addition, many other questions can be raised. Why is the Golgi apparatus
492 Nutrition and Gene Expression
TABLE 1
Temperature Sensitivity of Secretion from
PAT Cells
41 120' 20b 70
39 120' 20b 75
32 120' 20b 140
29 * 36 *
25 * * *
Note: * Not secreted with first order kinetics.
•.b Average value presented; differences between temper-
atures were not statistically significant.
so small in tendon cells when half their total protein production is secreted?69
Why are secretory vesicles in tendon cells difficult to see by electron mi-
croscopy? Why does secreted procollagen remain sensitive to endoglycosidase
H, 11 an indication that sugar rearrangements that should have occurred in the
Golgi have not taken place in the procollagen molecule? Why is the fast rate
of procollagen secretion so insensitive to temperature (Table 1), something
one would not expect if secretion were occurring by an energy intensive
process such as vesicle transport? Until more is understood about the process
of procollagen secretion, it is difficult to give a physical meaning to the
sixfold differences in secretion rates that appear to be critically dependent on
whether the procollagen molecule is triple helical.
expect that when secretion rates were reduced by addition of the inhibitor
that the pool of procollagen inside the cell would build up dramatically.
Instead, Kao et al. 34 observed that the internal procollagen pool remained
constant. They concluded that the inhibitor decreased procollagen translation
rates (procollagen degradation 5 could have played a role but was not dis-
cussed). This possibility for translational regulation could be tested directly
using this same cell type in primary culture (PAT cells). 64 In this case a,a'-
dipyridyl was given to cells fully induced with ascorbate, making half their
total protein production as procollagen, and to scorbutic cells, as a control.
The data was plotted as the ratio between counts incorporated into procollagen
and those into noncollagen protein (Figure 3). This controlled for the non-
specific decrease in protein synthesis ( ~40% )34 •64 resulting from a toxic effect
of chelation of ferrous ion by the inhibitor. As might be predicted, scorbutic
cells showed no specific change in procollagen production, confirming that
a cell inhibited at the proline hydroxylation step by a lack of ascorbate cannot
be further inhibited by a lack of ferrous ion. In contrast, fully induced cells
showed a specific and rapid reduction in procollagen translation rates. By 4
h the level had dropped by 2/3 and then leveled off with increasing time. In
the short time period of this experiment, there was no significant change in
procollagen mRNA levels.
The question can be raised that if there is a feedback linkage between
translation and secretion rates why is the drop in collagen production on
addition of a,a' -dipyridyl only 2/3 of the scorbutic rate. The answer is that
the linkage between these steps is better than it first appears. When one takes
into account that the inhibitor nonspecifically inhibits 1/3 of all protein syn-
thesis, then the procollagen translation rates need only drop by 2/3 to bring
them into synchrony with the ability of the cell to secrete what is made.
Translation regulation is also observed when one induces procollagen
production with ascorbate (Figure 4). In this case, the efficiency of translating
procollagen mRNA increases by ~2-fold and this continues until the cells
reach a fully induced state where upon it returns to the original rate. 64 In the
context of the kinetics of the induction process, secretion rates are rapidly
altered by ascorbate (< 1 h), but full induction of translation rates requires a
much longer time ( ~48 h) in large part because of the slow increase in
procollagen mRNA levels (starting at 12 hand requiring ~72 h to complete). 64
Until translation rates can equal secretion rates the pool of procollagen within
the ER is depressed, and a consequence is that the cell raises its translational
efficiency.
The kinetics of the induction process are incompatible with changes in
degradation rates playing a significant role. 5 One might assume that scorbutic
cells producing nonhelical collagen would have more degradation. This fol-
lows from the fact that nonhelical collagen (gelatin) is susceptible to most
proteases, but the triple helical collagen molecule requires specific proteases,
called collagenases, to denote their unusual ability to attack the triple helical
494 Nutrition and Gene Expression
1.2
.....
z
w
(!'
c(
...J
...J
0
(.)
z
0
z......
z
w
(!'
c(
...J
...J
0
(.)
0
0::
....,
0..
0
i=
c(
0::
-----------~-----------~-----------
HOURS
FIGURE 3. Effect of adding cx,cx' -dipyridyl (0.3 mM) to ascorbate-induced (•) and uninduced
("')PAT cells. 64 The rates of procollagen synthesis have been normalized to that of noncollagen
protein synthesis to correct for the nonspecific inhibition of protein synthesis ( -40% in 6 h).
Each point in the graph reflects a 2-h pulse ending at the time indicated. The fully induced and
uninduced controls are shown as dashed lines ± SD on the right ordinate.
•
...J
w
~
ffi
~
w
~
ii::
w
;;::
!;(
...J
w
a:
I I I I I
48 12 16 20 24 28 32 36 40 44 48 52 56 60
observed. While this appeared to be much more rapid than the induction in
PAT cells, it is not inconsistent with that data. The reason is that procollagen
mRNA levels in the human fibroblasts never dropped more than approximately
twofold. Since translational efficiency can increase approximately twofold
after ascorbate addition, there is no need for mRNA levels to increase in order
to obtain full induction. It is the slow increase in mRNA levels that retards
the induction process in PAT cells.
Owen et at. 47 have shown that for rat osteoblasts there is a strong ascorbate
induction of collagen synthesis from ~2.5 to~ 30% after~ 36 h. Even though
detailed kinetics were not done and no comparisons were made with procol-
lagen mRNA levels, this study again shows that ascorbate can have a dramatic
effect on the collagen pathway and the amount of procollagen synthesized.
One can ask why the earlier results with ascorbate were so variable when
the more recent studies show a common response to ascorbate. One important
difference is that earlier studies used fibroblastic cell lines making low levels
of collagen. As discussed before, this could be reflecting cell culture con-
ditions that are dominantly negative for collagen expression. 77 Or it could be
reflecting the fact that long-term cultures lower their collagen production by
a mechanism that is not reversible by ascorbate addition. In addition, a cell
making 1% procollagen no longer taxes the secretory machinery of the cell.
At that level of synthesis, changes in the secretion rate may still alter the
pool size, but the initial pool would be so small that these changes may not
feed back to alter translation rates. Therefore, even though a cell making low
levels of collagen could respond to ascorbate, its lack of increase in collagen
production could be due to multiple factors.
Finally, a discussion of translational control and collagen regulation would
not be complete without reference to some of the unusual features of the
translational machinery of the cell. As might be expected, embryonic tendon
cells have a high proportion of membrane-bound ribosomes to support the
high level of procollagen translation rates. 31 •66 This only drops slowly when
cells are grown without ascorbate in contrast to the more rapid drop in pro-
collagen synthesis. This leads to a large disparity between the relative number
of membrane-bound ribosomes and the level of secreted proteins. A finding
that promotes the concept that the number of membrane-bound ribosomes is
a characteristic trait of the differentiated state at the time when the cells were
isolated. Similarly, the pool of proline and glycine tRNA is elevated in PAT
cells and this too is unaffected by the absence of ascorbate but does drop as
cells age developmentally. 46 Also, there is evidence to support the model that
there are specific sites on membrane-bound ribosomes for each procollagen
chain helping to organize the interaction between collagen chains that results
in a type I collagen molecule with two a 1 and one a 2 chains. 78 •79 Needless
to say, much more needs to be known about procollagen translation and its
regulation before these observations can be understood in molecular terms.
Schwarz 497
other steps would have to be induced before the transcriptional activity could
increase. In the case of a transcription factor model, a fairly rapid increase
in transcriptional activity would be expected. Ascorbate is rapidly taken up
by the cell; 6 •69 so, there is no simple explanation for the delay in mRNA
induction using this model. Second is the fact that isoascorbate, an isomer
whose uptake by the cell is strongly selected against, once inside the cell is
as active as ascorbate at inducing procollagen production. 35 These data support
the idea that the proline hydroxylation step can not discriminate between the
isomers. Since other reducing agents can participate in the reaction in vitro
it is questionable whether a specific recognition site exists. Since the isomers
have the same reducing potential, they are interchangeable at this step. How-
ever, one can question whether a transcription factor that specifically bound
ascorbate would also bind its isomer with equal efficiency. Again, these
questions only make it clear that more needs to be known about the controls
regulating procollagen mRNA levels and how these controls are specifically
changed during the ascorbate induction process.
change for type I collagen because the more hydroxylated chains stabilize the
triple helical conformation. With type IV collagen, even fully hydroxylated
molecules exist as single chains and may be secreted as single chains. 12 •50
Therefore, using the model that the increase in secretion rates is a result of
the conformation change of the procollagen molecule, the observation that
the secretion of typed IV collagen is not significantly changed by ascorbate
is consistent with that model (although more conclusive data is needed to
prove the point).
The effect of ascorbate on procollagen translation rates is even more
difficult to establish with other cell types. For instance, indirect evidence has
shown that ascorbate causes a remodeling of the ER in chondrocytes making
collagen types II and X. 5 1 In type X more procollagen is made after ascorbate,
but the kinetics have not been studied in enough detail to show a linkage
between this step and secretion rates. 37 With type II the analysis is quite
complex. Ascorbate has not been shown to increase synthesis and in some
cases there is a decrease. 37 Part of the problem is a consequence of chondro-
cytes being able to produce both type I and type II collagen. 20 •24 The presence
of ascorbate, especially for cells grown on plastic, can encourage the transition
to a more "fibroblastic" phenotype-reducing type II production. 3 It remains
to be seen if a chondrocyte, in suspension culture or in other conditions that
stabilize the chondrocyte phenotype, will respond to ascorbate by increasing
their type II procollagen translation rates.
The effect of ascorbate on mRNA levels remains to be understood in
more detail. In the case of chondrocytes, type X mRNA increases 14-fold,
7 days after ascorbate addition. In the same cells, type II mRNA drops 50%
but this occurs independently of ascorbate. 37
What conclusions can be drawn about the role of ascorbate in regulating
other collagen types? Even though there are strong analogies in some situations
to the type I example, more studies will have to be done especially with cell
types in cell culture conditions permissive for high procollagen expression.
As was seen with secretion and type IV collagen, the differences as well as
the similarities to the type I situation will be informative about how specific
cell types regulate each collagen type.
Because ascorbate affects many pathways and because the collagen path-
way can be altered at many levels, it is not surprising that ascorbate can affect
collagen production indirectly. In these cases, the primary action of ascorbate
is not on the collagen pathway. As a consequence of that change, however,
there is an alteration in collagen synthesis. There are probably many such
interactions, but two well-documented cases will be discussed below.
The first involves the complete removal of ascorbate from the diet of the
guinea pig. The consequence is that the guinea pig stops eating after two
Schwarz 501
weeks and literally starves. 9 This unusual behavior may result from an im-
balance caused by a lack of an essential vitamin. In any case, collagen
production drops rapidly and can be shown to be equally affected by food
deprivation in the presence of ascorbate. 9 This is an unusual case. Lack of
appetite in the guinea pig is associated only with complete removal of the
vitamin from the diet and a comparable effect is not observed in humans.
Nevertheless, it is a good example of how in the whole organism the ascorbate
effect can become quite complex. Peterkofsky and her co-workers have ana-
lyzed this problem in detail and they have found that the lack of vitamin C
causes a change in the blood. 49 The serum from fasted animals can be shown
to reduce the collagen production in cells in culture. The active factor appears
to bind to and inhibit the activity of insulin-like growth factor I in the fasted
serum. 4 •8 How this factor alters the collagen pathway is still under investi-
gation.
The second case involves the oxidation of lipids to aldehydes. 7• 10 ·23 Chojk-
ier et a!. 7• 10 have shown that aldehydes can induce collagen production in
certain cells in culture. The human fibroblasts used were making about 3%
collagen, and this increased approximately twofold by either ascorbate or
aldehyde addition. In this case, the role of ascorbate is difficult to interpret
since in the highly oxygenated environment of cell culture, ascorbate breaks
down and can promote the oxidation of the lipids to aldehydes. This mech-
anism of collagen stimulation may be important in wound repair and in the
fibrosis due to injury seen in the lung and other tissues. It is probably not a
major route of ascorbate stimulation of collagen synthesis. Ascorbate is thought
to prevent lipid oxidation under normal circumstances. 21 In addition, ascorbate
and isoascorbate would be expected to have equal activities in stimulating
collagen if oxidation was the critical factor since the isomers are oxidized
outside the cell at about the same rate. 35 In fact, isoascorbate is about three-
to fivefold less effective than ascorbate at stimulating PAT cells because of
its inability to be taken up effectively by the cell. 35 Nevertheless, one has to
keep in mind when dealing with multifaceted molecules and multiregulated
pathways, alternative mechanisms have to be expected and can play important
roles in specific cases.
IX. CONCLUSIONS
Ascorbate acts as a specific inducer of collagen production in many cells
producing type I collagen. If it had worked directly on transcriptional regu-
lation of the collagen gene, it would have been a mainstream research topic,
but it would have revealed little new about the regulation of the collagen
pathway except perhaps a new promotor binding site. The fact that a molecule
with little more than a good reducing potential can act to specifically stimulate
procollagen synthesis requires a new look at protein regulation. The critical
concept that this molecule keeps teaching us about the collagen pathway (type
I) is that it is tightly coupled through feedback regulation. Therefore, ascorbate
502 Nutrition and Gene Expression
levels (or cell density) by controlling the activity of the proline hydroxylation
step can control the flow through the whole pathway. Hydroxylation levels
control whether the collagen molecule is triple helical. This conformation
changes the secretion rates by sixfold. This in tum can alter the pool inside
the ER; the pool size alters translational efficiency; and translational efficiency
alters the half-life of the procollagen mRNA.
It is also clear that not all collagen types respond to ascorbate in the same
way. But this may be an advantage in that the differences may reveal even
more about how cells can control tissue-specific functions. The action of
ascorbate on other collagen types promises to be an area that will yield
important new information about specific points of control within the collagen
pathway.
At the same time one has to be aware that in the whole organism ascorbate
can affect many biological pathways in nonmesenchymal tissues, and these
changes can feedback to alter collagen production. Likewise, the ability of
the molecule to form free radicals in an oxygenated environment can modulate
the collagen pathway indirectly. In the end, as we better understand the
complexities of ascorbate action on the cell, this will all add to our under-
standing of how the differentiated state can be controlled.
After 70 years since the discovery of ascorbate, we can finally give a
molecular model for why scurvy is a collagen disease. But to its credit, this
specific inducer of collagen synthesis has been a key to unlocking the complex
controls that regulate the differentiated state of the cell.
REFERENCES
I. Aronow, M. A., Gerstenfeld, L. C., Owen, T. A., Tassinari, M. S., Stein, G. S.,
and Lian, J. B., Factors that promote progressive development of the osteoblast phe-
notype in cultured fetal rate calvaria cells, J. Cell. Physiol., 143, 213, 1990.
2. Bates, C. J., Bailey, A. J., Prynne, C. J., and Levine, C. I., The effect of ascorbic
acid on the synthesis of collagen precursor by 3T6 mouse fibroblasts in culture, Biochim.
Biophys. Acta, 278, 372, 1972.
3. Benya, P. D. and Shaffer, J. D., Dedifferentiated chondrocytes reexpress the differ-
entiated collagen phenotype when cultured in agarose gels, Cell, 30, 215, 1982.
4. Berg, R. A. and Prockop, D. J., The thermal transition of a nonhydroxylated form of
collagen. Evidence for a role for hydroxyproline in stabilizing the triple-helix of collagen,
Biochem. Biophys. Res. Commun., 52, 115, 1973.
S. Bienkowski, R. S., Curran, S. F., and Berg, R. A., Kinetics of intracellular degradation
of newly synthesized collagen, Biochemistry, 25, 2455, 1986.
6. Blanck, T. J. J. and Peterkofsky, B., The stimulation of collagen secretion by ascorbate
as a result of increased proline hydroxylation in chick embryo fibroblasts, Arch. Biochem.
Biophys., 171, 259, 1975.
7. Brenner, D. A. and Chojkier, M., Acetaldehyde increases collagen gene transcription
in cultured human fibroblasts, J. Bioi. Chem., 262, 17690, 1987.
Schwarz 503
8. Chan, D. Lamande, S. R., Cole, W. G., and Bateman, J. F., Regulation ofprocollagen
synthesis and processing during ascorbate-induced extracellular matrix accumulation in
vitro, Biochem. 1., 269, 175, 1990.
9. Chojkier, M., Spanheimer, R., and Peterkofsky, B., Specifically decreased collagen
biosynthesis in scurvy dissociated from an effect on proline hydroxylation and correlated
with body weight loss, J. Clin. Invest., 72, 826, 1983.
10. Chojkier, M., Houglum, K., Solis-Herruzo, J., and Brenner, D. A., Stimulation of
collagen gene expression by ascorbic acid in cultured human fibroblasts: a role for lipid
peroxidation, J. Bioi. Chern., 264, 16957, 1989.
II. Clark, C. C., The distribution and initial characterization of oligosaccharide unit on the
COOH-terminal propeptide extensions of the pro- I and pro- 2 chains of type I pro-
collagen, J. Bioi. Chern., 254, 10798, 1979.
12. Crouch, E. and Bornstein, P., Characterization of a type IV procollagen synthesized
by human amniotic fluid cells in culture, J. Bioi. Chern., 254, 4197, 1979.
13. Cutroneo, K. R., Guzman, N. A., and Sharawy, M. M., Evidence for a subcellular
vesicular site of collagen prolyl hydroxylation, J. Bioi. Chern., 249, 5989, 1974.
14. Diegelmann, R. F., Bernstein, L., and Peterkofsky, B., Cell-free collagen synthesis
on membrane-bound polysomes of chick embryo connective tissue and the localization
of prolyl hydroxylation on the polysome-membrane complex, J. Bioi. Chern., 248, 6514,
1973.
15. Diliberto, E. J., Jr., Daniels, A. J., and Viveros, 0. H., Multicompartmental secretion
of ascorbic acid and its dual role in dopamine-'Y-hydroxylation, Am. J. Clin. Nutr., 54,
1163S, 1991.
16. Doty, P. and Nishihara, T., The molecular properties and thermal stability of soluble
collagen, in Recent Advances in Gelatin and Glue Research, Stainsberg, G., Ed., Per-
gamon Press, London, 1957, 92.
17. Eipper, B. A. and Mains, R. E., The role of ascorbate in the biosynthesis of neuroen-
docrine peptides, Am. J. Clin. Nutr., 54, 1153S, 1991.
18. Englard, S. and Seifter, S., The biochemical functions of ascorbic acid, Annu. Rev.
Nutr., 6, 365, 1986.
19. Fessler, L. I. and Fessler, J. H., Protein assembly of procollagen and effects of hy-
droxylation, J. Bioi. Chern., 249, 7637, 1974.
20. Finer, M. H., Gerstenfeld, L. C., Young, D., Doty, P., and Boedtker, H., Collagen
expression in embryonic chicken chondrocytes treated with phorbol myristate acetate,
Mol. Cell. Bioi., 5, 1415, 1985.
21. Frei, B., Ascorbic acid protects lipids in human plasma and low-density lipoprotein
against oxidative damage, Am. J. Clin. Nutr., 54, 1113S, 1991.
22. Friedrich, W., in Vitamins, Walter de Gruyter, Berlin, 1988, 929.
23. Geesin, J. C., Gordon, J. S., and Berg, R. A., Retinoids affect collagen synthesis
through inhibition of ascorbate-induced lipid peroxidation in cultured human dermal
fibroblasts, Arch. Biochem. Biophys., 278. 350, 1990.
24. Gerstenfeld, L. C., Finer, M. H., and Boedtker, H., Quantitative analysis of collagen
expression in embryonic chick chondrocytes having different developmental fates, J.
Bioi. Chern., 264,5112, 1989.
25. Green, H. and Goldberg, B., Collagen synthesis by human fibroblast strains, Proc.
Soc. Exp. Bioi. Med., 117, 258, 1964.
26. Green, H., Goldberg, B., and Todaro, G., Differentiated cell types and the regulation
of collagen synthesis, Nature (London), 212, 631, 1966.
27. Jimenez, S. A. and Yankowski, R., Role of molecular conformation on secretion of
chick tendon procollagen, J. Bioi. Chern., 253, 1420, 1978.
28. Kalcheim, C. and Levie!, V., Stimulation of collagen production in vitro by ascorbic
acid released from explants of migrating avian neural crest, Cell Diff, 22, 107, 1988.
504 Nutrition and Gene Expression
29. Kao, W.-Y.W., Kao, W.-C. C., and Schwarz, R. 1., Prolyl hydroxylase production
can be uncoupled from the regulation of procollagen synthesis, Exp. Cell Res., 157, 265,
1985.
30. Kao, W. W-Y. and Berg, R. A., Cell density-dependent increase in prolyl hydroxylase
activity in cultured L-929 cells requires de novo protein synthesis, Biochim. Biophys.
Acta, 586, 528, 1979.
31. Kao, W. W-Y., Flaks, J. G., and Prockop, D. J., Primary and secondary effects of
ascorbate on procollagen synthesis and protein synthesis by primary cultures of tendon
fibroblasts, Arch. Biochem. Biophys., 173, 638, 1976.
32. Kao, W. W-Y., Mai, S. H., Chou, K-L., and Ebert, J., Mechanism for the regulation
of post-translational modifications of procollagens synthesized by matrix-free cells from
chick embryos, J. Bioi. Chern., 258, 7779, 1983.
33. Kao, W. W-Y., Berg, R. A., and Prockop, D. J., Kinetics for the secretion of pro-
collagen by freshly isolated tendon cells, J. Bioi. Chern., 252, 8391, 1977.
34. Kao, W. W-Y., Prockop, D. J., and Berg, R. A., Kinetics for the secretion ofnonhelical
procollagen by freshly isolated tendon cells, J. Bioi. Chern., 254, 2234, 1979.
35. Kipp, D. and Schwarz, R. I., Effectiveness of isoascorbate versus ascorbate as an
inducer of collagen synthesis in primary avian tendon cells, J. Nutr., 120, 185, 1990.
36. Knaack, D., Podleski, T. R., and Salpeter, M. M., Ascorbic acid and acetylcholine
receptor expression, Ann. N.Y. Acad. Sci., 498, 77, 1987.
37. Leboy, P. S., Vaias, L., Uschmann, B., Golub, E., and Adams, S. L., Ascorbic acid
induces alkaline phosphatase, type X collagen, and calcium deposition in cultured chick
chondrocytes, J. Bioi. Chern., 264, 17281, 1989.
38. Lyons, B. L. and Schwarz, R. I., Ascorbate stimulation of PAT cells causes an increase
in transcription rates and a decrease in degradation rates of procollagen mRNA, Nucleic
Acids Res., 12, 2569, 1984.
39. Marchi, F. and Leblond, C. P., Radioautographic characterization of successive com-
partments along the rough endoplasmic reticulum-golgi pathway of collagen precursors
in foot pad fibroblasts of [3H]proline-injected rats, J. Cell Bioi., 98, 1705, 1984.
40. Margolis, R. L. and Lukens, L. N., The role of hydroxylation in the secretion of
collagen by mouse fibroblasts in culture, Arch. Biochem. Biophys., 147, 612, 1971.
41. Murad, S., Grove, D., Lindberg, K. A., Reynolds, G., Sivarajah, A., and Pinnell,
S. R., Regulation of collagen synthesis by ascorbic acid, Proc. Nat/. Acad. Sci. U.S.A.,
78, 2879, 1981.
42. Niki, E., Action of ascorbic acid as a scavenger of active and stable oxygen radicals,
Am. J. Clin. Nutr., 54, 1119S, 1991.
43. Nist, C., von der Mark, K., Hay, E. D., Olsen, B. R., Bornstein, P., Ross, R., and
Dehm, P., Localization of procollagen in chick corneal and tendon fibroblasts with
ferritin-conjugated antibodies, J. Cell Bioi., 65, 75, 1975.
44. Olsen, B. R., Berg, R. A., Kishida, Y., and Prockop, D. J., Collagen synthesis:
localization of prolyl hydroxylase in tendon cells detected with ferritin-labeled antibodies,
Science, 182, 825, 1973.
45. Olsen, B. R., Berg, R. A., Kishida, Y., and Prockop, D. A., Further characterization
of embryonic fibroblasts and the use of immunoferritin techniques to study collagen
biosynthesis. J. Cell Bioi., 64, 340, 1975.
46. Ouenzar, B., Weill, D., Agoutin, B., Keith, G., and Heyman, T., Relative content
of isoaccepting tRNAs for glycine and proline in avian tendon cells with different rates
of procollagen synthesis, Biochim. Biophys. Acta, 950, 429, 1988.
47. Owen, T. A., Aronow, M., Shalhoub, V., Barone, L. M., Wilming, L., Tassinari,
M. S., Kennedy, M. B., Pockwinse, S., Lian, J. B., and Stein, G. S., Progressive
development of the rate osteoblast phenotype in vitro: reciprocal relationships in expres-
sion of genes associated with osteoblast proliferation and differentiation during formation
of the bone extracellular matrix, J. Cell. Physioi., 143, 420, 1990.
Schwarz 505
48. Oyamada, I., Palka, J., Schalk, E. M., Takeda, K., and Peterkofsky, B., Scorbutic
and fasted guinea pig sera contain an insulin-like growth factor !-reversible inhibitor of
proteoglycan and collagen synthesis in chick embryo chondrocytes and adult human skin
fibroblasts, Arch. Biochem. Biophys., 276, 85, 1990.
49. Oyamada, I., Bird, T. A., and Peterkofsky, B., Decreased extracellular matrix pro-
duction in scurvy involves a humoral factor other than ascorbate, Biochem. Biophys. Res.
Commun., 152, 1490, 1988.
50. Pacifici, M., Independent secretion of proteoglycans and collagens in chick chondrocyte
cultures during acute ascorbic acid treatment, Biochem. J., 272, 193, 1990.
51. Pacifici, M. and Iozzo, R. V., Remodeling of the rough endoplasmic reticulum during
stimulation of procollagens secretion by ascorbic acid in cultured chondrocytes, J. Bioi.
Chern., 263, 2483, 1988.
52. Paz, M. A. and Gallop, P. M., Collagen synthesized and modified by aging fibroblasts
in culture, In Vitro, II, 302, 1975.
53. Pesciotta, D. M. and Olsen, B. R., The cell biology of collagen secretion, in Immu-
nochemistry of the Extracellular Matrix, Vol. II, Furthmayr, H., Ed., CRC Press, Boca
Raton, FL, 1982, I.
54. Peterkofsky, B., The effect of ascorbic acid on collagen polypeptide synthesis and proline
hydroxylation during the growth of cultured fibroblasts, Arch. Biochem. Biophys., 152,
318, 1972.
55. Peterkofsky, B., Regulation of collagen secretion by ascorbic acid in 3T3 and chick
embryo fibroblasts, Biochem. Biophys. Res. Commun., 49, 1343, 1972.
56. Peterkofsky, B. and Diegelmann, R., Use of mixture of protease-free collagenase for
specific assay of radioactive collage in the presence of other proteins, Biochemistry, 10,
988, 1971.
57. Peterkofsky, B. and Udenfriend, S., Conversion of proline to collagen hydroxyproline
in a cell-free system from chick embryo, J. Bioi. Chern., 238, 3966, 1963.
58. Peterkofsky, B. and Udenfriend, S., Enzymatic hydroxylation of proline in microsomal
polypeptide leading to formation of collagen, Proc. Nat/. Acad. Sci. U.S.A., 53, 335,
1965.
59. Pierce, G. B., Jones, A., Orfanakis, N. G., Nakane, P. K., and Lustig, L., Biosyn-
thesis of basement membrane by parietal yolk sac cells, Differentiation, 23, 60, 1982.
60. Pinnell, S. R., Murad, S., and Darr, D., Induction of collagen synthesis by ascorbic
acid, Arch. Dermatol., 123, 1684, 1987.
61. Quinones, S. R., Neblock, D. S., and Berg, R. A., Regulation of collagen production
and collagen mRNA amounts in fibroblast in response to culture conditions, Biochem.
J., 239, 179, 1986.
62. Rebouche, C. J,, Ascorbic acid and carnitine biosynthesis, Am. J. Clin. Nutr., 54,
1147S, 1991.
63. Ross, R. and Benditt, E. P., Wound healing and collagen formation, J. Cell Bioi.,
27, 83, 1965.
64. Rowe, L. B. and Schwarz, R. I., Role of procollagen mRNA levels in controlling the
rate of procollagen synthesis, Mol. Cell. Bioi., 3, 241, 1983.
65. Schwarz, R. 1., Mandell, R. B., and Bissell, M. J., Ascorbate induction of collagen
synthesis as a means for elucidating a mechanism for quantitative control of tissue-specific
function, Mol. Cell. Bioi., 1, 843, 1981.
66. Schwarz, R. I., Kleinman, P., and Owens, N., Ascorbate can act as an inducer of the
collagen pathway because most steps are tightly coupled, N. Y. Acad. Sci., 498, 172,
1987.
67. Schwarz, R. 1., Colarusso, L., and Doty, P., Maintenance of differentiation in primary
avian tendon cells, Exp. Cell Res., 102, 63, 1976.
68. Schwarz, R. I. and Bissell, M. J., Dependence of the differentiated state on the cellular
environment: modulation of collagen synthesis in tendon cells, Proc. Nat/. Acad. Sci.
U.S.A., 74, 4453, 1976.
506 Nutrition and Gene Expression
69. Schwarz, R. I., Procollagen secretion meets the minimum requirements for the rate-
controlling step in the ascorbate induction of procollagen synthesis, J. Bioi. Chern .. 260,
3045, 1985.
70. Schwarz, R. I., Cell-to-cell signaling in the regulation of procollagen expression in
primary avian tendon cells, In Vitro Cell. Dev. Bioi., 27a, 698, 1991.
71. Schwarz, R. I., Farson, D. A., and Bissell, M. J., Requirements for maintaining the
embryonic state of avian tendon cells in culture, In Vitro, 15, 941, 1979.
72. Spanheimer, R. G., Bird, T. A., and Peterkofsky, B., Regulation of collagen synthesis
and mRNA levels in articular cartilage of scorbutic guinea pigs, Arch. Biochern. Biophys.,
246, 33, 1986.
73. Spanheimer, R. C. and Peterkofsky, B., A specific decrease in collagen synthesis in
acutely fasted, vitamin C-supplemented, guinea pigs, J. Bioi. Chern., 260, 3955, 1985.
74. Szent-Gyorgyi, A., Observations on the function of peroxidase systems and the chemistry
of the adrenal cortex. Description of a new carbohydrate derivative, Biochern. J .. 22,
1387, 1928.
75. Szent-Gyorgyi, A. and Haworth, W. N., 'Hexuronic acid' (ascorbic acid) as the an-
tiscorbutic factor, Nature, 131, 24, 1933.
76. Tajima, S. and Pinnell, S. R., Regulation of collagen synthesis by ascorbic acid.
Ascorbic acid increases type I procollagen mRNA, Biochern. Biophys. Res. Cornrn .. 106,
632, 1982.
77. Valmassoi, j. and Schwarz, R.I., High serum levels interfere with the normal differ-
entiated state of avian tendon cells by altering translational regulation, Exp. Cell Res.,
176, 268, 1988.
78. Veis, A. and Kirk, T. Z., The coordinate synthesis and cotranslational assembly of type
I procollagen, J. Bioi. Chern., 264, 3884, 1989.
79. Veis, A., Leibovich, S. J., Evans, J., and Kirk, T. Z., Supramolecular assemblies of
mRNA direct the coordinated synthesis of type I procollagen chains, Proc. Natl. Acad.
Sci. U.S.A., 82, 3693, 1985.
80. Vogel, Z., Daniels, M.P., Chen, T., Xi, Z-Y., Bachar, E., Ben-David, L., Rosenberg,
N., Krause, M., Duksin, D., and Kalcheim, C., Ascorbate-like factor from embryonic
brain. Role in collagen formation, basement membrane deposition, and acetylcholine
receptor aggregation by muscle cells, Ann. N. Y. Acad. Sci., 498, 13, 1987.
Chapter 22
TABLE OF CONTENTS
0:oc_8;:-c49;;:o3--;c;69:-;c61c--:-4=/93/$0.00 +-$.5_0
© 1993 by CRC Press. Inc. 507
508 Nutrition and Gene Expression
I. INTRODUCTION
II. METALLOTHIONEIN
A. ROLES OF METALLOTHIONEIN
Since the discovery of metallothionein, the function of this protein has
been debated and a number of potential roles have been postulated and tested.
The ability to serve as a cellular detoxification system has been well docu-
mented. 15 When a cell or organism is challenged with increased amounts of
a potentially damaging heavy metal, such as cadmium, metallothionein syn-
thesis is increased. The increased MT may then bind the cadmium, thus
reducing the toxic effect of the heavy metal. What is unclear is if this mech-
anism evolved as a protective mechanism against heavy metals or if it con-
veniently serves this role in addition to another role, such as the regulation
of free zinc levels within cells or protection against free-radical damage. 16
Zinc, a micronutrient required in the diet, 11 also activates the increase in MT
synthesis. Increases in MT protein allows for binding of excess zinc, thereby
serving as a homeostatic regulatory system for zinc. 16 It remains a largely
unanswered question whether the regulation of the required metal zinc, or the
toxic metals, such as mercury or cadmium, has been the driving force during
the evolution of the metallothionein regulatory system. Antiquity of the me-
tallothionein gene suggests the protein is serving a required, essential role. 15
Others have proposed roles for MT serving as an intracellular free radical
scavenger 17 or part of a system protecting the cells from insults such as ionizing
radiation or alkylating agents. 18 - 20
510 Nutrition and Gene Expression
C. TURNOVER OF METALLOTHIONEIN
The turnover of MT has not been studied as well as the induction process.
However, evidence has shown that the type of metal ions bound to MT has
a direct influence on the stability of the protein to proteolytic activity. In
general, the cellular content of MT protein is a composite of dietary and
hormonal inputs vs. metal release and other factors that influence degradation.
Readers are referred to more complete discussions of MT turnover than space
permits in this chapter. 15 •16
(e.g., zinc) in the diet regulate levels of metallothionein in the kidney, liver,
intestine, and other tissues. 15 · 16 Increases in the abundance of MT is much
more striking in kidney, liver, and intestine, most likely due to changes in
the metabolic compartment that regulates transcriptional control by metals.
Early experiments done to quantitate changes in metallothionein levels by
dietary zinc used 35 S and/or 65 Zn labeling of cellular protein followed by
chromatography of cellular extracts. The increase in MT synthesis has since
been shown to be the result of increased MT gene transcription, with maximal
levels of MT mRNA abundance occurring a few hours before the increased
levels of protein are detected. 28 ·29
~-actin-
Metallothionein -
~-actin-
Metallothionein -
FIGURE I. Northern blot analysis of metallothionein mRNA (bottom bands) and ~-actin
mRNA (top bands). Total RNA was extracted from liver, kidney. intestine, thymus, spleen,
lung, and heart of rats that were fed diets supplying 5, 30, or 180 mg zinc per kg (as indicated)
for 2 weeks. Each dietary group was composed of five rats. Each lane had 7.4 j.Lg of RNA.
Hybridization was to a mixed 60-base probe for metallothionein-1 and -2 mRNA plus a 60-base
probe for ~-actin mRNA. (From Cousins, R. J. and Lee-Ambrose, L. M .. J . Nutr. , 122, 56,
1992. With permission.)
~-actin-
t.Aetallothionein 1-
~-act i n-
t.Aetallothionein 2 -
Northern Blot
Fetal Liver Metollothionein mRNA
1AL 5R 180R 30R 30AL
._550b
FIGURE 3. Northern blot analysis of fetal liver metallothionein mRNA . Pregnant rats were
fed dietary zinc at 1, 5, 30, or 180 mg/kg diet from days 13 through 20 of gestation. Intake was
either restricted (R) to that of the I mg zinc per kg group or ad libitum (AL). Total RNA from
fetal liver was fractionated in 1. 1% agarose gels. Hybridization of RNA transferred by Northern
blot was to a 32 P-labeled 60-base probe oligonucleotide DNA probe for metallothioneins I and
2. The 550b metallothionein mRNA band is indicated. (From Huber, K. L. and Cousins, R. J..
J . Nutr., 118 , 1570, 1988. With permission .)
C. OTHER STUDIES
Although metallothionein has been consistently considered a cytosolic
protein, immunocytochemical localization 42 has confirmed the presence of
metallothionein in the nucleus. Whether this is real or an artifactual contam-
516 Nutrition and Gene Expression
ination has not been definitively addressed. However, with the identification
recently of zinc-finger-containing transcription factors, the possibility of me-
tallothionein playing a role in nuclear regulation of zinc may exist. In fact,
Zeng et al. 8 have speculated that metallothionein may play a role in the
regulation of zinc available for these zinc-containing nuclear metalloproteins,
perhaps adding another level of control to the process of gene regulation.
Specifically, they have shown that apo-metallothionein can remove zinc from
the transcription factor Spl, decreasing the DNA-binding activity of the pro-
tein. Since the binding constant of MT for metals is considerably greater than
other proteins, they propose metal exchange from Spl to apo MT occurs.
Presumably, the nuclear level of MT protein could regulate the availability
of Zn for Sp 1 and influence regulation of specific genes.
Cousins and Lee-Ambrose 39 have investigated the relationship between
the dietary regulation of metallothionein and the uptake of zinc into the nucleus
in a variety of rat tissues to determine the changes in nuclear zinc brought
about by dietary change. Growing male rats were fed 5, 30, or 180 mg zinc
per kg for a 2-week period. Nuclei purified by sucrose gradient methods from
the liver, kidney, and spleen had approximately the same total zinc content
as measured by atomic absorption. Subsequently, a constant dietary zinc intake
of either 5, 30, or 180 mg zinc per kg was provided to rats for 2 d, followed
by gavage of liquified diet containing those same levels of zinc. By tagging
the gavaged zinc with 65 Zn, it was shown that substantial amounts of dietary
zinc entered cell nuclei shortly after the oral feeding. Two hours after this
oral dose, nuclei were isolated from the tissues. Intestine and liver accu-
mulated substantial amounts of zinc and directly reflected the dietary zinc
level as shown in Table 1. For example, nuclei from kidney accumulated up
to 6.2% of the zinc in the oral dose during the 2-h period. Additionally,
chromatography of extracts from liver nuclei showed that 65 Zn introduced
into the portal supply bound to specific fractions of nuclear proteins, including
one specific fraction that could contain the zinc-containing metal responsive
element binding protein (MRE-BP) that stimulates transcription of the me-
tallothionein genes. Clearly, this study shows the rapidity of the association
of dietary zinc with nuclear proteins, consistent with the known temporal
regulation of the metallothionein gene by zinc administration. Westin and
Schaffne~ 3
showed that a MREd binding protein from nuclear extracts of
HeLa cells binds DNA in a zinc-dependent fashion. Andersen et al. 27 and
Searle 23 have made similar observations. This supports the hypothesis that an
influx of zinc from the diet rapidly reaches the nucleus and influences tran-
scription. The interrelationships among dietary zinc, MREs, and MRE-BP
are presented in Figure 4.
At the interface between contemporary molecular biology and traditional
dietary studies is the elegant transgenic mouse work done by Palmiter and
others. 22 •23 Chimeric fusion genes were created using recombinant techniques.
Shay and Cousins 517
TABLE 1
Nuclear Zinc Derived from
Dietary Zinca
These genes had the mouse MT -1 promoter fused to the coding sequences of
a variety of genes including rat and human growth hormones. The gene
constructs were injected into the pronucleus of fertilized mouse ova. The
microinjected ova were then reimplanted into the oviducts of surrogate moth-
ers. Upon screening the offspring, it was found that approximately 25% were
expressing the transgenes. The level of growth hormone assayed in the trans-
genic animals was found to be regulated by zinc levels in coordination with
the native MT genes. This technology has many applications where dietary
zinc could provide the signal for transcriptional activation of trans genes. Zinc
is relatively nontoxic, and we have shown that the native MREs provide
excellent regulation by this micronutrient at the physiological range of dietary
intakes.
Of equal importance to zinc regulation of transgenes is the effect dietary
zinc could potentially have in metallothionein-related drug resistance. There
is convincing evidence that resistance to many alkylating agents (e.g., cis-
platin) used in chemotherapy is directly related to MT expression. 18-20 If
methods could be developed to block the role of dietary zinc in this activation,
drug resistance may be decreased.
Zinc = •
Dietary
Zinc Supply
~--,...•
.... MRE-BP
k ·~
~pl~m ~~
Nucleus 7 ~
C,,. . ~
, w- -
--
MRE Zn-regulated
Gene
7
of Balb/c mice treated with a variety of heavy metals given by i.p. injection.
Mercury elicited the strongest response, followed in order of response by Cd
> Pb > Cu > Ni > Zn > Mg. Other acute-phase reactants, such as alpha-
1-antitrypsin and alpha-1-antichymotrypsin, were unaffected by the metal
treatment. Additionally, albumin, which is downregulated during acute-phase,
was not repressed by the metal treatment, which indicates that the induction
of these mRNAs was not due to a metal-mediated inflammatory response.
Adrenalectomized animals were also challenged with metals, and the response
was still present, indicating that the induction by metals was not mediated
by the glucocorticoid induction pathway. They also noted that some DNA
sequence homologies exist between the MREs of the MT genes and sequences
in the promoter region of the alpha-1-acid glycoprotein and C-reactive protein
genes. Metal induction potential of these acute phase genes were tested in a
transiently tranfected cell system. Approximately 600 bp of the alpha-1-acid
Shay and Cousins 519
histidine and cysteine residues of the "LIM" motif was recognized by Freyd
et al. 10 The latter is found in some transcription factors and has some features
in common with the consensus sequence of zinc-finger proteins. Certainly,
the structural features of this protein coupled with the nearly absolute local-
ization to the intestine and induction at weaning give us strong circumstantial
evidence that this zinc-binding protein is crucially important to nutrient ab-
sorption. It will be both interesting and a challenge to design experiments
that define the nutritional regulation of CRIP.
V. SUMMARY
REFERENCES
I. Vallee, B. L. and Galdes, A., The metallobiochemistry of zinc enzmes, Adv. Enzymol.,
57, 283, 1984.
2. Lehninger, A. L., Principles ofBiochemistry, Worth Publishers. New York, 1982, 220.
3. Miller, J., McLachlan, A. D., and Klug, A., Repetitive zinc-binding domains in the
protein transcription factor IliA from Xenopus oocytes, EMBO J .. 4, 1609, 1985.
4. Engelke, D. R., Ng, S.-Y., Shastry, B. S., and Roeder, R. G., Specific interaction
of a purified transcription factor with an internal control region of 5S RNA genes, Cell,
19, 7l7, 1980.
5. Rosenberg, U. B., Schroder, C., Preiss, A., Kienlen, A., Cote, S., Riede, I., and
Jackie, H., Structural homology of the product of the Drosophila Kruppel gene with
Xenopus transcription factor IliA, Nature, 319, 336, 1986.
6. Tautz, D., Lehman, R., Schnurch, H., Schuh, R., Seifert, E., Kienlin, A., Jones,
K., and Jackie, H., Finger protein of novel structure encoded by hunchback, a second
member of the gap class of Drosophila segmentation genes, Nature (London), 327, 383,
1987.
7. Page, D. C., Mosher, R., Simpson, E. M., Fisher, E. M. C., Mardon, G., Pollack,
J., McGillivray, B., de Ia Chapelle, A., and Brown, L. G., The sex-determining region
of the human Y chromosome encodes a finger protein, Cell, 51, 1091, 1987.
8. Zeng, J., Heuchel, R., Schaffner, W., and Kagi, J. H. R., Thionein (apometallothi-
onein) can modulate DNA binding and transcription activation by zinc finger containing
factor Sp1, FEBS Lett., 279, 310, 1991.
9. Li, P. M., Reichert, J., Freyd, G., Horvitz, H. R., and Walsh, C. T., The LIM
region of a presumptive Caenorhabditis elegans transcription factor is an iron-sulfur and
zinc-containing metallodomain, Proc. Nat/. Acad. Sci. U.S.A., 88, 9210, 1991.
10. Freyd, G., Kim, S. K., and Horvitz, H. R., Novel cysteine-rich motif and homeodomain
in the product of the Caenorhabditis elegans cell lineage gene lin-11, Nature (London),
344, 876, !990.
11. Hambidge, K. M., Casey, C. E., and Krebs, N. F., Zinc, in Trace Elements in Human
and Animal Nutrition II, Mertz, W., Ed., Academic Press, Orlando, 1986, I.
12. Piletz, J. E. and Ganschow, R. E., Zinc deficiency in murine milk underlies expression
of the lethal-milk (lm) mutation, Science, 199, 181, 1978.
13. Lombeck, I., Schnippering, H. G., Ritz!, F., Feinendegen, L. H., and Bremner,
H. J., Absorption of zinc in acrodermatitis enteropathica, Lancet, I, 855, 1975.
14. Flagstad, T., Zinc absorption in cattle with a dietary picolinic acid supplement, J. Nutr.,
111' 1996, 1981.
15. Kagi, J. H. R. and Kojima, Y., Chemistry and biochemistry of metallothionein, in
Metallothionein ll, Experientia Supplementum, Vol. 52, Kagi, J. H. R. and Kojima, Y.,
Eds., Birkhauser Verlag, Basel, 1987, 25.
16. Dunn, M. A., Blalock, T. L., and Cousins, R. J., Metallothionein, Proc. Soc. Exp.
Bioi. Med., 185, 107, 1987.
17. Thornalley, P. J. and Vasak, M., Possible role for metallothionein in protection against
radiation-induced oxidative stress. Kinetic and mechanism of its reaction with superoxide
and hydroxyl radicals, Biochim. Biophys. Acta, 827, 36, !985.
18. Lohrer, H. and Robson, T., Overexpression of metallothionein in CHO cells and its
effect on cell killing by ionizing radiation and alkylating agents, Carcinogenesis, 10,
2279, 1989.
19. Kelley, S. L., Basu, A., Teicher, B. A., Hacker, M. P., Hamer, D. H., and Lazo,
J_ S., Overexpression of metallothionein confers resistance to anticancer drugs, Science,
241, 1813, 1988.
522 Nutrition and Gene Expression
20. Kaina, B., Lohrer, H., Karin, M., and Herrlich, P., Overexpressed human metal-
lothionein IIA gene protects Chinese hamster ovary cells from killing by alkylating agents,
Proc. Nat/. Acad. Sci. U.S. A., 87, 2710, 1990.
21. Durnam, D. M. and Palmiter, R., Transcriptional regulation of the mouse metallothi-
onein-1 gene by heavy metals, J. Bioi. Chern., 256, 6712, 1981.
22. Stuart, G. W., Searle, P. F., Cen, H. Y., Brinster, R. L. and Palmiter, R. D., A
12-base-pair DNA motif that is repeated several times in metallothionein gene promoters
confers metal regulation to a heterologous gene, Proc. Nat/. Acad. Sci. U.S.A., 81, 7318,
1984.
23. Searle, P. F., Zinc dependent binding of a liver nuclear factor to metal response element
MRE-a of the mouse MT-1 gene and variant sequences. Nucleic Acids Res., 18, 4683,
1990.
24. Hamer, D., Metallothionein, Annu. Rev. Biochem., 55, 913, 1986.
25. Seguin, C. and Prevost, J., Detection of a nuclear protein that interacts with a metal
regulatory element of the mouse MT-1 gene, Nucleic Acids Res., 16, 10547, 1988.
26. lmbert, J., Zafarullah, M., Culotta, V. C., Gedamu, L., and Hamer, D., Transcrip-
tion factor MBF-1 interacts with metal regulatory elements of higher eucaryotic MT genes,
Mol. Cell. Bioi., 9, 5315, 1989.
27. Andersen, R. D., Taplitz, S. J., Oberbauer, A.M., Calame, K. L., and Herschman,
R., Metal-dependent binding of a nuclear factor to the rat MT-1 promoter, Nucleic Acids
Res., 18, 6049, 1990.
28. McCormick, C. C., Menard, M.P., and Cousins, R. J., Induction of hepatic MT by
feeding zinc to rats of depleted zinc status, Am. J. Physiol., 240, E414, 1981.
29. Blalock, T. L., Dunn, M. A., and Cousins, R. J., Metallothionein gene expression in
rats: tissue-specific regulation by dietary copper and zinc, J. Nutr., 118, 222, 1988.
30. Richards, M. P. and Cousins, R. J., Metallothionein and its relationship to the me-
tabolism of dietary zinc in rats, J. Nutr., 106, 1591, 1976.
31. Richards, M. P. and Cousins, R. J., Zinc-binding protein: relationship to short term
changes in zinc metabolism, Proc. Soc. Exp. Bioi. Med., 153, 52, 1976.
32. Whanger, P. D. and Weswig, P. H., Effect of some copper antagonists on induction
of ceruloplasmin in the rat, J. Nutr., 100, 341, 1970.
33. Menard, M. P., McCormick, C. C., and Cousins, R. J., Regulation of intestinal
metallothionein biosynthesis in rats by dietary zinc, J. Nutr., 111, 1353, 1981.
34. Bremner, I. and Davies, N. T., The induction of metallothionein in rat liver injection
and restriction of food intake, Biochem. J., 149, 733, 1975.
35. Sato, M., Mehra, R. K., and Bremner, I., Measurement of plasma metallothionein-
1 in the assessment of the zinc status of zinc-deficient and stressed rats, J. Nutr., 114,
1683, 1984.
36. Grider, A., Kao, K. J., Klein, P. A., and Cousins, R. J., Enzyme-linked immuno-
sorbent assay for human metallothionein: correlation of induction with infection, J. Lab.
Clin. Med., 113, 221, 1989.
37. Grider, A., Bailey, L. B., and Cousins, R. J., Erythrocyte metallothionein as an index
of zinc status in humans, Proc. Natl. Acad. Sci. U.S.A., 87, 1259, 1990.
38. Huber, K. L. and Cousins, R. J., K562 cells respond to zinc and alter metallothionein
gene expression during differentiation, FASEB J., 6, 873A, 1992.
39. Cousins, R. J. and Lee-Ambrose, L. M., Nuclear zinc uptake and interactions and
metallothionein gene expression are influenced by dietary zinc in rats, J. Nutr., 122, 56,
1992.
40. Huber, K. L. and Cousins, R. J., Maternal zinc deprivation and interleukin-1 influence
metallothionein gene expression and zinc metabolism of rats, J. Nutr., 118, 1570, 1988.
41. Hurley, L. S., Teratogenic aspects of manganese, zinc and copper nutrition, Physiol.
Rev., 61, 249, 1981.
Shay and Cousins 523
42. Cherian, M. G., Templeton, D. M., Gallant, K. R., and Banerjee, D., Biosynthesis
and metabolism of metallothionein in rat during perinatal development, in Metallothionein
II, Experientia Supplementum, Vol. 52, Kagi, 1. H. R. and Kojima, Y., Eds., Birkhauser
Verlag, Basel, 1987, 499.
43. Westin, G. and Schaffner, W., A zinc-responsive factor interacts with a metal-regulated
enhancer element (MRE) of the mouse metallothionein-1 gene, EMBO J., 7, 3763, 1988.
44. Yiangou, M., Ge, X., Carter, K. C., and Papaconstantinou, J., Induction of several
acute-phase protein genes by heavy metals: a new class of metal-responsive genes, Bio-
chemistry, 30, 3798, 1991.
45. Shay, N. F. and Cousins, R. J., Identification and cloning of zinc-regulated mRNAs
from rat intestine, FASEB J., 6, A483, 1992.
46. Hempe, J. M. and Cousins, R. J., Cysteine-rich intestinal protein binds zinc during
transmucosal zinc transport, Proc. Natl. Acad. Sci. U.S.A., 88,9671, 1991.
47. Hempe, J. M. and Cousins, R. J., Cysteine-rich intestinal protein and intestinal me-
tallothionein: an inverse relationship as a conceptual model for zinc absorption in the rat,
J. Nutr., 122, 89, 1992.
48. Birkenmeier, E. H. and Gordon, J. I., Developmental regulation of a gene that encodes
a cysteine-rich intestinal protein and maps near the murine immunoglobulin heavy chain
locus, Proc. Nat/. Acad. Sci. U.S.A., 83, 2516, 1986.
Chapter 23
IRON-DEPENDENT REGULATION OF
FERRITIN SYNTHESIS
TABLE OF CONTENTS
0-8493-6961-4/93/$0.00 +$.50
© 1993 by CRC Press, Inc. 525
526 Nutrition and Gene Expression
I. OVERVIEW
Since the middle of the last century, it has been known that tissues contain
iron-positive granules called hemosiderin. 1 In 1937, Laufberger isolated and
crystallized another iron-rich compound from horse spleen and named it fer-
ritin to reflect its high iron content. 2 It has since been shown that ferritin is
a ubiquitous constituent of animal, plant, and microorganismal cells. Ferritin
consists of a protein shell made from subunits, usually 24 in number, sur-
rounding a cavity in which iron can be stored. Harrison et al. 3 provide an
excellent account of the physical features of ferritin and its function in iron
storage. In 1974 to 1975, two groups4 ·5 recognized heterogeneity in the sub-
units of the protein shell and eventually these were classified either as light
(L) or heavy (H) based on their slightly different molecular weights.
Drysdale and Munro 6 had shown in 1966 that iron induces ferritin syn-
thesis in the liver of the rat by a mechanism that is not quenched by acti-
nomycin D and therefore represents a posttranscriptional event. In 1976,
Zahringer et al. 7 confirmed and extended this observation by showing that in
rats not receiving extra iron a large number of ferritin mRNAs lie dormant
in their liver cell sap. When the rats were given iron supplements, these
reserve mRNAs became attached to ribosomes in the liver and started to
translate ferritin mRNA actively. They proposed that, at low levels of intra-
cellular iron, a specific protein binds to the anterior end (5' untranslated
region, 5'UTR) of ferritin mRNA, thus blocking initiation of ferritin protein
synthesis. When more iron enters the cell, the regulatory protein loosens so
that ferritin protein synthesis can proceed to meet the need for more ferritin
molecules to trap the excess iron. These and other studies of ferritin up to
1978 are reviewed elsewhere. 8 The present chapter deals in detail with research
performed during the past decade which has considerably increased our un-
derstanding of the mechanisms involved in the translational regulation of
ferritin mRNA availability in order to achieve iron homeostasis.
l-MRNA POLY A
H-MRNA POLY A
153 B
>lkb -so
RAT FERRITIN H-GENE
FIGURE 1. Upper diagram: Comparison of the number of nucleotide bases (B) and amino
acids (AA) of the coding regions of rat ferritin L and H subunit mRNAs. Also shown are the
features of the 5' and 3' untranslated regions with loop and stem structure at the same position
in the 5' regions of both mRNAs. Lower diagram: Comparison of the genomic structures of the
expressed rat ferritin L and H subunit genes. The exons are indicated by thick bars and the
introns by thin lines. The location of the TATA box, initiating methionine codon (ATG), ter-
mination codons (TAG,TAA), and polyA polymerase (AATAA) are indicated. (From Munro,
H. N. eta!., Ann. N.Y. Acad. Sci., 526, 117, 1988. With permission.)
liver H subunit as 100, rat liver shows 95% identity of sequence, mouse 94%,
and chicken 93%, thus suggesting more conservation.
The major features of the mRNAs of rat ferritin L and H subunits are
shown in Figure I. Despite closeness in size, the reading frames for rat L
and H are only 55% homologous; a similar divergence is seen when human
Land H subunits are compared, namely 58%. However, when the Chou and
Fasman rules 13 are applied, both subunits show five alpha-helices, thus sup-
porting a common ancestral gene. This suggests that L and H subunits may
interchange in the construction of the protein shell and thus give rise to
numerous isoferritins.
528 Nutrition and Gene Expression
Figure also shows that L and H subunit mRNAs have long 5' UTRs
which, for the most part, show little homology except for a sequence of 28
nucleotides close to the cap region. This region is highly conserved between
rat H and L and also between species as widely divergent as man and the
bullfrog. 14 On the basis of the reported sequences of this region for the L
chain of the human, rabbit, rat, and mouse, the consensus becomes:
UCUUGCUUCAACAGUGUUUGGACGGAAC L chain
UC~UGCUUCAACAGUGCUUGGACGGAAC H chain
The H ferritin of human, rat, mouse, and chicken are similar except for
substitution of C for U in two places in all four H sequences. On computer
analysis, the conserved sequence is able to form a double-stranded stem
supporting a loop (underlined) of six nucleotides. This sequence will be further
discussed under the section on regulation of ferritin mRNAs by iron.
Regarding the L and H ferritin genes of the rat, Southern blots show more
than 20 copies of the L gene 15 and 5 of the H gene. 16 Figure 1 displays the
exon and intron patterns of the single expressed L and H genes of the rat,
the others being considered as pseudogenes. The latter were evident when a
selection of L genes of the rat were sequenced and were found to contain no
introns, but to have polyadenylic acid tails and direct repeats in the adjacent
DNA, all of which is compatible with the random insertion of processed
pseudogenes into DNA by a retrovirus. Figure 1 shows that 3 introns (thin
lines) separate the 4 exons (thick lines) which encompass the 5 alpha-helices
of the L and H subunits, the small fifth alpha helix being present with the
fourth alpha helix on the last exon. Note also that the first and the last exons
also carry the 5' and 3' UTRs, respectively, of the mRNA. Irrespective of
whether the genes are from Lor H subunits, or are from man, rat, or chicken,
the middle two exons have identical lengths of 147 and 126 nucleotides,
respectively, representing 49 and 42 amino acids in the completed protein.
This is compelling evidence that the L and H genes share a common ancestry.
A. TRANSLATION
1. The Ferritin Model: The Iron-Responsive Element
As described in the introduction, injection of iron salts into the peritoneal
cavity of the rat is quickly followed by the synthesis of extra ferritin subunits
by a mechanism that is not quenched by prior injection of actinomycin D. 8
Munro et al. 529
DNA
4- Actinomycin
RNA
- Cordycepin
MRNA
~AAA
~~~r
/ PDLYRIBOSOMES
Fe~, ~Fe
DESFERR I-
OXAMINE '/
DPTA
FIGURE 2. Factors involved in regulation of ferritin subunit mRNAs. These include the supply
of mRNA from the nucleus, segregation of inactive ferritin mRNA, and its release for polyri-
bosome function when excess iron enters the cell. This is determined by the effect of the entering
iron on the free iron pool which can be restricted by chelation with desferioxamine which enters
the cell, but not with DPTA, which cannot enter. Note that iron can come from transferrin,
inorganic salts, or hemin, the latter through the action of heme oxygenase. (From Munro.
H. N. et al.. Ann. N.Y. Acad. Sci., 526. 117, 1988. With permission.)
-2.8
B
-0.97
2 3 4 ~ 6 78 9 10 II 12 13 14 I~ 16 17 18
FJGURE 3. Cytoplasmic distribution of the rat ferritin L subunit mRNA in control (above)
and iron-treated rats (below). Rats were injected intraperitoneally with ferric ammonium citrate
or saline (controls) and killed 3.5 h later. Sucrose gradients of liver homogenates were run. The
gradients were divided into 18 fractions from the top ("free'' mRNP) through monosomes to
the bottom (large polysomes). Each fraction was extracted from RNA, which was then run on
the Northern blot gel to show the quantity of L subunit RNA in each fraction using hybridisation
with labeled L subunit eDNA. The gel was then autoradiographed. (From Aziz , N. and Munro,
H. N., Nucleic Acid Res., 14,9 15 . 1986. With permission .)
migration of RNA alone on the gel, the binding of RNA to the protein retarded
532 Nutrition and Gene Expression
...
,~
- ;>'
AT(i
CAT ~
,., ~
5 CAT 3
.. ATG •0#
• 5• CAT 3
...
,~
&TG
CAT
•0#
~'
... '~
.....
5 CAT
CAT ~'
,~ ~o'
• 0#
EC EC EC s
5CAT3 5sCAT 3 5CAT
<(
z
0::
_J FT0-28 1
l{) 0 248 16
81-
82-
Free-
234567
FIGURE 5. RNA-protein complex formation with SIOO extracts from hepatoma cells (Ff0-
2B) treated with ferric ammonium citrate for 0, I, 2, 4. 8, and 16 h. Positions of complexes
Bl and B2 and free RNA are indicated. (From Leibold, E. A. and Munro. H. N .. Proc. Nat/.
Acad. Sci. U.S.A., 85.2171, 1988. With permission.)
the migration of the labeled 5' UTR-protein complex which appeared as two
bands, B-1 and B-2. When the cells were incubated with extra iron, there
was extensive loss of B-2 binding and a gain in B-1 binding. When the
fractions B-1 and B-2 were digested with RNAse T1, about 40 nucleotides
were protected from digestion including the 28-nucleotide section. Cross-
534 Nutrition and Gene Expression
linking using UV irradiation showed that proteins B-1 and B-2 were both
about 90 kDa. in molecular weight. Rouault et al., 26 using human H-mRNA,
also showed binding to human cell sap protein. In an extensive survey of the
binding proteins in 12 different species, Rothenberger et alY showed it to
be present in cytoplasmic extracts from human, mouse, chicken, frog, fish,
and fly, but was absent from bacterial, yeast, and plant sources. The proteins
found in animal sources varied from 95 to 100 kDa. Finally, like the rat/ 5
the mouse also displayed two binding proteins, thus confirming another de-
viation confined to these rodents, namely an insertion of eight amino acids
into the L subunit of the rae and the mouse 10 but not found in other species. 9
The next step was to purify the binding protein in order to characterize
and identify its structure. Rouault et al.2 6 •29 •30 describe a procedure in which
the regulatory RNA stem-loop structure (IRE) is used to trap the human
binding protein (IRE-BP). In order to do so, the segment of RNA containing
the IRE is reacted with biotin which makes the product capable of attaching
to avidin bound to an agarose column. After two cycles, an almost pure
product was obtained which migrated on an SDS/PAGE gel as a single band
with a molecular weight of 90 kDa. A peptide sequence from this purified
protein was used to prepare oligonucleotides to screen a human eDNA library.
A clone was identified which encoded a protein of 87 kDa. 29 We 31 also
employed biotinylation affinity chromatography to isolate the rat B-1 binding
protein, the yield being considerably increased by including six head-to-tail
IRE repeats. This protein had a molecular weight of 95 kDa and its N-terminal
amino acid sequence was close to that of the human. 29 •30 Walden et al. 28
employed a biochemical method using nucleotide binding dyes to isolate a
single polypeptide chain with a mol wt of about 90 kDa from rabbit liver.
Rouault et al. 29 noted in the amino acid sequence of their protein that it
included cysteine residues in motifs suggestive of those found in iron-sulfur
proteins. In particular, the mitochondrial enzyme aconitase shows 30% ho-
mology in its amino acid sequence compared with human binding protein. 32
This enzyme is active when all four Fe-S are present but can be inactivated
by loss of one iron atom. They therefore theorized that the gain or loss of
this iron atom may be the mechanism by which the IRE-BP senses iron levels
in the cell. The degree of homology of IRE-BP and aconitase is also em-
phasized by Hentze and Argos 34 who remark that another protein, isopro-
pylmalate isomerase, shows 23% homology with the binding protein se-
quence. It would be important to sequence the cytosolic form of aconitase
since this is the cell compartment in which the binding protein is located.
The activity of the binding protein may also vary with sulfhydryl levels in
the cytosoP 4 and indeed Barton et aJ.3 5 present a mechanism in response to
iron level involving multiple pools of IRE-BP, their characteristics, and ki-
netics including the important role of sulfhydryl groups (Figure 6).
As a step towards understanding how iron regulates the binding protein,
studies have focused on hemin and on chelatable cytosolic iron as likely
candidates. Figure 2 shows that chelatable free iron can be replenished from
Munro et al. 535
I
OXlDUD:I J'O»! REDUCED FORH I EI'WGEIIOUS CDI!nEl(
ln.cttve : low affinity I Active : high affinity I lllUtA bound
STUL£ I STAJLE I STA!LE
¥HS/
~Fe 1 HS'-R* lmRNA
~
F'\
0
mRN~
lll VIVO
i'Ri :
R*-
a m
RNA
s/ 'Fe :
-- ~ :~ -~- ~-:~-~- ~~~~--- --------------
IIVITIO
: HS/ 0 :
REDUCED FORM I
Active : •tah .tlinity I
I
UNSTABLE I
TABLE 1
Contrasting Effects of ALA on the Iron-
Dependent Induction of Ferritin and of Heme
Oxygenase Synthesis in Rat Fibroblasts
140 170
+ 690 290
+ 100 270 560
+ 500 110 590
+ 1000 130 1010
+ + 520 460
+ 100 + 530 480
+ 500 + 400 480
+ 1000 + 480 450
This thesis was explored in depth by Eisenstein et al. 40 who used rat
fibroblasts and rat hepatoma cells in culture to determine the effects of hemin,
other metalloporphyrins and nonheme iron (FEPIH) on the synthesis of ferritin
and on heme oxygenase, the latter being the enzyme that releases iron from
hemin. It was assumed that the level of activity of each enzyme reflects the
level of substrate, namely heme for heme oxygenase and chelatable iron for
ferritin. (This has been confirmed by later experiments [unpublished] in which
55
Fe was used to allow identification of the amounts of iron in different cell
components.) As shown in the published paper, 40 addition of hemin to the
medium induced both proteins, the oxygenase peaking first and releasing iron
for the subsequent increase in ferritin synthesis. Tin-mesoporphyrin, an in-
hibitor of heme breakdown by the oxygenase, delayed the induction of ferritin.
On the other hand, desferal, the chelator of free iron, inhibited ferritin syn-
thesis without affecting syntheses of heme oxygenase, thus suggesting that
free chelatable iron is the dominant factor in ferritin synthesis. This was
confirmed with free iron from added FEPIH, which selectively stimulated
ferritin synthesis but not heme oxygenase. Finally, addition to the cultured
cells of aminolevulinic acid (ALA), a regulator of the rate of heme synthesis,
increased the demands for iron by the heme synthesis pathway and thus
reduced the amount available for ferritin synthesis. These responses can be
prevented by addition to the medium of succinyl acetone (SA), an inhibitor
of ALA utilization (Table 1). Altogether these experiments show that ferritin
synthesis is regulated by chelatable free iron and not by hemin, making the
free cytosolic iron the preferred regulator of the IRE-BP.
Finally, optimum structure of the IRE for interaction with the IRE-BP
will be explored. Using the most recent Zuketl 1 computer-generated program
Munro eta!. 537
I
I
for predicting suboptional folding of RNA (Figure 7), we 42 have analyzed the
stem-loop structure of the IRE of the rat L subunit and have compared it with
the cleavage sites for single-stranded areas of the IRE using enzymes T-1 and
S-1, represented in the Figure 7 by dotted line arrows on the six-membered
loop and also the major bulge of three unpaired nucleotides (38 to 41) on one
side of the stem. Similarly, scission of double-stranded areas was accom-
plished by using enzyme V-1 and is represented by solid arrows. Wang et
al. 43 explored the structure of the bullfrog H subunit using the chemical
nuclease l, 10-phenanthroline-copper which cleaves single-stranded and bulged
areas. This supplemented the evidence from their application of T-1 while
V-1 was applied for identification of the double-stranded areas. In both of
these studies the predicted stem-loop structure has been confirmed.
In addition to studies of secondary structure described above, there have
been several reports of nucleotide replacement or deletion and the effect of
this manoever on IRE function. One example comes from a comparison of
the IRE of the Land the H subunits of mammals. As shown earlier, 14 all H
subunit IREs of mammals so far examined have a cytidylic acid replacing
the uridy lie acid of the L subunit IREs in two places (one in the sixth position
of the CAGUGU loop) without apparent Joss of function. Barton et al. 35
replaced two nucleotides in the loop without greatly altering the affinity of
the IRE to bind the specific protein as determined by in vitro competition
and saturation assays. Similarly, exchange of adenylic acid for cytidylic acid
in the unpaired bulge also left protein binding unaffected. In contrast, Hentze
et al. 45 deleted a cytidylic acid from the loop and from the stem of the human
H-IRE. The result with each deletion was Joss of response to iron, which
may be attributed to the alteration in secondary structure of the IRE rather
than to Joss of a specific nucleotide. An even more extensive loss of binding
occurred when one leg of the stem of rat L-mRNA was replaced by other
nucleotides that did not pair with the other leg of the stem, but was restored
538 Nutrition and Gene Expression
by inserting a new leg of nucleotides compatible with the other new leg. 42
Thus one stem can be replaced with another of a different nucleotide com-
position and can function so long as the nucleotides in both legs are able to
pair. This leads to the conclusion that the secondary conformation of the IRE
stem is more important than the primary sequence of its nucleotides so long
as they form a structure of paired nucleotides of adequate length. 35 This is
also the conclusion drawn by Leibold et a!. 46 who applied competition assays
to obtain the 50% binding affinity between ferritin native IRE and mutant
IRE. These conclusions require testing in vivo.
B. TRANSCRIPTION
I. Response of H and L Genes to Iron
The regulatory stem-loop motif in the 5' UTR of both the L- and H-
mRNAs of ferritin implies that activation of translation of these two subunits
will create new ferritin molecules with H and L subunits in proportion to the
dormant H-and L-mRNAs. However, in an experiment in which new ferritin
subunits in rat liver were labeled with 14C-leucine, we 9 •55 obtained a ratio of
new L to H subunits which increased substantially after an intraperitoneal
dose of iron dextran, implying either preferential synthesis of L-mRNA or
decreased rate of breakdown of L-mRNA. Using HeLa cells in culture, Cairo
et al. 56 found that transcription of L-mRNA increased after adding more iron
to the medium, but for technical reasons were unable to monitor the response
of H-mRNA. Using transcription run-off assays for measuring Land H mRNA
synthesis, we 57 were able to show a preferential increase in the rate of tran-
scription of L compared with H-mRNA by rat liver nuclei. The pattern of
increased abundance of labeled L-mRNA in the nuclei then appeared in the
ribonucleoprotein pool (Figure 2) and later in the polyribosomal fraction of
the cell. This resulted in production of isoferritins richer in the L subunit,
and therefore better able to store iron. 55 The different effects of iron on
transcription and on translation thus allow flexibility in the creation of ferritin
spheres with various LIH ratios.
IV. CONCLUSION
REFERENCES
I. Wixom, R. L., Prutkin, L., and Munro, H. N., Hemosiderin: nature, formation and
significance, in International Review Experimental Pathology. Richter, G. W. and Ep-
stein, M.A., Eds., Academic Press, New York, 1980, 22, 193.
2. Lautberger, M. V., Sur Ia cristallisation de Ia ferritine, Bull. Soc. Chim. Bioi., 19,
1575, 1937.
3. Harrison, P. M., Andrews, S. C., Artymiuk, P. J., Ford, G. C., Lawson, D. M.,
Smith, J. M.A., Treffry, A., and White, J. L., Ferritin, in Iron Transport and Storage,
Ponka, P., Schulman, H. M., and Woodworth, R. C., Eds., CRC Press, Boca Raton,
FL, 1990, 81.
4. Linder, M. C., Moor, J. 8., and Munro, H. N., Subunit heterogeneity in rat liver
apoferritin: differential responses of the subunits to iron administration, J. Bioi. Chem.,
249, 7707, 1974.
5. Adelman, T. G., Arosio, P., and Drysdale, J. W., Multiple subunits in human ferritin:
evidence for hybrid molecules, Biochem. Biophys. Res. Commun., 63, 1056, 1975.
6. Drysdale, J. W. and Munro, H. M., Regulation of synthesis and turnover of ferritin
in rat liver, J. Bioi. Chem., 241, 3630, 1966.
542 Nutrition and Gene Expression
7. Zahringer, J., Baliga, B. S., and Munro, H. N., Novel mechanism for translational
control in regulation of ferritin synthesis by iron, Proc. Nat/. Acad. Sci. U.S.A., 73,
857, 1976.
8. Munro, H. N. and Linder, M. C., Ferritin: structure, biosynthesis and role in iron
metabolism, Physiol. Rev., 58, 317, 1978.
9. Munro, H. N., Leibold, E. A., Aziz, N., Murray, M. T., White, K., and Rogers,
J., Ferritin gene structure and expression, in Iron Transport and Storage, Ponka, P.,
Schulman, H. M., and Woodworth, R. C., Eds., CRC Press, Boca Raton, FL, 1990,
134.
10. Beaumont, C., Dugast, I., Renaudie, F., Souroujun, M., and Grandchamp, B.,
Transcriptional regulation of ferritin H and L subunits in adult erythroid and liver cells
from the mouse, J. Bioi. Chern., 264, 7498, 1989.
II. Dickey, L. F., Sreedharan, S., Theil, E. C., Didsbury, J. R., Wang, Y-H., and
Kaufman, R. E., Differences in the regulation of messenger RNA for housekeeping and
specialized cell ferritin: a comparison of three different ferritin complementary DNAs,
the corresponding subunits and identification of the first processed pseudogenes in am-
phibia, J. Bioi. Chern., 262, 7901, 1987.
12. Von Dar!, M. and Bottke, W., Vollstandige Basensequenz des langsten Somaferritin-
clons: Aminosauresequenz des Somaferritins. EMBL-accession Nr X 56778, 1991.
13. Chou, P. Y. and Fasman, G. D., Empirical predictions of protein conformation, Annu.
Rev. Biochem., 47, 251, 1978.
14. Eisenstein, R. S., Bettany, A. J. E., and Munro, H. N., Regulation of ferritin gene
expression, in Advances in Inorganic Biochemistry, Eichhorn, G. L. and Marzilli, L. G.,
Eds., Elsevier, New York, 1990, 8, 91.
IS. Leibold, E. A. and Munro, H. N., Characterization and evolution of the expressed rat
ferritin light subunit gene and its pseudogene family, J. Bioi. Chern., 262, 7335, 1987.
16. Murray, M. T., White, K., and Munro, H. N., Conservation of ferritin heavy subunit
gene structure: implications for the regulation of ferritin gene expression, Proc. Nat/.
Acad. Sci. U.S.A., 84, 7438, 1987.
17. Aziz, N. and Munro, H. N., Both subunits of rat liver ferritin are regulated at a
translational level by iron induction, Nucleic Acids Res., 14, 915, 1986.
18. Dougherty, J. J., Croft, W. A., and Hoekstra, W. G., Effects of ferrous chloride and
iron dextran on lipid peroxidation in vivo in vitamin E and selenium adequate and deficient
rats, J. Nutr., Ill, 1784, 1981.
19. Floyd, R. A., Role of oxygen free radicals in carcinogenesis and brain ischemia, FASEB
J., 4, 258, 1990.
20. Munro, H. N., Leibold, E. A., Vass, J. K., Aziz, N., Rogers, J., Murray, M., and
White, K., Ferritin gene structure and expression, in Proteins of Iron Storage and
Transport, Spik, G., Montreuil, J., Crichton, R. R., and Mazurier, J., Eds., Elsevier,
Amsterdam, 1985, 331.
21. Aziz, N. and Munro, H. N., Iron regulates ferritin mRNA translation through a segment
of its 5' untranslated region, Proc. Nat/. Acad. Sci. U.S.A., 84, 8478, 1987.
22. Dickey, L. F., Wang, Y-H, Schull, G. E., Wortman, I. A., and Theil, E. C., The
importance of the 3' untranslated region in the translational control of ferritin mRNA,
J. Bioi. Chern., 263, 3071, 1988.
23. Caughman, S. W., Hentze, M. W., Rouault, T. A., Harford, J. B., and Klausner,
R. D., The iron-responsive element is the single element responsible for iron-dependent
translational regulation of ferritin biosynthesis: evidence for function as the binding site
for a translational repressor, J. Bioi. Chern., 263, 19048, 1988.
24. Goessen, B., Caughman, S. W., Harford, J. B., Klausner, R. D., and Hentze, M. W.,
Translational repression by a complex between the iron-responsive element of ferritin
mRNA and its specific cytoplasmic binding protein is position-dependent in vivo, EMBO
J., 9, 4127, 1990.
Munro et al. 543
25. Leibold, E. A. and Munro, H. N., Cytoplasmic protein binds in vitro to a highly
conserved sequence of the 5' untranslated region of ferritin heavy and light subunit
mRNAs, Proc. Nat/. Acad. Sci. U.S.A., 85, 2171, 1988.
26. Rouault, T. A., Hentze, M. W., Caughman, S. W., Harford, J, B., and Klausner,
R. D., Binding of a cytosolic protein to the iron responsive element of human ferritin
messenger RNA, Science, 241, 1207, 1988.
27. Rothenberger, S., Mollner, E. W., and Kuhn, L. C., The mRNA-binding protein
which controls ferritin and transferrin receptor expression is conserved during evolution,
Nucleic Acids Res., 18, 1175, 1990.
28. Walden, W. E., Patino, M. M., and Gaffield, L., Purification of a specific repressor
of ferritin mRNA translation from rabbit liver, J. Bioi. Chern., 264, 13765, 1989.
29. Rouault, T. A., Hentze, M. W., Haile, D. J., Harford, J, B., and Klausner, R. D.,
The iron-responsive element binding protein: a method for the affinity purification of a
regulatory RNA-binding protein, Proc. Nat/. Acad. Sci. U.S.A., 86, 5768, 1989.
30. Rouault, T. A., Tang, C. K., Kaptain, S., Burgess, W. H., Haile, D. J., Samaniego,
F., McBride, 0. W., Harford, J, B., and Klausner, R. D., Cloning of the eDNA
encoding an RNA-regulatory protein: the human iron-response element binding protein,
Proc. Nat/. Acad. Sci. U.S.A., 87, 7958, 1990.
31. Bomford, A. B., Barton, H. A., Pettingell, W. H., Munro, H. N., and Eisenstein,
R. S., The iron-responsive element-binding protein of the rat: biochemical and functional
studies of the purified protein. (Submitted).
32. Rouault, T. A., Stout, C. D., Kaptain, S., Harford, J, B., and Klansman, R. D.,
Structural relationship between an iron-regulated RNA-binding protein (IRE-BP) and
aconitase: functional implications, Cell, 64, 881, 1991.
33. Kaptain, S., Downey, W. E., Tang, C., Philpott, C., Haile, D., Orloff, D. G.,
Harford, J, B., Rouault, T. A., and Klausner, R. D., A regulated RNA binding protein
also possesses aconitase activity, Proc. Nat/. Acad. Sci. U.S.A., 88, 10109, 1991.
34. Hentze, M. W., Rouault, T. A., Harford, J, B., and Klausner, R. D., Oxidation-
reduction and the molecular mechanism of a regulatory RNA-protein interaction, Science,
244, 357, 1989.
35. Barton, H. A., Eisenstein, R. S., Bomford, A., and Munro, H. N., Determinants of
the interaction between the iron-responsive element-binding protein and its binding site
in rat L-ferritin mRNA, J. Bioi. Chern., 265, 7000, 1990.
36. Lin, J. J., Daniels-McQueen, S., Patino, M. M., Gaffield, L., Walden, W. E., and
Thach, R. E., Derepression of ferritin messenger RNA translation by hemin in vitro,
Science, 247, 74, 1990.
37. Lin, J. J., Patino, M. M., Garfield, L., Walden, W. E., Smith, A., and Thach,
R. E., Cross-linking of hemin to a specific site on the 90-kd repressor protein, Proc.
Nat/. Acad. Sci. U.S.A., 88, 6068, 1991.
38. Haile, D. J., Rouault, T. A., Harford, J. B., and Klausner, R. D., The inhibition of
the iron responsive element RNA-protein interaction by heme does not mimic in vivo
iron regulation, J. Bioi. Chern., 265, 12786, 1990.
39. Rogers, J, and Munro, H. N., Translation of light and heavy subunit mRNAs is regulated
by intracellular chelatable iron levels in the rat hepatoma cells, Proc. Nat/. Acad. Sci.
U.S.A., 84, 2277, 1987.
40. Eisenstein, R. S., Garcia-Mayol, D., Pettingell, W., and Munro, H. N., Regulation
of ferritin and heme oxygenase synthesis in rat fibroblasts by different forms of iron,
Proc. Nat/. Acad. Sci. U.S.A., 88, 688. 1991.
41. Jaeger, J, A., Turner, D. H. and Zuker, M., Predicting optimal and suboptimal
secondary structure for RNA, Methods Enzyrnol., 183, 281, 1990.
42. Bettany, A. J. E., Eisenstein, R. S., and Munro, H. N., Role of RNA secondary
structure in the interaction between the iron regulatory element of ferritin mRNA and its
binding protein, in press.
544 Nutrition and Gene Expression
43. Wang, Y-H, Sczekan, S. R., and Theil, E. C., Structure of the 5' untranslated regulatory
region of ferritin mRNA studied in solution, Nucleic Acids Res., 18, 4463, 1990.
44. Harrell, C. M., McKenzie, A. R., Patino, M. M., Walden, W. E., and Theil, E. C.,
Ferritin mRNA: interactions of iron regulatory element with translational regulator protein
P-90 and the effect on base-paired flanking regions, Proc. Nat/. Acad. Sci. U.S.A., 88,
4166. 1991.
45. Hentze, M. W., Caughman, S. W., Casey, J. L., Koeller, D. M., Rouault, T. A.,
Harford, J. B., and Klausner, R. D., A model for the structure and functions of iron-
responsive elements, Gene, 72, 201. 1988.
46. Leibold, E. A., Laudano, A., and Yang, Y., Structural requirements of iron responsive
elements for binding of the protein involved in both transferrin receptor and ferritin mRNA
post-transcriptional regulation, Nucleic Acids Res., 18, 1819, 1990.
47. Mullner, E. W. and Kuhn, L. C., A stem-loop in the 3' untrans1ated region mediates
iron-dependent regulation of transferrin receptor mRNA stability in the cytoplasm, Cell,
53, 815, 1988.
48. Mullner, E. W., Neupert, B., and Kuhn, L. C., A specific mRNA binding factor
regulates the iron-dependent stability of cytoplasmic transferrin receptor mRNA, Cell,
58, 373, 1989.
49. Casey, J. L., Hentze, M. W., Koeller, D. M., Caughman, S. W., Rouault, T. A.,
Klausner, R. D., and Harford, J. B., Iron-responsive elements: regulatory RNA se-
quences that control mRNA levels and translation, Science, 240, 924, 1988.
50. Casey, J. L., Koeller, D. M., Ramin, V. C., Klausner, R. D., and Harford, J. B.,
Iron regulation of transferrin receptor mRNA levels requires iron-responsive elements
and a rapid turnover determinant in the 3' untranslated region of the mRNA, EMBO J.,
8, 3693, 1989.
51. Koeller, D. M., Casey, J. L., Hentze, M. W., Gerhardt, E. M., Chan, L-N. L.,
Klausner, R. D., and Harford, J. B., A cytosolic protein binds to structural elements
within the iron regulatory region of the transferrin receptor mRNA, Proc. Nat/. Acad.
Sci. U.S.A., 86, 3574, 1989.
52. Cox, T. C., Bawden, M. J., Martin, A., and May, B. K., Human erythroid 5-
aminolevulinate synthase: promoter analysis and identification of an iron-responsive ele-
ment in the mRNA, EMBO J., 10, 1891, 1991.
53. Dandekar, T., Stripecke, R., Gray, N. K., Goosen, B., Constable, A., Johansson,
H., and Hentze, M., Identification of a novel iron-responsive element in murine and
human 5-erythroid amino levulinic acid synthase mRNA, EMBO J., 10, 1903, 1991.
54. Tanzi, R. E. and Hyman, B. T., Alzheimer's mutation, Nature (London), 350, 564,
1991.
55. Bomford, A., Conlon-Hollingshead, C., and Munro, H., Adaptive responses of rat
tissue isoferritins to iron administration, J. Bioi. Chern., 256, 948, 1981.
56. Cairo, G., Bardella, L., Schiaffonati, L., Arosio, P., Levi, S., and Barnelli-Zazzera,
A., Multiple mechanisms of iron-induced ferritin synthesis in HeLa cells, Biochern.
Biophys. Res. Cornrnun., 133, 314, 1985.
57. White, K. and Munro, H. N., Induction of ferritin subunit synthesis by iron is regulated
at both the transcriptional and translational levels, J. Bioi. Chern., 263, 8938, 1988.
58. Konijn, A.M. and Hershko, C., Ferritin synthesis in inflammation. I. Pathogenesis of
impaired iron release, Br. J. Haernatol., 37, 7, 1977.
59. Campbell, C. H., Solgonick, R., and Linder, M. C., Translational regulation of ferritin
synthesis in rat spleen: effects of iron and inflammation, Biochern. Biophys. Res. Corn-
rnun., 160, 453, 1989.
60. Dinarello, C. A., Biology of interleukin-1, FASEB J., 2, 108, 1988.
61. Rogers, J. T., Bridges, K. R., Durmowicz, G. P., Glass, J., Auron, P. E., and
Munro, H. N., Translational control during the acute phase response; ferritin synthesis
in response to interleukin-1, J. Bioi. Chern., 265, 14572, 1990.
Munro et al. 545
62. Wei, Y., Miller, S. C., Tsuji, Y., Torti, S. V., and Torti, F. M., Interleukin 1 induces
ferritin heavy chain in human muscle cells, Biochern. Biophys. Res. Cornrnun .. 169, 289.
1990.
63. Miller, L. L., Miller, S.C., Torti, S. V., Tsuji, Y., and Torti, F. M., Iron-independent
induction of ferritin H chain by tumor necrosis factor, Proc. Nat/. Acad. Sci. U.S.A..
88. 4946. 1991.
64. Tsuji, Y., Miller, L. L., Miller, S.C., Torti, S. V., and Torti, F. M., Tumor necrosis
factor-alpha and interleukin !-alpha regulate transferrin receptor in human diploid fibro-
blasts; relationship to the induction of ferritin heavy chain, J. Bioi. Chern .. 266, 7257.
1991.
65. Chou, C-C., Gatti, R. A., Fuller, M. L., Concannon, P., Wong, A., Chada, S.,
Davis, R. C., and Salser, W. A., Structure and expression of ferritin genes in a human
promyelocyte cell line that differentiates in vitro. Mol. Cell. Bioi.. 6. 566, 1986.
66. Testa, U., Kuehn, L., Petrini, M., Quaranta, M. T., Pelosi, E., and Peschle, C.,
Differential regulation of iron regulatory element-binding protein(s) in cell extracts of
activated lymphocytes versus monocytes-macrophages, J. Bioi. Chern., 266, 13925, 1991.
67. Liau, G., Chan, L. M., and Feng, P., Increased ferritin gene expression is both promoted
by cAMP and a marker of growth arrest in rabbit vascular smooth muscle cells, J. Bioi.
Chern., 266, 18819, 1991.
Chapter 24
TABLE OF CONTENTS
0-8493-6961-4/93/$0.00 + $.50
© 1993 by CRC Press. Inc. 547
548 Nutrition and Gene Expression
I. INTRODUCTION
minal and the ligand binding domains of the receptors. 3 •35 This highly con-
served region consists of 66 amino acids which permit the receptor molecules
to recognize and bind to specific DNA regulatory sequences, the steroid
response elements, located within or near to the promoter regions of steroid
responsive genes. Contained within this DNA binding domain are nine highly
conserved cysteine residues, eight of which are thought to form two zinc
fingers, with each finger coordinately binding one zinc atom. 36 It is believed
that one zinc finger determines target gene specificity by distinguishing be-
tween different steroid response elements, while the other zinc finger contacts
the sugar-phosphate backbone of the DNA helix to stabilize binding of the
receptor to the steroid response element. 37 -40
It is remarkable that despite such a high degree of conservation of amino
acid sequence and protein structure, different steroid receptors remain capable
of distinguishing between different hormonally regulated genes, activating
transcription only of the appropriate target genes in the appropriate cell or
tissue type. It is possible that other regions of the receptors may participate
in determining target gene specificity. Alternatively, cell- or tissue-specific
factors may participate in regulating receptor function, perhaps through direct
interaction with the receptor molecules. Although only a speculative notion,
the NH 2 -terminal region, which exhibits the most variability among different
receptors, may be responsible for promoting such receptor-specific functions.
This could explain variations in the cell- or tissue-specific expression of many
hormonally regulated genes. For example, expression of the tyrosine ami-
notransferase gene is selectively induced in parenchymal cells of liver, but
not in other tissues, following glucocorticoid treatment, 41 One could speculate
that a liver-specific factor may interact with the NH 2 -terminal portion of the
glucocorticoid receptor to allow glucocorticoid-specific induction of the ty-
rosine aminotransferase gene in the liver, while absence of an analogous
factor in other cell types would inhibit glucocorticoid induction of tyrosine
aminotransferase gene expression in nontarget tissues. Alternative explana-
tions may also apply, however, and in the case of the tyrosine aminotransferase
gene it has been reported that tissue specific methylation of CpG dinucleotides
in certain regions of DNA may inhibit expression of tyrosine aminotransferase
gene in nonexpressing cells. 42 Theories such as these are currently being
investigated in an effort to determine how specific gene expression is mediated
in the presence of similar steroid receptors.
Nonetheless, given the extensive sequence and structural homology that
exists among the different steroid hormone receptors, it is not surprising to
learn that the receptors all act through essentially the same pathway to exert
their diverse biological functions. Each of the steroid hormone receptors is
believed to occur in an oligomeric complex containing two molecules of heat
shock protein 90 and perhaps a smaller heat shock protein or other protein
in addition to receptor. 43 •44 While the receptor is associated with this oligo-
meric complex, it is maintained in an unactivated state, which is operationally
Tully eta!. 551
cpm
!
~
500
CD
E
~ 400
~
300
20
100
interact directly with the glucocorticoid receptor through Schiff's base for-
mation, to alter its sedimentation profile. Other lysine-reactive compounds,
including dinitrofluorobenzene, picryl sulfonic acid, succinate, or p-nitro-
phenol acetate, had no effect on receptor sedimentation profiles. 94 Thus, the
glucocorticoid receptor exhibits an alteration in conformation or subunit com-
position as a result of exposure specifically to the physiologically active form
of vitamin 8 6 , pyridoxal phosphate. The requirement for the carboxaldehyde
moiety of pyridoxal phosphate further supports the notion of a direct inter-
action between the receptor and the vitamin. Pyridoxal phosphate treatment
has also been shown to have similar effects on other steroid receptors. Both
progesterone86 and estrogen receptors 87 exhibit a reduction in sedimentation
coefficient on sucrose density gradients following treatment with pyridoxal
phosphate. These observations suggest that pyridoxal phosphate may act through
a single, common mechanism to influence multiple members of the steroid
hormone receptor family of proteins. However, it still remained unclear whether
the vitamin was acting directly on the receptor or on another protein in the
oligomeric complex.
UNACTIVATED RECEPTORS
• TRYPSIN • NABH 4
• TRYPSIN
o PPAL. NABH4• TRYPSIN
ov MY
vv
'
0 20 40 60 80 100 120
FRACTION
FIGURE 2. The influence of trypsin on the metabolism of rat thymocyte glucocorticoid re-
ceptor. Cytosols from thymocytes incubated with ['H] dexamethasone were treated with Wor-
thington trypsin 10 f.l-g/ml for 30 min at 4cC. The trypsin reaction was stopped by the addition
of I mg of soybean trypsin inhibitor (Sigma), and samples were treated with dextran-coated
charcoal as described under Methods prior to chromatography on Sephacryl. The dosed circles
were from untreated (unactivated) cytosol, and the open triangles were from samples to which
a small amount of solid sodium borohydride was added prior to trypsin. The open circles were
derived from samples of cytosol treated with 5 mM pyridoxal phosphate for 3 h at 0 to 4°C,
reduced by the addition of solid sodium borohydride, and then trypsin treated. The patterns are
representative of three similar experiments. (From Cidlowski. J. A., Biochemistry, 19, 6162,
1980. With permission.)
Tully et al. 557
3.0
~~ 2.0
5!!.
<';
1.0
>-
I- 2.0 2.0
>
I-
u<{
I- 1.0 1.0
<{
u
sequences derived from the mouse mammary tumor virus promoter, and that
variation of intracellular pyridoxal phosphate concentration also affects tran-
scription mediated through other hormonally regulated promoters.
VII. SUMMARY
These studies on transcriptional activation by the glucocorticoid, andro-
gen, progesterone, and estrogen receptors provide the most direct evidence
to date that pyridoxal phosphate, the active form of vitamin B6 , acts not only
to affect some physical properties of steroid hormone receptors, but also
influences the physiological function of these molecules. The demonstration
that alterations in intracellular pyridoxal phosphate concentration have dra-
matic effects on hormonally regulated target gene expression suggests that
vitamin B6 may act as a physiological regulator of steroid hormone action.
Despite the efforts of many laboratories, surprisingly little is known about
how steroid hormone action may be modulated in different tissues and cell
types. Although the specific mechanisms through which receptor-mediated
562 Nutrition and Gene Expression
HeLo CELLS
2.0 r
>- ..=-
1-
>
i=
r-
u 1.0 r-.--
<{
1-
<{
u
-
CON PYR4DXY
pERENF1CAT
+
pH EO
ESTROGEN
RECEPTOR
FIGURE 5. Estrogen receptor-mediated gene expression is modulated by alterations in intra-
cellular pyridoxal phosphate concentration. HeLa cells were transfected with the estrogen receptor
expression vector pHEO and the estrogen-regulated CAT reporter pERENFICAT. Cells were
exposed to control medium (CON) 16 h after transfection medium. medium supplemented with
I mM pyridoxine (PYR), or medium supplemented with 5 mM 4-deoxypyridoxine (4DXY) prior
to 24-h treatment with 5 nM 17 beta-estradiol or vehicle and CAT activity determination. The
data shown reflect results from at least three individual transfection experiments. (From Allgood,
V. E. and Cidlowski, J. A., J. Bioi. Chern., 267. 3819, 1992. With permission.)
gene expression is induced have been much more clearly identified, much
less is known about the mechanisms cells may employ to regulate the mag-
nitude of their response to steroid hormones. The observations which have
been described here are consistent with the hypothesis that pyridoxal phosphate
interacts with the steroid hormone receptors, and, in so doing, alters both
physical properties of the receptors and their transcriptional activation ca-
pacity. Thus by regulating the intracellular concentration of pyridoxal phos-
phate, differentiated cell types may be able to modify the magnitude of their
response to steroid hormone stimulation. Identification of a site on the steroid
receptors with which pyridoxal phosphate interacts will facilitate proof of this
hypothesis. However, it is clear that, regardless of mechanism, vitamin B6
acts to modulate the capacity of many different steroid hormone receptors to
induce target gene expression. Thus, vitamin B6 has an important role in the
regulation of steroid hormone action.
Tully et al. 563
REFERENCES
I. Beato, M., Transcriptional control by nuclear receptors. FASEB J., 5, 2044, 1991.
2. Munck, A., Mendel, D. B., Smith, L. I., and Orti, E., Glucocorticoid receptors and
actions, Am. Rev. Respir. Dis., 141, S2, 1990.
3. Evans, R. M., The steroid and thyroid hormone receptor superfamily, Science, 240,
889, 1988.
4. Vanderbilt, J. N., Miesfeld, R., Maler, B. A., and Yamamoto, K. R., Intracellular
receptor concentration limits glucocorticoid-dependent enhancer activity, Mol. Endocri-
nol .. 1, 68, 1987.
5. Gehring, U., Mugele, K., and Ulrich, Cellular receptor levels and glucocorticoid re-
sponsiveness of lymphoma cells, J. Mol. Cell. Endocrinol .. 36, 107. 1984.
6. Ballard, P. L., Glucocorticoids and differentiation, in Glucocorticoid Hormone Action.
Monographs in Endocrinology, Vol. 12. Baxter, J. D. and Rousseau, G. G., Eds.,
Springer-Verlag, Berlin, 1979, 493.
7. Kalinyak, J. E., Griffin, C. A., Hamilton, R. W., Bradshaw, J. G., Perlman, A. J.,
and Hoffman, A. R., Developmental and hormonal regulation of glucocorticoid receptor
messenger RNA in the rat, J. Clin. Invest., 84. 1843, 1989.
8. Littlefield, B. A., Cidlowski, N. B., and Cidlowski, J. A., Modulation of glucocorticoid
effects and steroid receptor binding in butyrate-treated HeLa S 3 cells, Arch. Biochem.
Biophys., 201, 174, 1980.
9. Littlefield, B. A. and Cidlowski, J. A., Increased steroid responsiveness during sodium
butyrate-induced "differentiation" of HeLa S, cells, Endocrinology, 114, 566, 1984.
10. Cidlowski, J. A. and Michaels, G. A., Alteration in glucocorticoid binding site number
during the cell cycle in HeLa cells, Nature (London), 266, 643, 1977.
11. Tomkins, G. M., Gelehrter, T., Granner, D., Martin, D., Samuels, H. H., and
Thompson, E. B., Control of gene expression in higher organisms, Science, 166, 1474,
1969.
12. Griffin, M. J. and Ber, R., Cell cycle events in the hydrocortisone regulation of alkaline
phosphatase in HeLa S, cells, J. Cell Bioi., 40, 297, 1969.
13. Allgood, V. E. and Cidlowski, J. A., Novel role for vitamin B6 in steroid hormone
action: a link between nutrition and the endocrine system, J. Nutr. Biochem., 2, 641,
1991.
14. Allgood, V. E., Powell-Oliver, F. E., and Cidlowski, J. A., Influence of vitamin B6
on the structure and function of the glucocorticoid receptor, Ann. N.Y. Acad. Sci., 585,
452, 1990.
15. Compton, M. M. and Cidlowski, J. A., Vitamin B6 and glucocorticoid action, Endocrine
Rev., 7, 140, 1986.
16. DiSorbo, D. M. and Litwack, G., Changes in the intracellular levels of pyridoxal 5'-
phosphate affect the induction of tyrosine aminotransferase by glucocorticoids, Biochem.
Biophys. Res. Commun., 99, 1203, 1981.
17. Valeriote, F. A., Auricchio, F., Tomkins, G. M., and Riley, D., Purification and
properties of rat tyrosine aminotransferase, J. Bioi. Chern., 244, 3618, 1969.
18. Allgood, V. E., Powell-Oliver, F. E., and Cidlowski, J. A., Vitamin B6 influences
glucocorticoid receptor-mediated gene expression, J. Bioi. Chern., 265, 12424, 1990.
19. Beato, M., Gene regulation by steroid hormones, Cell, 56, 355, 1989.
20. O'Malley, B. W., The steroid receptor superfamily: more excitement predicted for the
future, Mol. Endocrino/., 4, 363, 1990.
21. Reichman, M. E., Foster, C. M., Eisen, L. P., Torain, B. F., and Simons, S. S.,
Limited proteolysis of covalently labeled glucocorticoid receptors as a probe of receptor
structure, Biochemistry, 23, 5376, 1984.
22. Sherman, M. R., Moran, M. C., Tuazon, F. B., and Stevens, Y.-E., Structure,
dissociation, and proteolysis of mammalian steroid receptors, J. Bioi. Chern., 258, 10366,
1983.
564 Nutrition and Gene Expression
43. Mendel, D. B. and Orti, E., Isoform composition and stoichiometry of the -90-kDa
heat shock protein associated with glucocorticoid receptors, J. Bioi. Chern., 263, 6695,
1988.
44. Denis, M., Wikstrom, A.-C., Gustafsson, J.-A., the molybdate-stabilized nonactivated
glucocorticoid receptor contains a dimer of Mr 90,000 non-hormone-binding protein, J.
Bioi. Chern., 262, 11803, 1987.
45. Currie, R. A. and Cid1owski, J. A., Physicochemical properties of the cytoplasmic
glucocorticoid receptor complex in HeLa S3 cells, J. Steroid Biochem., 16, 419, 1982.
46. Holbrook, N. J., Bodwell, J, E., Jeffries, M., and Munck, A., Characterization of
nonactivated and activated glucocorticoid receptor complexes from intact rat thymus cells,
J. Bioi. Chern., 258, 6477, 1983.
47. Vedeckis, W. V., Subunit dissociation as a possible mechanism of glucocorticoid receptor
activation, Biochemistry, 22, 1983, 1983.
48. Burnstein, K. and Cidlowski, J. A., Regulation of gene expression by glucocorticoids,
Annu. Rev. Physiol., 51, 683, 1989.
49. Yamamoto, K. R. and Alberts, B. M., Steroid receptors: elements for modulation of
eukaryotic transcription, Annu. Rev. Biochem., 45, 721, 1975.
50. Rousseau, G. G., Control of gene expression by glucocorticoid hormones, Biochem. J.,
224, 1, 1984.
51. Ringold, G. M., Steroid hormone regulation of gene expression, Annu. Rev. Pharm.
Toxicol., 25, 529, 1985.
52. Drouin, J., Sun, Y. L., and Nemer, M., Glucocorticoid repression of proopiomelan-
ocortin gene transcription, J. Steroid Biochem., 34, 63, 1989.
53. Mordacq, J. C. and Linzer, D. I. H., Co-localization of elements required for phorbol
ester stimulation and glucocorticoid repression of proliferin gene expression, Genes De-
velop., 3, 760, 1989.
54. Picard, D. and Yamamoto, K. R., Two signals mediate hormone-dependent nuclear
localization of the glucocorticoid receptor, EMBO J., 6, 3333, 1987.
55. Nordeen, S. K., Sub, B. J., Kuhne!, B., and Hutchison, C. A., III, Structural
determinants of a glucocorticoid receptor recognition element, Mol. Endocrinol., 4, 1866,
1990.
56. Aronica, S.M. and Katzenellenbogen, B.S., Progesterone receptor regulation in uterine
cells: stimulation by estrogen, cyclic adenosine 3' ,5' -monophosphate, and insulin-like
growth factor I and suppression by antiestrogens and protein kinase inhibitors, Endocri-
nology, 128, 2045, 1991.
57. Groul, D. J., Harrigan, M. T., and Bourgeois, S., Modulation of glucocorticoid-
induced responses by cyclic AMP in lymphoid cell lines, in Gene Regulation by Steroid
Hormones, Vol. 4, Roy, A. K. and Clark, J. H., Eds., Springer-Verlag, Berlin, 1989,
41.
58. Tryfiates, G. P., Vitamin B6 Metabolism and Role in Growth, Food and Nutrition Press,
Westport, CT, 1980, I.
59. Merrill, A. H. and Henderson, J. M., Diseases associated with deficiencies in vitamin
8 6 metabolism or utilization, Annu. Rev. Nutr., 7, 137, 1987.
60. Merrill, A. H., Henderson, J. M., Wang, E., McDonald, B. W., and Millikan,
W. J., Metabolism of vitamin 8 6 by human liver, J. Nutr., 114, 1664, 1964.
61. McCormick, D. B., Two interconnected B vitamins: riboflavin and pyridoxine, Physiol.
Rev., 69, 1170, 1989.
62. Wada, H. and Snell, E. E., The enzymatic oxidation of pyridoxine and pyridoxamine
phosphates, J. Bioi. Chern., 236, 2089, 1961.
63. Dempsey, W. B. and Christensen, H. N., The specific binding ofpyridoxal5-phosphate
to bovine plasma albumin, J. Bioi. Chern., 237, 1113, 1962.
64. Anderson, B. B., Newmark, P. A., and Rawlins, M., Plasma binding of vitamin 8 6
compounds, Nature (London), 250, 502, 1974.
566 Nutrition and Gene Expression
65. Lumeng, L. and Li, T. K., Mammalian vitamin 8 6 metabolism: regulatory role of
protein binding and the hydrolysis of pyridoxal 5 '-phosphate in storage and transport, in
Vitamin B6 Metabolism and Role in Growth, Tryfiates, G. P., Ed., Food and Nutrition
Press, Westport, CT, 1980, 27.
66. Brin, M., Vitamin B6 : Chemistry, absorption, metabolism, catabolism, and toxicity, in
Human Vitamin B6 Requirements, National Academy of Sciences, Washington, D.C.,
1976.
67. Leklem, J. E., Vitamin 8 6 , in Handbook of Vitamins, 2nd ed., Machlin, L. J., Ed.,
Marcel Dekker, New York, 1991, chap. 9.
68. Hayashi, H., Wada, H., Yoshimura, T., Esaki, N., and Soda, K., Recent topics in
pyridoxal 5' -phosphate enzyme studies, Annu. Rev. Biochem., 59, 87, 1990.
69. Sauberlich, H. E., Biochemical systems and biochemical detection of deficiency, in The
Vitamins, Sebrell, W. H. and Harris, R. S., Eds., Academic Press, New York, 1968,
31.
70. Walsh, C., Enzymatic reactions requiring pyridoxal phosphate, in Enzymatic Reaction
Mechanisms, W. H. Freeman, San Francisco, 1979, chap. 24.
71. Buffoni, F., Histamine and related amine oxidases, Pharmacal. Rev., 18, 1163, 1966.
72. Kahlson, G. and Rosengren, E., Histamine, Annu. Rev. Pharmacal., 5, 305, 1965.
73. Lovenberg, W., Weissbach, H., and Udenfriend, S., Aromatic L-amino acid decar-
boxylase, J. Bioi. Chern., 237, 89, 1962.
74. Roberts, E. and Frankel, S., Gamma-amino butyric acid in brain: its formation from
glutamic acid, J. Bioi. Chern., 187, 55, 1950.
75. Leklem, J. E., Vitamin 8 6 metabolism and function in humans, in Clinical and Phys-
iological Applications of Vitamin B6 , Leklem, J. E. and Reynolds, R. D., Eds., Alan
R. Liss, New york, 1988, 3.
76. Axelrod, A. E., Immune processes in vitamin deficiency states, Am. J. Clin. Nutr., 24,
265, 1971.
77. Chandra, R. K. and Purl, S., Vitamin 86 modulation of immune responses and in-
fection, in Vitamin 86: its role in health and disease, Reynolds, R. D. and Leklem, J.
E., Eds., Alan R. Liss, New York, 1984, 163.
78. Harutyunyan, E. G., Malashkevich, V.N., Kochkina, V.M., and Torchinsky, Y. M.,
Conformational changes in chicken heart cytosolic aspartate aminotransferase as revealed
by X-ray crystallography, in Chemical and Biological Aspects of Vitamin B6 , Evange-
lopoulos, A. E., Ed., Alan R. Liss, New York, 1984, 205.
79. Jansonius, J. N., Eichele, G., Ford, G. C., Kirsch, J. F., Picot, D., Thaller, C.,
and Vincent, M. G., Crystallographic studies on the mechanism of action of mitochon-
drial aspartate aminotansferase, in Chemical and Biological Aspects of Vitamin B6 , Evan-
gelopoulos, A. E., Ed., Alan R. Liss, New York, 1984, 195.
80. Gehring, H., Sandmeier, E., Kirsten, H., Carrassi, R., Christen, P., andJansonius,
J. N., Aspartate aminotransferase: studies on conformational adaptations and 'H-transfer
during transamination, in Chemical and Biological Aspects of Vitamin B6 Catalysis,
Evangelopoulos, A. E., Ed., Alan R. Liss, New York, 1984, 223.
81. Taylor, J. E., Metzler, D. E., and Arnone, A., Modeling inhibitors in the active site
of aspartate aminotransferase, Ann. N.Y. Acad. Sci., 585, 58, 1990.
82. Nishigori, H., Moudgil, V. K., and Toft, D., Inactivation of avian progesterone receptor
binding to ATP-Sepharose by pyridoxal 5 '-phosphate, Biochem. Biophys. Res. Commun.,
80, 112, 1978.
83. Modak, M. J., Observations on the pyridoxal 5'-phosphate inhibition of DNA poly-
merases, Biochemistry, 15, 3620, 1976.
84. Cake, M. H., DiSorbo, D. M., and Litwack, G., Effect of pyridoxal phosphate on
the DNA binding site of activated hepatic glucocorticoid receptor, J. Bioi. Chern., 253,
4886, 1978.
85. Cidlowski, J. A. and Thanassi, J. W., Extraction of nuclear glucocorticoid receptor
complexes with pyridoxal phosphate, Biochem. Biophys. Res. Commun., 82, 1140, 1978.
Tully et al. 567
86. Nishigori, H. and Toft, D., Chemical modification of the avian progesterone receptor
by pyridoxal 5'-phosphate. J. Bioi. Chern, 254. 9155, 1979.
87. Muldoon, T. G. and Cidlowski, J. A., Specific modification of rat uterine estrogen
receptor by pyridoxal 5'-phosphate, J. Bioi. Chem., 255, 3100, 1980.
88. Hiipakka, R. A. and Liao, S., Effect of pyridoxal phosphate on the androgen receptor
from rat prostate: inhibition of receptor aggregation and receptor binding to nuclei and
to DNA-cellulose, J. Steroid Biochem.. 13. 841. 1980.
89. Silva, C. M., Tully, D. B., Petch, L. A., Jewell, C. M., and Cidlowski, J, A.,
Application of a protein-blotting procedure to the study of human glucocorticoid receptor
interactions with DNA, Proc. Nat/. Acad. Sci. U.S.A., 84. 1744, 1987.
90. Holley, J,, Bender, D. A., Coulson, W. F., and Symes, E. K., Effects of vitamin B6
nutritional status on the uptake of ['H)-oestradiol into the uterus, liver, and hypothalamus
of the rat, J. Steroid Biochem.. 18. 161, 1983.
91. Bowden, J. F., Bender, D. A., Coulson, W. F., and Symes, E. K., Increased uterine
uptake and nuclear retention of ['H] oestradiol through the oestrous cycle and enhanced
end-organ sensitivity to oestrogen stimulation in vitamin 8 6 deficient rats, J. Steroid
Biochem., 25, 359, 1986.
92. Bunce, G. E. and Vessal, M., Effect of zinc and/or pyridoxine deficiency upon oestrogen
retention and oestrogen receptor distribution in the rat uterus, J. Steroid Biochem., 26,
303, 1987.
93. Symes, E. K., Bender, D. A., Bowden, J. F., and Coulson, W. F., Increased target
tissue uptake of, and sensitivity to, testosterone in the vitamin 8 6 deficient rat, J. Steroid
Biochem., 20, 1089, 1984.
94. O'Brien, J. M. and Cidlowski, j. A., Interaction of pyridoxal phosphate with gluco-
corticoid receptors from HeLa S, cells. J. Steroid Biochem., 14, 9. 1981.
95. Viceps-Madore, D., Cidlowski, J. A., Kittler, j. M., and Thanassi. J. W., Prepa-
ration, characterization, and use of monoclonal antibodies to vitamin 8 6 • J. Bioi. Chern.,
258, 2689, 1983.
96. Chandler, V. L., Maler, B. A., and Yamamoto, K. R., DNA sequences bound spe-
cifically by glucocorticoid receptor in vitro render a heterologous promoter hormone
responsive in vivo, Cell, 33, 489. 1983.
97. Cato, A. C. B., Skroch, P., Weinmann, J., Butkeraitis, P., and Ponta, H., DNA
sequences outside the receptor-binding sites differently modulate the responsiveness of
the mouse manunary tumour virus promoter to various steroid hormones, EMBO J., 7,
1403, 1988.
98. Ham, J., Thomson, A., Needham, M., Webb, P., and Parker, M., Characterization
of response elements for androgens, glucocorticoids and progestins in mouse mammary
tumour virus, Nucleic Acids Res., 16, 5263, 1988.
99. Allgood, V. E. and Cidlowski, J. A., Vitamin 86 modulates transcriptional activation
by multiple members of the steroid hormone receptor superfamily. J. Bioi. Chern., in
press.
569
INDEX