Personalized Nutrition
Personalized Nutrition
Personalized Nutrition
PERSONALIZED
NUTRITION
Principles and Applications
Edited by
Frans Kok
Laura Bouwman
Frank Desiere
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
2008 by Taylor & Francis Group, LLC
CRC Press is an imprint of Taylor & Francis Group, an Informa business
This book contains information obtained from authentic and highly regarded sources. Reprinted
material is quoted with permission, and sources are indicated. A wide variety of references are
listed. Reasonable eorts have been made to publish reliable data and information, but the author
and the publisher cannot assume responsibility for the validity of all materials or for the conse-
quences of their use.
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 photocopying,
microlming, 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-prot 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 identication and explanation without intent to infringe.
QP144.G45P47 2007
612.3--dc22 2007012503
Table of Contents
Section I
Scientific Principles of Personalized Nutrition .....................................................1
Section II
Personalized Nutrition and Stakeholders in Society........................................149
Section III
The Future of Personalized Nutrition ...............................................................235
Index ......................................................................................................................279
9281_C000.fm Page v Thursday, July 26, 2007 7:30 PM
Foreword
PERSONALIZED NUTRITION: HISTORY
AND PROMISES?
Personalized nutrition is an emerging but controversial new discipline among nutri-
tion scientists. Due to its possible genetic component, it has become an attractive
topic for the media. So far, the science fiction stories of a lady doing food shopping,
guided by a PDA containing her genome information are not taken very seriously.
Of course, in the developed world, personalized food choice is taken for granted.
Did you realize that while ordering Italian three-taste gelato, you can choose from
over a million combinations and variations, if you consider tastes, sizes, toppings,
and packaging? The same is true for coffee or take-away pizzas.
In the context of this book, personalized nutrition is related to a differentiated
health effect, with the key questions targeted at the possible scientific basis for this
phenomenon. In order to put this in the right perspective, let us have a look at the
historical development of the diet and health relationship. Until the early twentieth
century, nutritional sciences dealt with hygiene and processing. This was followed
by the incorporation of biochemistry and (molecular) physiology: the vitamins were
described, energy metabolism was understood, etc. During the second half of the
twentieth century, knowledge of cellular and molecular biological processes allowed
for further optimizing of our diet toward a balanced macro- and micronutrient
composition. This contributed to a prolongation of life span and produced a com-
mercial wave of supplements.
Gradually, nutrition scientists indicated the involvement of diet in a large number
of diseases and disorders (e.g., colon cancer, cardiovascular disorders, type 2 diabetes
mellitus, a number of inflammation-related health problems, and many more. During
the 1990s, this development triggered the introduction of functional foods, dietary
components with added health value. So far, these functional foods, apart from
the aforementioned supplements (vitamins, cofactors, minerals, and so forth, which
I do not consider to be real functional foods), have resulted in only a limited number
of successful products (cholesterol-lowering stanols, probiotics, a number of specific
fatty acids). Except for the cholesterol-lowering products, where an established
biomarker (the relationship between CVD and HDL/LDL cholesterol) was estab-
lished by the biomedical research community, it was very difficult to obtain scientific
proof of efficacy for most other functional foods. In many cases, the advertisement
and PR efforts by far outreached the scientific efforts accompanying the market
introduction.
Why does nutrition science have such a hard job in providing evidence for health
claims related to a dietary component? In pharmacological and biomedical research,
bioactive compounds are developed to treat a well-characterized disease. In contrast,
nutrition deals with prevention of disease and optimizing health. Here, biomarkers
9281_C000.fm Page vi Thursday, July 26, 2007 7:30 PM
quantifying the health status are essentially missing, and much of the nutrition
research (the large observational and intervention cohorts) rely on disease endpoints
instead of health endpoints. Also in the gold standard of diet and health research,
the (cross-over) dietary intervention trial, the quantification of the effect is a major
issue. Usually, the observed effects are small, and great efforts have to be made to
unravel the intervention effect from potential confounding variables. In other words,
the confounding parameters have a larger impact than the intervention effects. The
recent omics-related observations in human intervention studies, using very accurate
biomarkers, confirm that intraindividual variation is much smaller than interindividual
variation. Differences between study subjects may be much larger than differences
affected by the dietary treatment.
The keys for personalized nutrition actually are these confounders that make
the life of nutrition scientists so difficult. Age, gender, lifestyle (e.g., exercise),
phenotype (e.g., body mass index), genetic makeup, and epi-genomic imprinting
all possibly determine our nutritional needs, the way we respond to nutrition, and
thus our personal diet-and-health relationship. Infant nutrition clearly differs from
a sports diet. Now, a triad of questions arise:
This book at least attempts to answer these questions. My personal opinion is that
indeed this relationship exists to a much greater extent than assumed until now, and
that nutrition science will need to do a much better job in accurately identifying and
quantifying the subtle differences in health status related to dietary treatment. A
complete merger of nutrition with a number of fundamental scientific disciplines
(molecular biology, biochemistry, bioinformatics, statistics, etc.) will be essential to
reach the goal.
Now, what about this specific and overemphasized part of personalized nutrition,
the impact of genetic differences? Of course, we know the examples of cholesterol
and folate, where genetic predisposition partly determines plasma concentration and
related health effects. In the case of cholesterol, a phenotypic assessment of LDL
and HDL provides a solid phenotypic biomarker, which makes genetic testing (at
least partly) unnecessary. In fact, this phenotypic readout is the gross result of at
least 20 genetic different makeups. The HDL/LDL relationship partly determines
biomedical intervention and the consumers food choice. In the case of folate, the
occurrence of a genetic polymorphism in one of the enzymes involved in folate
biotransformation results in a relatively low plasma folate concentration. In extreme
cases, this leads to embryonic deformation (spina bifida) due to impaired DNA
synthesis. Folate is essential for DNA synthesis as it provides the methyl group for
nucleotide synthesis. Folate is cheap, and some countries have turned to folate
supplementation to the general population (e.g., by fortification of bread). In other
words, a genetically based differential dietary advice is overruled by mass treatment.
9281_C000.fm Page vii Thursday, July 26, 2007 7:30 PM
Possible negative consequences in persons efficiently dealing with folate (high folate
levels have been shown to overmethylate DNA and are thus potentially mutagenic
and carcinogenic) were considered nonrelevant. Time will tell whether a personalized
approach would be better.
A number of disorders related to nutrition are packed with genetic variation, but
the effect of nutritional modulation of the phenotypic outcome of these variations
is difficult to assess as yet. Nutrition is the worst-case scenario for this approach in
science as multiple minor genetic differences can occur, possibly modulated by
multiple food bioactives, usually with low receptor affinity, and resulting in multiple
minor changes in gene expression and resulting phenotypic expression.
Eventually, nutrition science may very likely determine a large number of per-
sonalized nutrition and health relationships. However, this is only a small part of
the equation. Food consumption nowadays is only partially driven by health con-
cerns. It is still much more driven by convenience and price. Food is pleasure
rightfully is a universally held credo, and science will have a hard job in promoting
healthy diet if this aspect is compromised. Therefore, a personalized diet needs to
be both optimized toward personal health and also for personal convenience, plea-
sure, and affordability. What a challenge!
Preface
The primary role of diet is to provide an individual with optimal levels of nutrients
to meet metabolic requirements, and to give the consumer a feeling of satisfaction
and well being through the pleasure of eating. In addition, particular diets, foods
and food components can provide additional physiological, cognitive and psycho-
logical benefits, and result in biological activities beyond their widely accepted
nutritional effects. Moreover, diet not only helps to achieve and maintain optimal
health and development but can also play an important role in reducing the risk of
specific diseases and disorders.
However, a diet that serves the health of one individual does not necessarily
work for all individuals due to differences in (genetic) predisposition, and environ-
mental and lifestyle factors. Hence, diagnostic tools that reflect both the overall
health status and (genetic) predisposition of a particular person at a given time need
to be developed. The knowledge obtained thereby will facilitate appropriate nutri-
tional advice to individuals according to their immediate and long-term health needs
and, ultimately, we can foresee the time when nutritional products will be tailored
for individual consumers.
The success of the introduction of personalized nutrition in society depends on many
factors. The scientific background for this must be established, the regulatory system
must be in place, ethical issues need to be addressed, services and products with clear
health benefits must be available, and the accompanying communication must be clear.
This textbook has the aim of defining the area of personalized nutrition both
from a biomedical and social science perspective. A selected group of leading
scientists in the field will comprehensively address the molecular, physiological,
epidemiologic, and public health aspects of personalized nutrition, highlighted with
examples from major diseases. Another group of well-known social scientists will
discuss the behavioral, ethical, and consumer perspectives that will influence a
legitimate successful introduction of personalized nutrition.
We expect our book will be useful for the education of students in several
disciplines, for example, nutrition, behavioral and communication science. More-
over, stakeholders involved in personalized nutrition in government, health care, and
business may use the book as a reference guide.
Our book is divided in three sections: (1) Scientific Principles of Personalized
Nutrition, (2) Personalized Nutrition: Consumer and Society, and (3) Future Per-
spectives on Personalized Nutrition.
In the first section, Scientific Principles of Personalized Nutrition, the state of
the art of nutrigenomics technologies, including transcriptomics, proteomics, and
metabolomics, are discussed. Subsequently, the use of genomics technology for a
better understanding of the molecular mechanisms involved in major diet-related
chronic disorders i.e., chronic inflammation, cardiovascular disease, type 2
diabetes, cancer, and obesity is addressed.
9281_C000.fm Page x Thursday, July 26, 2007 7:30 PM
Frans J. Kok
Laura Bouwman
Frank Desiere
9281_C000.fm Page xi Thursday, July 26, 2007 7:30 PM
The Editors
Frans J. Kok PhD, is full professor of nutrition and health and head of the Division
of Human Nutrition at Wageningen University, The Netherlands. Moreover, as dean of
Wageningen Graduate Schools, he is responsible for securing the quality of academic
research and training at the university.
Dr. Kok was trained in human nutrition at Wageningen University and in epi-
demiology at Harvard University, Cambridge, Massachusetts. His scientific expertise
covers topics such as energy balance and body composition, diet in disease preven-
tion, and sensory science. He has authored more than 250 original scientific publi-
cations and is a member of the Dutch Health Council and the Academic Board of
Wageningen University. As a faculty member, he has lectured in many courses on
nutrition and health, including the European Nutrition Leadership Program, as well
as in courses in the U.S., Africa, and Asia.
Frank Desiere, PhD, MBA, started his scientific career in the earlier days of biotech-
nology doing research in fermentation technology, microbiology, and biological conver-
sion of xenobiotics, and later joined the Nestl Research Center in Lausanne, Switzerland,
to work on the ecology and evolution of bacteriophages in milk fermentation. Later, he
initiated a bioinformatics effort at Nestl and worked on microbial genomics, transcrip-
tomics, and proteomics. Dr. Desiere also initiated Peptideatlas (www.peptideatlas.org)
for the integration of proteomics data with human and other genomes. He was then
responsible for a project involving personalized nutrition at Nestl that initiated consumer
and patient applications of personalized nutrition concepts in the marketplace. As a result
of this initiative, his interests evolved into the commercial application of scientific con-
cepts in science and nutrition, management of innovation and new business ideas. After
having completed an MBA at IMD Lausanne, Dr. Desiere now works as business
development manager at Roche Diagnostics in Basel, Switzerland. His scientific activities
have resulted in more than 30 peer-reviewed publications and numerous presentations
at international scientific conferences.
9281_C000.fm Page xii Thursday, July 26, 2007 7:30 PM
9281_C000.fm Page xiii Thursday, July 26, 2007 7:30 PM
Contributors
Lydia A. Afman Hannelore Daniel
Nutrition, Metabolism, and Genomics Molecular Nutrition Unit
Division of Human Nutrition Center of Life and Food Sciences
Wageningen University Technical University of Munich
Wageningen Freising-Weihenstephan
The Netherlands Germany
Josh Conway
Cogent Research, LLC J. Bruce German
Cambridge, Massachusetts University of CaliforniaDavis
Davis, California
Dolores Corella
Department of Preventive Medicine Michael J. Gibney
University of Valencia UCD Agriculture and Food Science
Genetic and Molecular Epidemiology Centre
Unit University College
Valencia Dublin
Spain Ireland
9281_C000.fm Page xiv Thursday, July 26, 2007 7:30 PM
Danielle R. Reed
Maria A. Koelen Monell Chemical Senses Center
Wageningen University Philadelphia, Pennsylvania
Sub-Department of Communication
Science, Communication Manuela Rist
and Innovation Group Molecular Nutrition Unit
Wageningen Technical University of Munich
The Netherlands Freising-Weihenstephan
Germany
Michiel Korthals Amber Ronteltap
Applied Philosophy Marketing and Consumer Behavior
Social Sciences Group
Wageningen University Wageningen University
Wageningen Wageningen
The Netherlands The Netherlands
9281_C000.fm Page xv Thursday, July 26, 2007 7:30 PM
Section I
Scientific Principles
of Personalized Nutrition
9281_S001.fm Page 2 Wednesday, July 18, 2007 4:31 PM
9281_C001.fm Page 3 Thursday, July 26, 2007 7:04 PM
1 Nutrigenomics
and Transcriptomics:
Applications of Microarray
Analyses in Nutrition
Research
Lydia A. Afman and Michael Mller
CONTENTS
1.1 INTRODUCTION
This chapter describes how nutrients can influence gene expression and describes
how this knowledge can be applied in nutritional research by utilization of transcrip-
tional profiling. Two research approaches are discussed: a mechanistic approach and
3
9281_C001.fm Page 4 Thursday, July 26, 2007 7:04 PM
Systems-biology
databases and bioinformatics
Nutrient
Metabolomics and
Functions functional
Transporter genomics
Molecular-
biology tools Transcription
Tet-on + factors Molecular
Proteins Proteomics
biomarkers
Transgenics mechanisms
RNAi and targets
tdnAd
Nucleus
1.3 TRANSCRIPTOMICS
1.3.1 TECHNOLOGY
Transcriptomics is both a sensitive and well-validated technique, which employs
high-density oligonucleotide microarray analysis to study the number of mRNA
copies per gene for virtually all actively transcribed genes. This technique enables
determination of expression levels of genes of the whole genome, meaning hundreds
to thousands of genes at the same time and within one study. As nutrients influence
transcription of genes, this technique can be applied to measure diet-induced changes
in every tissue of interest. A challenge in nutritional research is the relatively small
effect of dietary interventions on physiological parameters. Similarly, the effects of
nutrition on gene expression patterns are also rather small and not easy to detect.
Consequently, the development of a highly sensitive and validated microarray anal-
ysis platform is required along with a robust research hypothesis that is adequately
addressed by the experimental design to avoid so-called fishing expeditions.
(Ptitsyn, Zvonic et al. 2006; Zvonic, Ptitsyn et al. 2006). Another undesirable factor
is pooling of samples, but increasing the number of biological replicates will enhance
accuracy by decreasing the false-positive rate and result in reliable data (Kendziorski,
Irizarry et al. 2005). Once a transcriptomics study is performed, RNA quality and
quantity should be verified and subsequent labeling and hybridization of RNA should
preferably be conducted by the same technician to keep variation between arrays
low. Preprocessing of data requires specific attention, as the small gene expression
changes in dietary studies require a highly reliable algorithm (Irizarry, Warren et al.
2005). The most challenging part of the process occurs after the array data are
obtained, when the data have to be analyzed in terms of their biological meaning.
Several commercial and noncommercial tools have been developed that assist with
the extraction of significantly changed genes and the visualization of changed path-
ways or related networks (Curtis, Oresic et al. 2005).
this method less suitable for application in short or time-course intervention studies.
Another characteristic of an appropriate nutrigenomics design is a high number of
replicates; the accompanying high costs, however, are often the limiting factor in
this respect. A third aspect of a sound study design is the magnification of the signal-
to-noise ratio, leading to an increased change on selecting the correct but rather
small diet-induced effects. Such an effect could be achieved by magnifying the
response through a nutritional challenge of the body.
The number of human studies that describe nutrition-related transcriptome pro-
filing is limited. Most of the papers describe differential gene expression in muscle
and adipose tissue biopsies in nutrition-related chronic diseases.
(Mootha, Lindgren et al. 2003; Patti, Butte et al. 2003). Moreover, they showed that
these changes have most likely been mediated by the peroxisome proliferator-
activated receptor coactivator-1 (PGC1), which was found to be downregulated in
these subjects as well. Similarly, microarray analyses performed on muscle biopsies
before and after 3 d of high-fat feeding showed a downregulation in expression of
genes involved in oxidative phosphorylation or mitochondrial function. This high-
fat feeding intervention also resulted in the downregulation of PGC1 and PGC1,
similar to the findings in diabetic muscle tissue (Sparks, Xie et al. 2005). To confirm
this, a comparable intervention study was performed with C57Bl/6 mice fed a high-
fat diet for 3 weeks. The gene expression changes due to high-fat feeding in the
mouse study were of a greater magnitude than those observed in the human study,
but were all related to a decrease in the expression of genes involved in oxidative
phosphorylation and mitochondrial biosynthesis and its master regulator, PGC1. The
authors suggest that high-fat diet may explain the reduction in oxidative phospho-
rylation previously observed in muscle of type 2 diabetes patients and prediabetic
individuals (Kelley, He et al. 2002). In conclusion, this is a good example of a
nutrigenomics study demonstrating that diet-induced changes in inbred mice that
are genetically identical animals are of a greater magnitude than comparable inter-
ventions performed in humans with different genotypes.
With the use of transcriptomics techniques, the aforementioned studies were
able to identify pathways involved in different stages of the development of type 2
diabetes. In addition, these studies were able to demonstrate that these pathways
were changed in the same direction after intervention with a high-fat diet, implying
that this pathway can be influenced by diet, which points toward a mechanism in
diet-induced type 2 diabetes. The question still remains whether transcriptional
analyses of this pathway can be used as a diagnostic tool for the early predisease
state and whether and which dietary adjustments can lead to upregulation in oxidative
phosphorylation, mitochondrial biosynthesis and PGC1, and ultimately result in the
prevention of type 2 diabetes.
KEY READINGS
Afman, L. and Mller, M. (2006). Nutrigenomics: from molecular nutrition to prevention of
disease. J Am Diet Assoc 106(4): 56976.
Desvergne, B., Michalik, L. et al. (2006). Transcriptional regulation of metabolism. Physiol
Rev 86(2): 465514.
Mller, M. and Kersten, S. (2003). Nutrigenomics: goals and strategies. Nat Rev Genet 4(4):
31522.
Mutch, D.M., Wahli, W., and Williamson, G. Nutrigenomics and nutrigenetics: the emerging
faces of nutrition. FASEB J 19(12): 160216, October 2005.
REFERENCES
Afman, L. and Mller, M. (2006). Nutrigenomics: from molecular nutrition to prevention of
disease. J Am Diet Assoc 106(4): 56976.
Calvano, S.E., Xiao, W. et al. (2005). A network-based analysis of systemic inflammation in
humans. Nature 437(7061): 10327.
Clement, K., Viguerie, N. et al. (2004). Weight loss regulates inflammation-related genes in
white adipose tissue of obese subjects. FASEB J 18(14): 165769.
Cobb, J.P., Mindrinos, M.N. et al. (2005). Application of genome-wide expression analysis
to human health and disease. Proc Natl Acad Sci USA 102(13): 48016.
Curtis, R.K., Oresic, M. et al. (2005). Pathways to the analysis of microarray data. Trends
Biotechnol 23(8): 42935.
Desvergne, B., Michalik, L. et al. (2006). Transcriptional regulation of metabolism. Physiol
Rev 86(2): 465514.
9281_C001.fm Page 12 Thursday, July 26, 2007 7:04 PM
CONTENTS
2.1 INTRODUCTION
Proteome analysis of a particular cell or organ, or of body fluids such as plasma,
urine, and others, makes it possible to identify biomarkers that respond to alterations
in diet or any other treatment, and that may have a predictive value for the assessment
of a humans health status. To date, proteomics has not been applied on a large scale
in human studies related to nutrition, yet it has obvious advantages over transcrip-
tome profiling techniques in that it directly assesses the molecular entities that carry
out the biological functions. This chapter summarizes the different approaches in
proteomics research and provides some examples that demonstrate the state of the
art of biomarker discovery based on proteome analysis and their implications for
the development of personalized nutrition approaches.
Human metabolism can be described as the orchestrated interplay of a huge
number of proteins that perform different functions as receptors, enzymes, signaling
molecules, and transporters or structural components, and that are expressed at the
level of the cell, the organ, or whole organism. Proteins are the transmitters of
metabolic functions, and therefore changes in the level of an individual protein or
sets of proteins may indicate an altered metabolic condition. Protein synthesis and
protein degradation, and thereby metabolic functions, can be affected by the overall
composition of the diet, the levels of specific nutrients and nonnutrient components
of foods and, indeed, many other aspects of lifestyle, in the most complex ways.
Analysis of the proteome as the whole protein complement of the genome expressed
in a particular cell, an organ, or body fluid also called proteomics allows
13
9281_C002.fm Page 14 Wednesday, July 18, 2007 1:47 PM
changes in protein expression patterns, and the identity of the proteins themselves
to be determined simultaneously. Moreover, for individual proteins, posttranslational
modifications that may be crucial for protein function, or even amino acid sub-
stitutions (polymorphisms), can be detected. The potential value of proteomics for
assessing markers of health has been recognized for some years [1], but only very
few studies so far have employed proteome analysis as a tool for research in human
nutrition and health [2].
Biouid sample
2D-gel Tryptic
separation digestion
Peptide mixture
Mixture of proteins
LC-separation
Column
Excised protein
Loading on chips
Mixture of peptides
generated. The mixture of peptides isolated from the gel spot after digestion with
the protease is usually submitted to matrix-assisted laser desorption/ionization time-
of-flight mass spectrometry (MALDI-TOF-MS) analysis to determine the corre-
sponding peptide masses that are characteristic for a given protein and serve as the
peptide mass fingerprint. The obtained mass spectrum is evaluated with computer
programs that apply various algorithms for interpretation of the peptide pattern and
predict the protein identity based on a comparison with masses predicted by virtual
digestion of identified open-reading frames in a given genome [8].
In contrast to gel-based proteomics analysis, shotgun proteomics is a gel-free
approach that uses different liquid chromatography techniques for separation of
complex peptide mixtures coupled with mass spectrometry identification [9]. It starts
with the isolation of the protein-containing sample, which may also be prefraction-
ated by chromatographic or electrophoretic techniques followed by a digestion step
to generate a complex mixture of peptides. Multidimensional chromatographic tech-
niques are subsequently used to separate and further fractionate the obtained peptide
mixtures, which are then submitted for analysis by electrospray ionization (ESI) or
MALDI-based mass analysis. Sophisticated mathematical algorithms are used to
compare the peptide sequence data derived from mass spectra with sequence infor-
mation in protein databases to identify the proteins.
Major technological advancements in protein chemistry have led to chip-based
protein sample arrays with different chromatographic surfaces to selectively bind
9281_C002.fm Page 16 Wednesday, July 18, 2007 1:47 PM
proteins with specific chemical properties. One of the evolved technologies is the
surface-enhanced laser desorption ionization time-of-flight (SELDI-TOF) platform
for high-throughput analysis of proteins in complex biological specimens [10]. It
uses a protein chip array system with different chromatographic properties, including
hydrophobic, hydrophilic, anion exchange, cation exchange, or immobilized-metal
affinity surfaces. After addition of an energy-absorbing matrix to the chip, the
proteins are analyzed by MALDI-TOF mass analysis. The output from the proteome
analysis based on chips via SELDI-TOF is a trace of relative intensity vs. m/z (mass-
to-charge ratio) of the detected proteins, providing a pattern that can be compared
from different samples or that can be used for cluster analyses for significant protein
abundance differences between samples. The future of more simple proteome anal-
ysis tools may be the use of antibody libraries that contain specific antibodies raised
against an expressed open-reading frame and making proteome analysis similar to
high-throughput microplate assays that allow essentially every protein to be identi-
fied and quantified easily [11].
Eux Proteome of
blood cells
Eux
Inux Plasma
proteome
Eux
Eux Urine
proteome
FIGURE 2.2 The importance of human blood as a source of biological information with the
possibilities of analyzing plasma, blood cell proteomes, and urine as easily obtainable samples.
9281_C002.fm Page 17 Wednesday, July 18, 2007 1:47 PM
the condition of individual organs and the physiological processes associated with
it. Moreover, proteins in the blood are actively participating in many life-sustaining
body functions such as the transport of nutrients and metabolites, inflammation,
hormone signaling, and many more. The blood proteome therefore promises to be
an ideal biological entity for assessing markers of health or disease.
Analysis of blood proteins, however, depends on many variables, most impor-
tantly, on the methods of sampling, sample processing, and storage. A particular
problem is that plasma/serum contains around 22 abundant proteins that represent
99% of the protein mass. The most prominent protein is albumin, with around
45,000 g/ml, whereas the concentration of other proteins, such as the alpha-
fetoprotein, may be as low as 0.005 g/ml [12]. This huge concentration difference
makes analysis of the plasma proteome very difficult and requires the removal of
the most abundant proteins from the sample to allow a reliable analysis of the minor
components. During the course of the Human Proteome Project (HPP), a group of
38 expert labs has focused on the human plasma proteome. The Human Plasma
Proteome Project (HPPP) has worked out standard operational procedures and has
made recommendations for sample analysis and processing [13]. A wealth of bio-
logical information is already available from the HPPP and is in the public domain
(http://www.plasmaproteomedatabase.org/). A total of 2446 unique gene products
and more than 4900 protein entities were identified when specific isoforms were
taken into account. In silico methods were employed to determine the origin and
nature of the various plasma proteins (see Figure 2.3). This analysis delivered a
surprisingly high number of unexpected protein classes, including a large number
Cytoplasma 53%
Cellular proteins 58% Membrane-associated 19%
Other extracellular or secreted 14%
Immunoglobulins 8% Nuclear 9%
Cytokine and related 5%
Unknowns 21%
Common circulatory proteins 28%
Classic plasma proteins 13% Coagulation and complement factors 26%
Binding proteins 14%
Inhibitors 13%
Proteases and other enzymes 19%
FIGURE 2.3 The origins and nature of the proteins identified in human plasma by the HPPP.
9281_C002.fm Page 18 Wednesday, July 18, 2007 1:47 PM
of intracellular proteins, and even proteins of nuclear origin. Whether they are
indicators of normal tissue renewal processes or a measure of tissue (organ-specific)
damage is currently under investigation. In this context, the isoform-specific proteins
(in case of selected enzymes, these are well-established clinical markers) in plasma
are interesting for further exploration. The nutritional status is known to affect the
levels of distinct plasma proteins, and organ-specific processes appear also to be
detectable in plasma as some of the proteins found in plasma are derived from
individual organs. In the HPPP, 362 of the plasma proteins were of hepatic origin
and 345 proteins were cardiovascular related, and about 110 proteins could be
related to inflammation such as cytokines, adhesion molecules, and chemokines [14].
Plasma peptides are usually too small in molecular mass for standard proteome
analysis. However, they are of particular interest because of their specific functions
in biological processes in which they act as hormones, growth factors, adipokines,
etc. The plasma peptidome defined as the fraction with peptides of < 10,000 or
15,000 Da therefore requires special techniques for separation and analysis.
Usually, a prefractionation of plasma based on ultrafiltration techniques with mem-
branes of different cutoff values is used and then further chromatographic separation
steps combined with mass analysis are employed to characterize the low-molecular-
weight peptides in the sample. The plasma peptidome is considered to represent up
to 5000 different peptides [15].
Proteome analysis may also be applied to circulating blood cells. The fraction
of mononuclear cells can easily be collected from blood samples by commercial
separation kits, and these cells may serve as reporter cells as they are in circulation
for extended time periods and reach different body compartments. Proteome analysis
of human peripheral mononuclear cells has not been performed in the context of
biomarker discovery or clinical diagnostics. In contrast, several studies have targeted
the platelet proteome, and a high-resolution platelet proteome map that comprises
more than 2000 different protein features has been generated [16,17].
Other body fluids such as urine, saliva, tears, seminal fluid, synovial fluid, and
cerebrospinal fluid have also been submitted to proteome analysis in search for
biomarkers of disease and toward improved clinical diagnostics [18].
samples were obtained and large, abundant proteins removed. Bioinformatic analysis
of MALDI-TOF spectra revealed two significant peaks that could classify partici-
pants based on diet with 76% accuracy. One of the peaks (at 2740 m/z) was identified
as the B-chain of -2-HS glycoprotein, a serum protein previously found to vary
with diet and thought to be involved in insulin resistance and immune function [25].
The adipocyte fatty acidbinding protein (A-FABP) is considered to be a cyto-
solic fatty acid chaperone. However, mice with a targeted disruption of the A-FABP
gene show protection from insulin resistance, hyperglycemia, and atherosclerosis,
suggesting that A-FABP could have additional functions. 2D-gel-based separations
of proteins from cultured adipocytes identified the cellular fatty acid-binding protein
as a prominent secretion product. When obese patients were concomitantly screened
for plasma levels of A-FABP, it was observed that concentrations of A-FABP were
significantly higher in overweight and obese as compared to lean persons and after
adjustment for age and sex, serum A-FABP concentrations correlated significantly
with blood pressure, fasting insulin, waist circumference, and dyslipidemia. A-FABP
therefore appears to be a new biomarker that is associated with obesity and the
metabolic syndrome [26].
analysis of hemoglobin A1) or by monitoring the urine proteome, which may indicate
disturbed kidney function at an early stage as the disease progresses. Detecting serum
autoantibodies in autoimmune diseases or quantitative measurements of serum aller-
gen-specific IgE levels in type I allergy are already possible via proteome analysis
in chip formats with minute amounts of serum. A detailed analysis of the protein
components of the lipoproteins of the LDL and HDL classes may provide better
diagnostics for arthrosclerosis and heart disease, in particular, when different iso-
forms related to different organs of origin or because of genetic variations are taken
into account. However, before proteomics moves into the real world and these
new and more convenient assay systems become available, nutritional science needs
to explore with existing technologies the power of proteomics in characterizing
human metabolism and diet responses in well-defined studies.
REFERENCES
1. Chambers, G. et al., Proteomics: a new approach to the study of disease, J Pathol,
192, 280, 2000.
2. Kussmann, M. et al., Proteomics in nutrition and health, Comb Chem High Through-
put Screen, 8, 679, 2005.
3. Gorg, A. et al., The current state of two-dimensional electrophoresis with immobilized
pH gradients, Electrophoresis, 21, 1037, 2000.
4. Cordwell, S.J. et al., Subproteomics based upon protein cellular location and relative
solubilities in conjunction with composite two-dimensional electrophoresis gels,
Electrophoresis, 21, 1094, 2000.
5. Corthals, G.L. et al., The dynamic range of protein expression: a challenge for
proteomic research, Electrophoresis, 21, 1104, 2000.
6. Adam, G.C. et al., Profiling the specific reactivity of the proteome with non-directed
activity-based probes, Chem Biol, 8, 81, 2001.
7. Conrads, T.P. et al., Utility of accurate mass tags for proteome-wide protein identi-
fication, Anal Chem, 72, 3349, 2000.
8. Chamrad, D.C. et al., Interpretation of mass spectrometry data for high-throughput
proteomics, Anal Bioanal Chem, 376, 1014, 2003.
9. Wolters, D.A. et al., An automated multidimensional protein identification technology
for shotgun proteomics, Anal Chem, 73, 5683, 2001.
10. Issaq, H.J. et al., The SELDI-TOF MS approach to proteomics: protein profiling and
biomarker identification, Biochem Biophys Res Commun, 292, 587, 2002.
11. LaBaer, J. and Ramachandran, N., Protein microarrays as tools for functional pro-
teomics, Curr Opin Chem Biol, 9, 14, 2005.
12. Qian, W.J. et al., Comparative proteome analyses of human plasma following in vivo
lipopolysaccharide administration using multidimensional separations coupled with
tandem mass spectrometry, Proteomics, 5, 572, 2005.
13. Rai, A.J. et al., Hupo plasma proteome project specimen collection and handling:
towards the standardization of parameters for plasma proteome samples, Proteomics,
5, 3262, 2005.
14. Ping, P. et al., A functional annotation of subproteomes in human plasma, Proteomics,
5, 3506, 2005.
15. Richter, R. et al., Composition of the peptide fraction in human blood plasma: database
of circulating human peptides, J Chromatogr B Biomed Sci Appl, 726, 25, 1999.
9281_C002.fm Page 22 Wednesday, July 18, 2007 1:47 PM
16. Garcia, A. et al., Applying proteomics technology to platelet research, Mass Spectrom
Rev, 24, 918, 2005.
17. ONeill, E.E. et al., Towards complete analysis of the platelet proteome, Proteomics,
2, 288, 2002.
18. Veenstra, T.D. et al., Biomarkers: mining the biofluid proteome, Mol Cell Proteomics,
4, 409, 2005.
19. Alaiya, A. et al., Clinical cancer proteomics: promises and pitfalls, J Proteome Res,
4, 1213, 2005.
20. Chatterjee, S.K. and Zetter, B.R., Cancer biomarkers: knowing the present and pre-
dicting the future, Future Oncol, 1, 37, 2005.
21. Liao, H. et al., Use of mass spectrometry to identify protein biomarkers of disease
severity in the synovial fluid and serum of patients with rheumatoid arthritis, Arthritis
Rheum, 50, 3792, 2004.
22. Davidsson, P. and Sjogren, M., Proteome studies of CSF in AD patients, Mech Ageing
Dev, 127, 133, 2006.
23. Meier, M. et al., Identification of urinary protein pattern in type 1 diabetic adolescents
with early diabetic nephropathy by a novel combined proteome analysis, J Diabetes
Complications, 19, 223, 2005.
24. Pisitkun, T. et al., Identification and proteomic profiling of exosomes in human urine,
Proc Natl Acad Sci USA, 101, 13368, 2004.
25. Mitchell, B.L. et al., Evaluation of matrix-assisted laser desorption/ionization-time
of flight mass spectrometry proteomic profiling: identification of alpha 2-HS glyco-
protein B-chain as a biomarker of diet, Proteomics, 5, 2238, 2005.
26. Xu, A. et al., Adipocyte fatty acid-binding protein is a plasma biomarker closely
associated with obesity and metabolic syndrome, Clin Chem, 52, 405, 2006.
27. Kung, L.A. and Snyder, M., Proteome chips for whole-organism assays, Nat Rev Mol
Cell Biol, 2006.
9281_C003.fm Page 23 Thursday, July 26, 2007 7:05 PM
3 Metabolomics
and the Personalized
Metabolic Signature
Michael J. Gibney, Marianne Walsh,
and Lorraine Brennan
CONTENTS
3.1 INTRODUCTION
Whereas traditional nutrition and metabolism research sets out a priori the metab-
olites of interest in the hypothesis to be tested, nutritional metabolomics attempts
to capture the presence of the maximum number of small chemicals regardless of
their identity or relevance to any advance hypothesis. It is thus a hypothesis-
generating exercise. The totality of all signals in a sample is reduced in a two-stage
process to one point in two-dimensional space, and metabolic signatures that are
comparable will cluster closely together and, as the metabolomic profiles differ, the
points separate in proportion to the differences. Metabolomic profiles of many
diseases have been studied, and distinct clusters have been identified. Similarly, met-
abolic profiles arising from pharmacological intervention have also been characterized
and are found to cluster together for similar drugs. To date little or no serious work
has been published on nutritional metabolomics, but a number of clear challenges
exist. In nutrition, metabolic noise will be relatively high because food contains
23
9281_C003.fm Page 24 Thursday, July 26, 2007 7:05 PM
many nonnutrients, which are absorbed and which will be detectable in biofluids.
This, coupled with the fact that the effect of diet on metabolomic profiles is likely
to be milder than that of disease or drugs, makes the challenge of nutritional
metabolomics very special. However, whether it is through targeted metabolomic
profiles or universal metabolomic profiles, nutritional metabolomics offers great poten-
tial for nutrition research.
Traditionally, nutritional science has proceeded in a hypothesis-driven manner
in which a priori, end points were known, were part of the hypothesis, and were
the basis of experimental measure. Although that will remain so, new advances in
biology have led to the development of a parallel strategy that is hypothesis gener-
ating rather than hypothesis testing. Metabolomics is part of that new biology. As
ever in science, terminology becomes important, and two terms coexist in this field,
metabolomics and metabonomics. For completeness we will present the original
definitions of both and leave it to the reader to distinguish between the two. Meta-
bonomics is defined as the quantitative measurement of the time-related multi-
parametric metabolic response of living systems to pathophysiological stimuli or
genetic modification. On the other hand, metabolomics has been defined in the
literature as the comprehensive and quantitative analysis of all metabolites .
Throughout this chapter we will use the term metabolomics exclusively but remind
the reader that either term can be used.
In relation to human studies, metabolomics is the measurement of the maximum
number of metabolites present in a biofluid. A range of analytical methods can be
used to generate data. However, mass spectrometry (MS) in its various hyphenated
derivations (LC-MS, GC-MS) and high-resolution 1H nuclear magnetic resonance
spectroscopy (1H NMR) are the dominant platforms. Both platforms have their
advantages and disadvantages, and best practice is to apply both, if possible. For
the purpose of this chapter we will focus more on NMR applications but in no way
wish to claim it is a superior technique. In the case of 1H NMR, each biofluid has
a characteristic spectroscopic fingerprint that reflects the metabolic composition (see
Figure 3.1 for 1H NMR spectra of different biofluids). The intensities of peaks, in
an NMR spectrum, belonging to a certain metabolite are determined by its concen-
tration, and assignment can be made based on chemical shifts (position in spectra),
spin multiplicities, and addition of authentic material. Typical metabolomic studies
generate large amounts of data that cannot easily be analyzed in an unbiased way.
To overcome this problem, a number of pattern recognition techniques have been
applied to robustly analyze NMR spectra.
The most commonly used method to analyze metabolomics data is through
principal component analysis (PCA). This statistical method reduces a large number
of variables to a smaller number of variables, called principal components. The
principles underlying the reduction of a complex multisignal NMR output to a single
point in two-dimensional space through PCA is best illustrated with a 3-peak output
rather than with the several hundred peaks normally found in NMR spectra.
Figure 3.2A and Figure 3.2B show how these three peaks can be processed: first,
the multipeak output is summarized by one point in multidimensional space and
then reduced to a lower two-dimensional space. In the resulting scores plot, each
point represents all the data contained in the spectrum belonging to one individual.
9281_C003.fm Page 25 Thursday, July 26, 2007 7:05 PM
// //
Creatinine
TMAO
Citrate
DMA
Glycine
Alanine
C
4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2 1.0
Chemical shift (ppm)
//
Propionate
Propionate
Acetate
Lactate
Pyruvate Alanine
Choline
B
4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2 1.0
Chemical shift (ppm)
Lactate
Valine
Alanine
Acetoacetate
Lactate Glutamine
Acetate
A
4.4 4.2 4.0 3.8 3.6 3.4 3.2 3.0 2.8 2.6 2.4 2.2 2.0 1.8 1.6 1.4 1.2 1.0
Chemical shift (ppm)
FIGURE 3.1 Typical 1H NMR spectra of biofluids obtained from a healthy volunteer: (a) plasma,
(b) saliva, and (c) urine. TMAO, trimethylamine-N-oxide; DMA, dimethylamine. (From Am
J Clin Nutr, 2006, 84:531539. With permission.)
C B
b
FIGURE 3.2A Three NMR or MS peaks a, b, and c, must be reduced to one point in multidi-
mensional space. Three coordinates A, B, and C are used to achieve this. The signal strength
of peak a is laid on coordinate A proportionate to the size of the signal. Peak b is now placed
parallel to coordinate B, and similarly peak c is set parallel to coordinate C, each proportionate
to the size of the relevant signal. The final point is the only position these three peaks can give
in three-dimensional space, and the same holds true for k peaks in K-dimensional space.
9281_C003.fm Page 26 Thursday, July 26, 2007 7:05 PM
PC 1
PC 2
FIGURE 3.2B The figure on the left should be imagined as a bunch of grapes in multidi-
mensional space. The center (nonfilled) grape is found, and a pencil is inserted into the bunch
of grapes through the center multiple times from all possible angles until maximum variance
to the bunch of grapes is found. A second pencil is similarly inserted at a right angle to the
first. Half of each pencil is lost, and the distance each grape is from each pencil [principal
components (PC) 1 and 2] is measured as shown for one point with an arrow on the right
figure. Now, a two-dimensional plot retaining maximum information on multidimensional
space is developed.
Hence, observation points that cluster close together have similar spectra, whereas
those that are far apart will have very different spectra. The spectral variables
responsible for the positioning of the observations in the scores plot can be obtained
from the loadings plot. Although PCA is the initial unsupervised data analysis
carried out in most metabolomic studies, it is usually followed by more sophisticated
supervised techniques such as Partial Least Squares Discriminant Analysis (PLS-DA).
The power of these statistical techniques to reduce complex NMR profiles to
single points in two-dimensional space is shown in Figure 3.3A and Figure 3.3B.
Figure 3.3A gives typical NMR spectra for urine samples collected in the morning
or in the evening by the same individual. Clearly, the samples taken at different time
points exhibit differences in the NMR profiles which are clearly discernible to the
Night
Morning
11 10 9 8 7 6 5 4 3 2 1
Chemical shift (ppm)
FIGURE 3.3A 1H NMR spectra of urine from the same subject sampled first thing in the
morning and last thing in the evening.
9281_C003.fm Page 27 Thursday, July 26, 2007 7:05 PM
Morning 12 Morning
Night 10 Night
10 8
6
0 4
2
t(2)
t(2)
0
10 2
4
6
20 8
10
12
20 10 0 10 10 0 10
t(1) t(1)
PCA PLSDA
FIGURE 3.3B Principal component analysis (PCA) and partial least squares discriminant
analysis (PLS-DA) of NMR data from human urine samples collected both in the early
morning and late evening.
human eye, as shown by asterisks. When PCA and PLS-DA, in particular, are applied,
quite clear separation of the nightmorning samples is seen because the samples
taken at each time point have comparable signatures but nighttime and morning-
time signatures are quite different. To date, metabolomic profiling of biofluids have
shown unique signatures for cases and controls in many chronic diseases such as
cardiovascular disease, cancers, osteoarthritis, and several neurodegenerative diseases.
The metabolomic profiles of human biofluids are determined by a number of
identifiable factors, although the quantitative effect of each individual factor remains,
as yet, unknown. It is evident from later sections that the presence or absence of a
disease or, if present, the severity of the disease is one determinant of a metabolic
signature. However, for the purposes of this section of the chapter, the focus will
be on physiological factors determining the nature of the metabolomic profiles in
human biofluids. For clarity in what can be a rather confused landscape, environ-
mental factors, including stress, will not be considered. This is not to say that these
are unimportant but, rather, it is advantageous to focus on core determinants of
metabolomic profiles that are amenable to dietary influence. Thus, this chapter will
focus on the effects of phenotype and endogenous metabolism on food with an
evaluation of the effects of both nutrients and nonnutrients, and also on the role of
the colonic microflora.
It is the essence of life that biological systems turn over constantly with a simulta-
neous set of anabolic and catabolic pathways. The balance of these is determined
by a variety of endocrine-based factors. The anabolic phase is dominant in growth,
reproduction, and weight gain, whereas the catabolic factors are dominant in aging,
9281_C003.fm Page 28 Thursday, July 26, 2007 7:05 PM
illness, and weight loss. This constant entry and exit of metabolites from plasma
plays a major role in determining the metabolic profile of that biofluid in the normal
sampling state of fasting. Phenotype therefore must play a very significant role in
shaping fasting plasma metabolomic profiles. For example, protein turnover is
strongly related to lean body mass and, thus, per unit of plasma, the rate of exit and
entry of amino acids into plasma will be far higher in individuals with a higher as
compared to a lower lean body mass per unit weight. Height and girth will reflect
skeletal differences, and greater skeletal mass will mean greater traffic of related
metabolites in and out of plasma. Clearly, as percentage body fat increases, the
constant anabolism and catabolism in adipose tissue determines the flow of nones-
terified fatty acids, glycerol, glucose, and triacylglycerol-rich lipoproteins and their
remnants to and from the liver via the plasma metabolome. Diet can clearly alter
the nature of the plasma metabolome in the postprandial phase, and the nature of
that effect will be dependent on the nature of the ingested meal. However, this is an
acute event that in the short term will not influence the nature of the fasting plasma
metabolome. If a modification to diet is sustained such that it can be described as
chronic, then it will lead eventually to a readjustment of gene expression and to a
redefining of the parameters of anabolic and catabolic pathways, and that will
influence the fasting plasma metabolome. Thus, an acute change of the ratio of
polyunsaturated to saturated fatty acids in the diet will have a transient effect on the
postprandial metabolome, whereas a chronic change will lead to a sustained read-
justment of the expression of many genes, resulting in altered fasting levels of plasma
cholesterol fractions, haemostatic functions, insulin responsiveness, and the like.
Phenotype will also influence urinary metabolomic profiles in that age, sex, height,
weight, and the endocrine environment all shape the constituents of urine.
There is no doubt from an extensive literature that chronic changes in the human
diet significantly alter metabolic homeostasis and that the altered metabolic homeo-
stasis will in turn alter the acute response to a nutrient load. There is, however, a
major difference in the relative effects of food and nutrients on urinary and plasma
metabolomic profiles. In the case of plasma, nutrients are preferentially retained
unless fairly high renal thresholds are exceeded. In contrast, xenobiotics, which
abound in the nonnutrient element of food, are preferentially secreted into urine. An
extensive review of the metabolism of plant chemicals or phytochemicals shows an
average half-life for excretion into urine of about 8 h, with a maximum of about 20
h. These phytochemicals do not enter normal metabolic pathways, but they do
operate as receptorligands, triggering a number of metabolic responses such as is
encountered with onion peeling, or the consumption of coffee, or the habit of
cigarette smoking.
In effect, the plasma metabolomic profile is dominated by the body composition
dimension of phenotype and by metabolites of a multitude of pathways. It can be
altered by diet, but only after chronic exposure has adjusted gene expression and,
thus, the management of metabolic pathways. In the case of urine, phenotypic
9281_C003.fm Page 29 Thursday, July 26, 2007 7:05 PM
characteristics will play a significant role, but they will be subject to considerable
interference by recently ingested nonnutrients. In one study of urinary metabolomic
profiles in subjects consuming chamomile tea, a significant number of outliers were
characterized by high levels of trimethylamine oxide (TMAO) indicative of the recent
consumption of seafood. A recent study by the authors has shown that strict and
total control over food choice for 24 h prior to biofluid collection has no effect on
reducing variability in plasma or saliva, but does have a very significant effect in
reducing between-person variability in urinary metabolomic profiles. The authors
speculated that this is best explained by the reduction in between-person variability
in nonnutrient intake rather than any change in metabolic homeostasis. Thus, in
human nutrition, urinary collections might have to be collected with strict control
over the prior 24-h food choice, an issue for discussion by international groups setting
standards for this area of research (www.nugo.org).
original databases appropriate for the given phenotype to ascertain where in the spec-
trum of dietary variability the unknown lies. Thus, the dietary profile of the subject
would be characterized. This does not quantify food and nutrient intake patterns and
will not replace such areas of investigation. However, it does overcome the enormous
bottleneck that exists in dietary phenotyping, namely, energy underreporting, in
which subjects misreport the consumption of a food or, if consumed, its frequency
of consumption and portion size. In this scenario, the true overall pattern of food
and nutrient intake will have been ascertained, and quantification at that point may
lead to reduced manipulation of the truth, because the truth will have been ascertained
by objective modern technologies.
A parallel question that will arise is whether, once a metabolomic profile has
been established, any inference might be made as to the biological future of someone
with that metabolic profile, such as their likely metabolic response, favorable or
otherwise, to some change in diet. Until recently, this has been seen as belonging to
the realm of nutrigenetics, which studies how common genetic variants (single-
nucleotide polymorphisms) influence the responsiveness to dietary change. However,
a recent study in rats has shown that knowledge of the metabolic profile of an animal
prior to pharmacological intervention can accurately predict responsiveness to the
test drug. At first this seems somewhat unexpected, but on reflection, the metabolomic
profile is the ultimate signature of the product of phenotype as well as gene expression
and single-nucleotide polymorphism and, as such, is the universal biomarker.
that would not only identify all the metabolites of this human nutrition metabolome
but also quantify them in the various biofluids. The European Nutrigenomics Organ-
isation (www.nugo.org) is committed to defining the human nutrition metabolome
and to creating an open-source database on the biological details of all compounds
in the human nutritional metabolome (www.nugowiki.org). This database will link
with that of the Human Metabolome Database (www.metabolomics.ca), which is a
major project of Genome Canada. The basic philosophy is that one day we will have
access to a list of all nutrients present in plasma, for example, that are important in
human nutrition and the pathways they regulate. Additionally, the factors controlling
the balance of these nutrients, such as genetic factors, will be described in detail,
and the normal and abnormal concentration ranges will be known. Thus, normal
values for plasma cholesterol are well established, and so we can recognize low and
high levels. In an ideal world we could recognize normality for the concentrations
of the totality of plasma nutrients and related compounds in a metabolomic signature
and, if that can be achieved, we should be able to recognize metabolic perturbations
at an early stage.
KEY READINGS
Bollard, M.E., Stanley, E.G., Lindon, J.C., Nicholson, J.K., and Holmes, E. NMR-based meta-
bonomic approaches for evaluating physiological influences on biofluid composition.
NMR Biomed 18: 14362, 2005.
German, J.B., Hammock, B.D., and Watkins, S.M. Metabolomics: building on a century of
biochemistry to guide human health. Metabolomics 1: 39, 2005.
Gibney, M.J., Walsh, M.C., Brennan, L., Roche, H.M., German, B., and van Ommen, B.
Metabolomics in human nutrition: opportunities and challenges. Am J Clin Nutr 82:
497503, 2005.
9281_C003.fm Page 32 Thursday, July 26, 2007 7:05 PM
Keun, H.C. Metabonomic modeling of drug toxicity. Pharmacol Ther 109: 92106, 2006.
Lamers, R.J., van Nesselrooij, J.H., Kraus, V.B., Jordan, J.M., Renner, J.B., Dragomir, A.D.,
Luta, G., van der Greef, J., and DeGroot J. Identification of a urinary metabolite
profile associated with osteoarthritis. Osteoarthritis Cartilage 13:7628, 2005.
Nicholson, J.K., Holmes, E., and Wilson, I.D. Gut microorganisms, mammalian metabolism
and personalized health care. Nat Review Microbiol 3: 4318, 2005.
9281_C004.fm Page 33 Wednesday, July 18, 2007 1:49 PM
4 Nutrition, Genomics,
and Cardiovascular
Disease Risk
Jose M. Ordovas and Dolores Corella
CONTENTS
4.1 INTRODUCTION
The existence in the population of hyper- and hyporesponders to dietary modification
and to any other therapeutic approach is a well-known phenomenon. Whereas some
individuals are able to improve their plasma lipid profile or body weight using the
current dietary guidelines, others are unable to gain any significant benefit. The basis
for these interindividual differences are complex, and current knowledge suggests
that it is partially genetic. During the last two decades, evidence of interaction
between traditional lipid metabolism candidate genes, dietary components, and
cardiovascular risk factors (i.e., plasma lipids and obesity-related traits) has been
slowly gaining momentum. Several experimental approaches have been used for this
research, including observational and interventional studies. However, the replication
among studies is still limited, mostly driven by less-than-optimal experimental
designs. Current studies are starting to incorporate experimental approaches (i.e.,
increased coverage of the genetic variation at each locus) that could provide more
solid results. However, other limitations, such as small populations that limit the
statistical power especially to examine higher-level interactions (genegenediet
behavioral factors) have yet to be incorporated in nutrigenetic studies. Moreover,
the current quality of the dietary and behavioral information adds another major
33
9281_C004.fm Page 34 Wednesday, July 18, 2007 1:49 PM
limitation, specifically for observational studies. These needs can be met through
the collaboration of experts in the different fields involved, ranging from basic
scientists to computational biologists and behavioral scientists. Once more solid
evidence is achieved, nutrigenomics could be applied toward primary prevention
and treatment of cardiovascular diseases in the general population and subjects at
high risk.
The effect of dietary changes on cardiovascular disease (CVD)-relevant traits
(i.e., plasma lipid measures, glucose levels, body weight, and blood pressure) differs
significantly among individuals [1]. Some individuals appear to be relatively insen-
sitive (hyporesponders) to dietary intervention, whereas others display enhanced
sensitivity (hyperresponders) [1]. This phenomenon has been more extensively
researched in relation to changes in dietary fat and plasma lipid concentrations for
the prevention of CVD compared to other pathological conditions. Although common
knowledge associates low-fat diets with reductions in total and plasma low-density
lipoprotein cholesterol (LDL-C), the clinical evidence shows dramatic interindividual
differences in response and may be one of the underlying causes of the limited
success of dietary recommendations in the prevention of diseases observed by recent
randomized clinical trials [2].
A growing body of data supports the hypothesis that the interindividual vari-
ability in response to dietary modification is determined by genetic factors [3].
Indirect evidence comes from the general observation that the phenotypic response
to diet is determined partly by the baseline value of the phenotype, which is itself
affected by genetic factors [1]. Some of the main challenges are the following:
(1) how to uncover and elucidate the many potential genediet interactions; (2) how
to include in the experimental designs the ability to test for genegenediet inter-
actions; (3) how to investigate more complex and realistic scenarios, including the
contributions of other behavioral factors (i.e., physical activity, smoking, socioeco-
nomic status, traditions, and social environment); and (4) how to collect reliable and
standardized information of relevant covariates [4]
Several studies have found specific candidate genes to be associated with the
variability in response of LDL-C levels in response to changes in dietary fat, but so
far, the humbling reality is that these findings have been highly inconsistent. These
conflicting outcomes reflect both the complexity of the mechanisms involved in
dietary responses and the limitations of the current experimental designs used to
address these problems, some of which are due to the challenges listed in the previous
paragraph. In addition, we need to keep in mind that the practical translation to the
public of any nutrigenetic finding needs to be supported by a more holistic approach
to health rather than applying the tunnel-vision approach of focusing exclusively on
the changes in one particular risk factor. To this effect, it is true that low-fat diets
can result in reduced plasma LDL-C levels, but also in lowering of HDL-C and
increased triacylglycerol (TAG) concentrations, which may be particularly harmful
for some persons. For example, it has been shown that individuals with a predom-
inance of small, dense LDL particles (subclass pattern B), a phenotype that is
associated with an increased risk of coronary heart disease, benefit more from a
low-fat diet [5] than do those with the subclass pattern A (larger LDL). A significant
proportion of the latter group unexpectedly exhibited a more atherogenic pattern B
9281_C004.fm Page 35 Wednesday, July 18, 2007 1:49 PM
subclass after consuming a low-fat diet. Intervention studies are increasingly focus-
ing on the interindividual differences in response to diet rather than on the mean
effect analyzed for a population. Moreover, new evidence indicates that the variabil-
ity in response is an intrinsic characteristic of the individual, rather than being the
result of differing dietary compliance with the experimental protocols. Jacobs et al.
[6] found that individual TAG responses to a high-fat or to a low-fat diet were vastly
different, suggesting that many patients with hypertriglyceridemia are not treated
optimally if general advice for either a low-fat or a high-fat diet is given. Under-
standing the mechanisms driving this variation will not only allow us to better grasp
the biology of dietary response but also to identify individuals who can benefit from
a particular dietary intervention.
and glucose metabolism [8]. PPARs are among the best-studied fatty-acid-regulated
nuclear receptors [9]. After uptake into target cells, a subset of them are transported
to the nucleus in association with fatty acid binding proteins (FABPs), which facil-
itates their interaction with PPARs. Several PPAR subtypes have been described.
PPAR-alpha (PPARA) plays a key role in lipid oxidation and inflammation, whereas
PPAR-gamma (PPARG) is involved in cell (adipocyte) differentiation, glucose lipid
storage, and inflammation. PPAR-delta (PPARD, also known as PPAR-beta) may
play an important role in development, lipid metabolism, and inflammation [10].
Many of the previously published nutrigenetic [i.e., single-gene/single-nucleotide
polymorphisms (SNPs)] studies focused on genes that are the subject of regulation
by PPARs and other nuclear receptors [11]. Polymorphisms in promoter regions of
these genes may disrupt, or at least alter, the communication with these transcription
factors, which would have significant consequences in a persons response to dietary
factors that are ligands (i.e., PUFAs) of the transcription factors. It is also obvious
that polymorphisms within the transcription factors themselves will have a significant
impact on the way each one of us responds to dietary components [1214].
The quantitative evidence for genediet interactions between common SNPs at
candidate genes and dietary factors related to lipid metabolism is increasing. How-
ever, we reiterate that caution is needed before applying these results to clinical
practice for three primary reasons: (1) the meaning of statistically significant
results is subject to differing interpretations and often depends on the study design,
(2) many initial genenutrientphenotypes associations were not replicated in sub-
sequent studies, and (3) gene variations may influence phenotypes differently in
individuals from different ancestral backgrounds owing to genegene (epistatic)
interactions.
modeling methods may have an even more pronounced effect in diverse population
groups. So, it is not surprising that when different study populations are combined with
different analytical techniques, there are so many inconsistencies in the literature [14].
So far, most of the initial descriptions of genediet interactions have been based
on observational studies; regardless of the apparent solidity of any initial reported
finding, every genediet interaction needs to go through a series of verification steps,
which should at least include (1) its replication on several other properly sized
populations, (2) its testing on an interventional study in people preselected on the
basis of their genotypes for the SNP under consideration, and (3) additional in vitro
and ex vivo tests of functionality to provide the mechanistic knowledge that will
support the reported statistical interactions. Moreover, genotype-nutrient-phenotype
analyses may be improved by determining ancestral backgrounds of each study
participant. These additional data are necessary because SNPs may be expressed
differently among individuals of differing ethnicities because of varying gene-gene
and gene-nutrient interactions.
The enumeration of those studies focusing on the interaction between genetic
variants at candidate genes, nutrients, and CVD risk factors have been the subject
of recent reviews [7,15,16], and we will not reproduce their contents in this chapter.
Rather, we will focus on capturing the current trends in nutrigenetic research.
We will begin by focusing on one of the rising genes from the last few years:
apolipoprotein A5 (APOA5). Totally unknown until 2001, APOA5 has become a
major player in lipoprotein metabolism and specifically in triglyceride-rich lipopro-
tein (TRL) metabolism [17]. Five common APOA5 SNPs have been reported in
several populations: 1131T > C, 3A > G, 56C > G IVS3 + 476G > A, and 1259T >
C, and these APOA5 gene variants have been associated with increased TAG con-
centrations [1821]. With the exception of the 56C > G SNP, the SNPs are reported
to be in significant linkage disequilibrium [22]. Despite the relative good consistency
with plasma TAG concentrations, the association between APOA5 SNPs (or haplo-
types) and CVD risk is still controversial [19,23,24]. Learning from previous expe-
rience, it is reasonable to implicate gene environment interactions modulating the
association with CVD risk factors as well as with the disease risk itself. Consistent
with this hypothesis, several intriguing interactions are beginning to emerge. In the
Framingham Heart Study, we have reported a genediet interaction between the
APOA5 gene variation and the polyunsaturated fatty acids (PUFA) in relation to
plasma lipid concentrations and lipoprotein particle size [25]. Furthermore, we have
demonstrated that obesity modulates the effect of the APOA5 gene variation in
carotid intimal medial thickness (IMT), a surrogate measure of atherosclerosis bur-
den. This association remained significant even after the adjustment for TG [26].
More recently, we have found a genediet interaction between the APOA5 gene and
total fat intake in relation to body mass index (BMI), overweight, and obesity risk
[27]. The homogeneity of this interaction was detected in both men and women and
followed a dose-response relationship. In comparison with wild-type subjects, the
carriers of the variant C allele at the 1131T > C SNP appeared to be protected
from an increase in body weight when consuming more dietary fat. Interactions
between BMI and APOA5 had been previously reported by Aberle et al. [28]. More-
over, some interesting peculiarities are emerging that deserve further examination.
9281_C004.fm Page 38 Wednesday, July 18, 2007 1:49 PM
One relates to the specificity of the interactions in terms of the location of the APOA5
tagging SNPs, one in the promoter region (1131T > C) and the other affecting the
primary structure of the protein (56C > G). We have reported that the complex
interactions between APOA5 SNPs, dietary intake, BMI, and IMT appear to be
related to the promoter polymorphism, whereas the APOA5 56C > G SNP appears
to associate with the different traits independently of environmental triggers [2527].
The second aspect to consider is the specificity of the interactions in terms of dietary
fat, being different for saturated, monounsaturated, and polyunsaturated n-6 and n-3.
We investigated the interaction between these APOA5 SNPs and dietary fat in
determining plasma fasting TGs, remnant-like particle (RLP) concentrations, and
lipoprotein particle size in Framingham Heart Study participants. Significant
genediet interactions between the 1131T > C SNP and PUFA intake were found
in determining fasting TGs, RLP concentrations, and lipoprotein particle size, but
these interactions were not found for the 56C > G polymorphism [25]. The 1131C
allele was associated with higher fasting TGs and RLP concentrations in only the
subjects consuming a high-PUFA diet (>6% of total energy), regardless of gender.
Interestingly, when we further analyzed the effects of n-6 and n-3 fatty acids, we found
that the PUFAAPOA5 interactions were specific for dietary n-6 fatty acids. Thus,
higher n-6 (but not n-3) PUFA intake increased fasting TGs, RLP concentrations, and
VLDL size and decreased LDL size in APOA5 1131C carriers, suggesting that n-6
PUFA-rich diets are related to a more atherogenic lipid profile in these subjects. This
could shed some light into the data coming from postprandial studies [2933] that will
be discussed in the following text. Similar specificities in terms of SNP, exclusive for
the 1131T > C, and dietary fat, primarily monounsaturated fat, were found in relation
to the interaction between fat intake and BMI within the Framingham Study [27].
These observations suggest some new hypotheses regarding some of the
observed geographical differences in CVD risk as well as the changes in risk
observed following Westernization of lifestyles. In this regard, it is important to pay
attention to the differences in allele frequencies of the APOA5 SNPs identified so
far. These frequencies appear to be higher in Asian compared to White populations
[18]. If the interactions observed in Framingham apply to other geographical areas,
we should expect that an increased fraction of the Asian population will be placed
at higher CVD risk because of the current trend toward higher dietary fat intakes,
especially if they are driven by increases in PUFA n-6. Moreover, we found in the
Framingham Study that some individuals (carriers of the C allele at the 1131T >
C SNP) may consume a high percentage of fat, especially MUFA, in their habitual
diet without adversely affecting their BMI and their risk of overweight and obesity [27].
The studies described earlier provide evidence of the current transition from the
traditional approach of studying one SNP per gene to a more comprehensive exam-
ination using multiple SNPs and haplotypes. Moreover, they represent a sample of
the exploding interest for obesity-related phenotypes and their complications. Given
the current public attention to this problem, it is not surprising that increasing number
of researchers aim their focus on the investigation of genediet and obesity-related
parameters.
Some of the recent reports examine obvious but previously ignored candidate
genes. This is the case with the loci encoding the carnitine palmitoyltransferase I
9281_C004.fm Page 39 Wednesday, July 18, 2007 1:49 PM
(CPT1), a key enzyme in beta-oxidation of fatty acids. Robitaille et al. [34] screened
the genes for CPT1A (11q13) and CPT1B (22qter) for common mutations in a
French-Canadian population and selected nine variants within CPT1B and one
variant within CPT1A for further analyses in relation with obesity based on the
minor allele frequency (>10%) or on potential functional impact. A significant
association between obesity phenotypes (BMI, weight, and waist girth) and CPT1B
c.282-18C > T and p.E531K variants was observed. When subjects were divided
into six groups according to p.E531K genotypes and further on the basis of fat using
the median value as a cutoff point (34.4% of energy), BMI, weight, and waist girth
were higher in E531/K531 on a high-fat diet compared to E531/K531 subjects under
a low-fat diet. There was no difference among E531/E531 and K531/K531. This
interaction is complex to interpret as the statistically significant interaction was
driven by heterozygotes, and no differences were observed for the homozygous at
each of the extremes. Similar results were obtained with the CPT1A p.A275T variant
as BMI and waist girth were higher in A275/A275 on a high-fat diet compared to
A275/A275 subjects on a low-fat diet. Among carriers of the T275 allele, obesity
indices were not affected by fat intake. These findings suggest that obesity-related
traits might be modulated by an interaction between CPT1 variants and fat intake,
but their interpretation is not straightforward.
Other studies were initially designed to examine the genetics of obesity and to
identify significant and valuable nutritional interactions. This was the case with
NUGENOB [35]. This study examined 42 SNPs at 26 candidate genes for obesity
in 549 adult obese women recruited from eight European centers in a case-only
study. The nutritional variables assessed in this study were the dietary fiber intake
(grams per day), the ratio of dietary polyunsaturated fat to saturated fat (P:S ratio),
and the percentage of energy derived from fat in the diet as calculated from a weighed
3-d food record (%E). Some weak, confirmatory evidence for genotype-by-nutrient
interactions in obesity was detected in this case-only study [i.e., hepatic lipase gene
(LIPC); adiponectin (ADIPOQ) and PPARG3]. Although this design was initially
considered useful for detecting interactions, the authors state that few genotype-by-
nutrient interactions have been suggested in obese European women after completing
the initial goals of the study.
Our own studies focused on the gene for human perilipin (PLIN), which is on
chromosome 15q26, in the region of a linkage locus for diabetes, hypertriglyceridemia,
and obesity. In a study to examine the regulation of lipolysis in obese and nonobese
women [36], it was found that obese women exhibited two- to fourfold higher basal
rates of lipolysis in fat cells obtained through a biopsy in the abdominal subcutaneous
area. Similar differences in lipolysis rates were observed following noradrenaline
induction. It was also noted that adipocyte perilipin content was reduced in obese
women. This study also examined the effect of a polymorphism (11482G > A) in
intron 6 of the perilipin locus in relation to perilipin content as well as the rates of
lipolysis. Adipose tissue from women who were homozygous for the A allele at
position 11482 exhibited 50 to 100% higher rates of lipolysis and 80% lower perilipin
content. Following this study, several polymorphisms at the perilipin locus have been
examined for potential associations with BMI or obesity risk. We have examined
four SNPs (PLIN1: 6209T > C, PLIN4:11482G > A, PLIN5: 13041A > G, and
9281_C004.fm Page 40 Wednesday, July 18, 2007 1:49 PM
It is unclear at this time why no association was observed in French and Chinese
ethnic groups despite being reasonably powered to detect an association, if present.
If indeed complex traits such as obesity are a consequence of interactions between
genetic and environmental factors [41], such geneenvironment interactions could
explain the discrepant findings among populations. It could simply be that the French
and Chinese are exposed to different environments when compared with whites in
Spain or California, and Malays and Asian Indians in Singapore. Do such gene
environment interactions operate in relation to the association between polymor-
phisms at the perilipin locus and obesity? In fact, the interaction between polymorphisms
at this locus and gender has already been described in whites and some of the Asian
groups. Recently, one study [42] has reported that individuals carrying the A allele
for the 11482G > A polymorphism are resistant to weight loss with caloric restriction.
Another [43] reported resistance to weight gain following treatment with the PPARG
agonist rosiglitozone in individuals carrying this polymorphism. Therefore, over-
weight and obese individuals carrying this allele may not benefit from traditional
dietary approaches to losing weight, and alternative approaches may be needed. In
this regard, an interesting observation was made regarding the potential regulatory
effects of a plant extract on perilipin, hormone-sensitive lipase, and other parameters
of lipid metabolism in obese women [44].
Up to this point, we have dealt primarily with associations between polymor-
phisms at the perilipin locus and their associations with the degree of obesity per
se, as opposed to the downstream effects of obesity, such as insulin resistance. Jang
et al. [45] studied 177 obese Korean men and women who were treated with caloric
restriction (300 kcal per day) and found that the presence of the A11482 allele was
associated with greater weight loss in response to caloric restriction and that this
weight loss was primarily in the visceral fat compartment. Because visceral obesity
is generally associated with increased plasma-free fatty acid concentrations, one
may expect a greater reduction in plasma-free fatty acids in these individuals. Instead,
these individuals experienced an increase in plasma-free fatty acids despite a greater
reduction in visceral fat mass.
It is important to remember that fatty acids serve not only as a source of energy
but also as signaling molecules. Free fatty acids result in impaired glucose homeostasis
and may increase the risk of type 2 diabetes mellitus. To test the hypothesis that
dietary fat may interact with polymorphisms at the perilipin locus to modulate dia-
betes-related traits, we reexamined data from the Singapore population, taking into
consideration dietary macronutrient intake [46]. We found evidence of an interaction
between dietary fat (specifically saturated fat) intake, polymorphisms at the perilipin
locus (11482G > A and 114995A > T), and insulin resistance. Greater intake of energy
from saturated fats was associated with increased insulin resistance among individuals
who were homozygous for the rare alleles but not individuals who carried the common
allele. This effect was independent of BMI and consistent with the associations with
anthropometric measures; these interactions were observed only in women.
Overall, observational studies have provided substantial support to the concept of
genediet interactions modulating cardiovascular risk factors; however, very few find-
ings have been consistently replicated; among several concerns related to this approach,
we want to underscore the need for more reliable assessment of dietary intake.
9281_C004.fm Page 42 Wednesday, July 18, 2007 1:49 PM
ACKNOWLEDGMENTS
This work was supported by NIH grants HL72524, HL54776, and DK075030 and
by contracts 53-K06-5-10 and 58-1950-9-001 from the U.S. Department of Agri-
culture Research Service. (JMO), and contracts 53-K06-5-10 and 58-1950-9-001
from the U.S. Department of Agriculture Research Service (JMO), and the Spanish
Ministerio de Educacin y Ciencia (PR2006-0258) and the Spanish Ministerio de
Sanidad (CB06/03/0035).
SUGGESTED LECTURES/URLS
www.nugo.org.
http://www.athero.org/.
http://www.isnn.info/.
http://www.nutragenomics.com.
9281_C004.fm Page 45 Wednesday, July 18, 2007 1:49 PM
REFERENCES
1. Katan, M. B.; Beynen, A. C.; de Vries, J. H.; et al. Existence of consistent hypo- and
hyperresponders to dietary cholesterol in man. American journal of epidemiology
123: 221234; 1986.
2. Prentice, R. L.; Caan, B.; Chlebowski, R. T. et al. Low-fat dietary pattern and risk
of invasive breast cancer: the Womens Health Initiative Randomized Controlled
Dietary Modification Trial. Jama 295: 629642; 2006.
3. Loktionov, A. Common gene polymorphisms and nutrition: emerging links with
pathogenesis of multifactorial chronic diseases (review). Journal of nutritional bio-
chemistry 14: 426451; 2003.
4. Greenwood, D. C.; Gilthorpe, M. S.; Cade, J. E. The impact of imprecisely measured
covariates on estimating gene-environment interactions. BMC medical research meth-
odology 6: 21; 2006.
5. Krauss, R. M. Dietary and genetic effects on low-density lipoprotein heterogeneity.
Annual review of nutrition 21: 283295; 2001.
6. Jacobs, B.; De Angelis-Schierbaum, G.; Egert, S.; et al. Individual serum triglyceride
responses to high-fat and low-fat diets differ in men with modest and severe hyper-
triglyceridemia. The Journal of nutrition 134: 14001405; 2004.
7. Ordovas, J. M.; Corella, D. Nutritional genomics. Annual review of genomics and
human genetics 5: 71118; 2004.
8. Jump, D. B. Fatty acid regulation of gene transcription. Critical reviews in clinical
laboratory sciences 41: 4178; 2004.
9. Clarke, S. D. The multi-dimensional regulation of gene expression by fatty acids:
polyunsaturated fats as nutrient sensors. Current opinion in lipidology 15: 1318; 2004.
10. Tobin, J. F.; Freedman, L. P. Nuclear receptors as drug targets in metabolic diseases:
new approaches to therapy. Trends in endocrinology and metabolism: TEM 17:
284290; 2006.
11. Mandard, S.; Muller, M.; Kersten, S. Peroxisome proliferator-activated receptor alpha
target genes. Cellular and molecular life sciences 61: 393416; 2004.
12. Soriguer, F.; Morcillo, S.; Cardona, F. et al. Pro12Ala polymorphism of the PPARG2
gene is associated with type 2 diabetes mellitus and peripheral insulin sensitivity in a
population with a high intake of oleic acid. The journal of nutrition 136: 23252330; 2006.
13. Tai, E. S.; Corella, D.; Demissie, S.; et al. Polyunsaturated fatty acids interact with the
PPARA-L162V polymorphism to affect plasma triglyceride and apolipoprotein C-III con-
centrations in the Framingham Heart Study. The journal of nutrition 135: 397403; 2005.
14. Nelson, T. L.; Fingerlin, T. E.; Moss, L. K.; et al. Association of the peroxisome
proliferator-activated receptor gamma gene with type 2 diabetes mellitus varies by
physical activity among non-Hispanic whites from Colorado. Metabolism: clinical
and experimental 56: 388393; 2007.
15. Corella, D.; Ordovas, J. M. Single nucleotide polymorphisms that influence lipid metab-
olism: interaction with dietary factors. Annual review of nutrition 25: 341390; 2005.
16. Ordovas, J. M.; Corella, D. Genes, diet and plasma lipids: the evidence from obser-
vational studies. World review of nutrition and dietetics 93: 4176; 2004.
17. Pennacchio, L. A.; Olivier, M.; Hubacek, J. A.; et al. An apolipoprotein influencing
triglycerides in humans and mice revealed by comparative sequencing. Science 294:
169173; 2001.
18. Lai, C. Q.; Tai, E. S.; Tan, C. E.; et al. The APOA5 locus is a strong determinant of
plasma triglyceride concentrations across ethnic groups in Singapore. Journal of lipid
research 44: 23652373; 2003.
9281_C004.fm Page 46 Wednesday, July 18, 2007 1:49 PM
19. Lai, C. Q.; Demissie, S.; Cupples, L. A.; et al. Influence of the APOA5 locus on
plasma triglyceride, lipoprotein subclasses, and CVD risk in the Framingham Heart
Study. Journal of lipid research 45: 20962105; 2004.
20. Talmud, P. J.; Hawe, E.; Martin, S.; et al. Relative contribution of variation within
the APOC3/A4/A5 gene cluster in determining plasma triglycerides. Human molecular
genetics 11: 30393046; 2002.
21. Talmud, P. J. Rare APOA5 mutations-Clinical consequences, metabolic and functional
effects: an ENID review. Atherosclerosis; 2007.
22. Pennacchio, L. A.; Olivier, M.; Hubacek, J. A.; et al. Two independent apolipoprotein
A5 haplotypes influence human plasma triglyceride levels. Human molecular genetics
11: 30313038; 2002.
23. Talmud, P. J.; Martin, S.; Taskinen, M. R.; et al. APOA5 gene variants, lipoprotein
particle distribution, and progression of coronary heart disease: results from the
LOCAT study. Journal of lipid research 45: 750756; 2004.
24. Dallongeville, J.; Cottel, D.; Montaye, M.; et al. Impact of APOA5/A4/C3 genetic
polymorphisms on lipid variables and cardiovascular disease risk in French men.
International journal of cardiology 106: 152156; 2006.
25. Lai, C. Q.; Corella, D.; Demissie, S.; et al. Dietary intake of n-6 fatty acids modulates
effect of apolipoprotein A5 gene on plasma fasting triglycerides, remnant lipoprotein
concentrations, and lipoprotein particle size: the Framingham Heart Study. Circula-
tion 113: 20622070; 2006.
26. Elosua, R.; Ordovas, J. M.; Cupples, L. A. et al. Variants at the APOA5 locus,
association with carotid atherosclerosis, and modification by obesity: the Framingham
Study. Journal of lipid research 47: 990996; 2006.
27. Corella, D.; Lai, C. Q.; Demissie, S.; et al. APOA5 gene variation modulates the
effects of dietary fat intake on body mass index and obesity risk in the Framingham
Heart Study. Journal of molecular medicine 85: 119128; 2007.
28. Aberle, J.; Evans, D.; Beil, F. U.; et al. A polymorphism in the apolipoprotein A5
gene is associated with weight loss after short-term diet. Clinical genetics 68:
152154; 2005.
29. Martin, S.; Nicaud, V.; Humphries, S. E.; Talmud, P. J. Contribution of APOA5
gene variants to plasma triglyceride determination and to the response to both fat
and glucose tolerance challenges. Biochimica et biophysica acta 1637: 217225;
2003.
30. Kim, J. Y.; Kim, O. Y.; Koh, S. J.; et al. Comparison of low-fat meal and high-fat
meal on postprandial lipemic response in non-obese men according to the 1131T >
C polymorphism of the apolipoprotein A5 (APOA5) gene (randomized cross-over
design). Journal of the American College of Nutrition 25: 340347; 2006.
31. Jang, Y.; Kim, J. Y.; Kim, O. Y.; et al. The 1131T>C polymorphism in the
apolipoprotein A5 gene is associated with postprandial hypertriacylglycerolemia;
elevated small, dense LDL concentrations; and oxidative stress in nonobese Korean
men. The American journal of clinical nutrition 80: 832840; 2004.
32. Masana, L.; Ribalta, J.; Salazar, J.; et al. The apolipoprotein AV gene and diurnal
triglyceridaemia in normolipidaemic subjects. Clinical Chemistry and Laboratory
Medicine 41: 517521; 2003.
33. Moreno, R.; Perez-Jimenez, F.; Marin, C.; et al. A single nucleotide polymorphism
of the apolipoprotein A-V gene 1131T>C modulates postprandial lipoprotein metab-
olism. Atherosclerosis 189: 163168; 2006.
34. Robitaille, J.; Houde, A.; Lemieux, S.; et al. Variants within the muscle and liver
isoforms of the carnitine palmitoyltransferase I (CPT1) gene interact with fat intake
9281_C004.fm Page 47 Wednesday, July 18, 2007 1:49 PM
51. van Ommen, B.; Stierum, R. Nutrigenomics: exploiting systems biology in the nutri-
tion and health arena. Current opinion in biotechnology 13: 517521; 2002.
52. Kaput, J.; Ordovas, J. M.; Ferguson, L.; et al. The case for strategic international
alliances to harness nutritional genomics for public and personal health. The British
journal of nutrition 94: 623632; 2005.
9281_C005.fm Page 49 Wednesday, July 18, 2007 1:50 PM
5 Nutrigenomics
and Chronic Inflammation
Lynnette R. Ferguson and Martin Philpott
CONTENTS
49
9281_C005.fm Page 50 Wednesday, July 18, 2007 1:50 PM
Pro-inflammatory genes
Infiltration/ Inflamed and mediators
recruitment tissue (COX-2, INOS, IL)
Inflammatory
immune cells
Pro-inflammatory mediators
(e.g., TNF-, IL-1)
FIGURE 5.1 Overview of the process of chronic inflammation, resulting in damage to host
tissues and activation of proinflammatory genes.
9281_C005.fm Page 51 Wednesday, July 18, 2007 1:50 PM
transcription factors such as nuclear factor-B (NF-B) and AP-1. Although ROS
and RNS may act directly to produce damage to DNA, lipid, or proteins, they also
commonly act through secondary products such as the peroxidation product, 4-
hydroxy-2-nonenal. NF-B, a family of ubiquitously expressed transcription factors,
regulates the expression of genes encoding proteins essential for stress response,
maintenance of intercellular communication, cell cycle control, and apoptosis.
disease. The bulk of functional polymorphisms thus far identified occur in upstream
promoter sequences, leading to changes in the expression of the respective pro- or
anti-inflammatory gene products. Interleukin Genetics is an example of a company
that focuses attention on polymorphisms in a single gene. As pointed out by Kornman,8
variants in the genes for the family of interleukin 1 (IL-1) proteins often lead to
modified inflammatory response, and may result in increased susceptibility to a range
of diseases, including coronary artery disease, AD, gastric cancer, and periodontitis.
Similarly, they may modify the onset and progression of the deterioration associated
with aging.
A number of other such loci have also been associated with multiple disorders.
For example, polymorphisms in the cytotoxic T-lymphocyte-associated 4 (CTLA-4)
gene have been associated with autoimmune thyroidosis9 and type I diabetes melli-
tus,10 and have been suggested as a general susceptibility factor in autoimmune
disease.11 Caspase recruitment domain-containing protein 15 (CARD15) functions
as an intracellular receptor for bacteria, signaling the presence of pathogens by the
production of NF-B. Ogura et al.12 provided evidence for certain polymorphisms
in CARD15 significantly increasing susceptibility to Crohns disease in humans,
and others have more recently shown a key involvement in colorectal cancer.13 The
products of the HLA (human leukocyte antigen; HLA) family of genes play a critical
role in regulation of the immune response.14 There are recognized associations
between several different HLA polymorphisms and a range of inflammatory diseases.
Thus, individuals carrying the HLA-DQA1*0501, DQB1*0201 genotype show a
more than 200-fold increased risk of developing celiac disease, whereas other related
factors may be involved in the development of malignant lymphoma in individuals
who already have celiac disease. Similar considerations pertain to the development
of rheumatoid arthritis.14
The recognition of ROS involvement in chronic inflammation and tissue destruc-
tion suggests that either genes or nutrients involved in regulating antioxidant
response will play a significant role in modulating the development and progression
of inflammation. At a cellular level, redox homeostasis is generally achieved through
the balance of ROS generation with mechanisms for their removal, including dietary
antioxidants and intracellular antioxidant defense mechanisms. Chen and Kunsch15
describe the antioxidant response element (ARE) as one of a class of cytoprotective
regulatory genes. This element is located in the regulatory regions of a number of
genes, including phase II detoxification enzymes proteins such as glutathione-S-
transferases and gamma-glutamylcysteine synthase. It is bound by the transcription
factor Nrf2, leading to activation of the expression of other leucine-zipper-containing
transcription factors that have profound effects on immune and inflammatory
responses. Factors that decrease expression of these genes enhance inflammatory
responses to ROS, and also enhance the progression and development of autoimmune
disease.
There are several polymorphisms in the PS-1 and APP genes that increase
production of Abeta (1-42), and are likely to be the major causes of early onset
familial AD.16 High levels of ROS and RNS, and increased vulnerability to oxidative
stress appear to be a consequence of elevated levels of Abeta (1-42), and may also
contribute significantly to neuronal apoptosis and death in familial early-onset AD.17
9281_C005.fm Page 53 Wednesday, July 18, 2007 1:50 PM
NSAIDS
Amino Acids Omega-3 Fatty Acids
Micronutrients Probiotics/Prebiotics
Pro-inflammatory genes
Infiltration/ and mediators
Inflamed
recruitment (COX-2, INOS, IL)
tissue
Inflammatory
immune cells
Pro-inflammatory mediators
(e.g., TNF-, IL-1)
FIGURE 5.2 Key points within the process of chronic inflammation where nutrients and
nonnutrients interact, reducing the likelihood of chronic inflammation leading to a disease
end point. Various micronutrients and amino acids are required for an essential early inflam-
matory response, whereas diverse nutrients and nonnutrients can inhibit later events and reduce
concomitant damage to host tissues during chronic inflammation.
ACKNOWLEDGMENTS
We thank the Auckland Cancer Society, The Cancer Society of New Zealand, and The
Foundation for Research, Science and Technology (New Zealand) for funding support.
KEY READINGS
Ferguson, L. R., Karunasinghe, N., and Philpott, M., Susceptibility to cancer and DNA damage
to lymphocytes: can it be counteracted by dietary selenium?, in Nutritional Biotech-
nology in the Feed and Food Industries: Proceedings of Alltechs Twenty Second
Annual Symposium, Lyons, T.P., Jacques, K.A., and Hower, J.M., Eds., Nottingham
University Press, Nottingham, 2006, p. 143.
Kundu, J. K. and Surh, Y. -J., Breaking the relay in deregulated cellular signal transduction as
a rationale for chemoprevention with anti-inflammatory phytochemicals, Mutat. Res.,
591, 123, 2005.
Philpott, M. and Ferguson, L. R., Immunonutrition and cancer, Mutat. Res., 551, 29, 2004.
Yamada, R. and Ymamoto, K., Recent findings on genes associated with inflammatory disease,
Mutat. Res., 573, 136, 2005.
REFERENCES
1. Dobrovolskaia, M. A. and Kozlov, S. V., Inflammation and cancer: when NF-kappaB
amalgamates the perilous partnership, Curr. Cancer Drug Targets, 5, 325, 2005.
2. Ditsworth, D. and Zong, W. X., NF-kappaB: key mediator of inflammation-associated
cancer, Cancer Biol. Ther., 3, 1214, 2004.
3. Greten, F. R. et al., IKKbeta links inflammation and tumorigenesis in a mouse model
of colitis-associated cancer, Cell, 118, 285, 2004.
4. Luo, J. L. et al., Inhibition of NF-kappaB in cancer cells converts inflammation-
induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression,
Cancer Cell, 6, 297, 2004.
5. Pikarsky, E. et al., NF-kappaB functions as a tumour promoter in inflammation-
associated cancer, Nature, 431, 461, 2004.
6. Andreakos, E., Foxwell, B., and Feldmann, M., Is targeting Toll-like receptors and
their signaling pathway a useful therapeutic approach to modulating cytokine-driven
inflammation?, Immunol. Rev., 202, 250, 2004.
9281_C005.fm Page 58 Wednesday, July 18, 2007 1:50 PM
7. Yamada, R. and Ymamoto, K., Recent findings on genes associated with inflammatory
disease, Mutat. Res., 573, 136, 2005.
8. Kornman, K. S., Interleukin 1 genetics, inflammatory mechanisms, and nutrigenetic
opportunities to modulate diseases of aging, Am. J. Clin. Nutr., 83, 475S, 2006.
9. Yanagawa, T. et al., CTLA-4 gene polymorphism associated with Graves disease in
a Caucasian population, J. Clin. Endocrinol. Metab., 80, 41, 1995.
10. Marron, M. P. et al., Insulin-dependent diabetes mellitus (IDDM) is associated with
CTLA-4 polymorphisms in multiple ethnic groups, Human Mol. Genet., 6, 1275, 1997.
11. Kristiansen, O. P., Larsen, Z. M., and Pociot, F., CTLA-4 in autoimmune diseasesa
general susceptibility gene to autoimmunity? Genes Immun., 1, 170, 2000.
12. Ogura, Y. et al., A frameshift mutation in NOD2 associated with susceptibility to
Crohns disease, Nature, 411, 603, 2001.
13. Roberts, R. L. et al., Caspase recruitment domain-containing protein 15 mutations in
patients with colorectal cancer, Cancer Res., 66, 2532, 2006.
14. Howell, W. M., Calder, P. C., and Grimble, R. F., Gene polymorphisms, inflammatory
diseases and cancer, Proc. Nutr. Soc., 61, 447, 2002.
15. Chen, X. L. and Kunsch, C., Induction of cytoprotective genes through Nrf2/antiox-
idant response element pathway: a new therapeutic approach for the treatment of
inflammatory diseases, Curr. Pharm. Des., 10, 879, 2004.
16. Walker, D. G. et al., Gene expression changes by amyloid beta peptide-stimulated
human postmortem brain microglia identify activation of multiple inflammatory pro-
cesses, J. Leukoc. Biol., 79, 596, 2006.
17. Mohammad Abdul, H. et al., Mutations in amyloid precursor protein and presenilin-
1 genes increase the basal oxidative stress in murine neuronal cells and lead to
increased sensitivity to oxidative stress mediated by amyloid beta-peptide (1-42), HO
and kainic acid: implications for Alzheimers disease, J. Neurochem., 96, 1322, 2006.
18. Prokunina, L. et al., A regulatory polymorphism in PDCD1 is associated with sus-
ceptibility to systemic lupus erythematosus in humans, Nat. Genet., 32, 666, 2002.
19. Nielsen, C. et al., Association of a putative regulatory polymorphism in PDCD1 with
susceptibility to type 1 diabetes, Tissue Antig., 62, 492, 2003.
20. Hu, Y. J. and Diamond, A. M., Role of glutathione peroxidase 1 in breast cancer: loss
of heterozygosity and allelic differences in the response to selenium, Cancer Res.,
63, 3347, 2003.
21. Ferguson, L. R., Karunasinghe, N., and Philpott, M., Susceptibility to cancer and
DNA damage to lymphocytes: can it be counteracted by dietary selenium?, in Nutri-
tional Biotechnology in the Feed and Food Industries: Proceedings of Alltechs Twenty
Second Annual Symposium, Lyons, T.P., Jacques, K.A., and Hower, J.M., Eds., Not-
tingham University Press, Nottingham, 2006, p. 143.
22. Curran, J. E. et al., Genetic variation in selenoprotein S influences inflammatory
response, Nat. Genet., 37, 1234, 2005.
23. Philpott, M. and Ferguson, L. R., Immunonutrition and cancer, Mutat. Res., 551, 29, 2004.
24. Bao, B. et al., Zinc modulates mRNA levels of cytokines, Am. J. Phys. Endo.
Metab., 285, E1095, 2003.
25. Halliwell, B., Oxidative stress, nutrition and health. Experimental strategies for opti-
mization of nutritional antioxidant intake in humans, Free Rad. Res., 25, 57, 1996.
26. DOdorico, A. et al., Reduced plasma antioxidant concentrations and increased oxi-
dative DNA damage in inflammatory bowel disease, Scand. J. Gastro., 36, 1289, 2001.
27. Rahman, I., Oxidative stress and gene transcription in asthma and chronic obstructive
pulmonary disease: antioxidant therapeutic targets, Curr. Drug Targets Inflamm.
Allergy, 1, 291, 2002.
9281_C005.fm Page 59 Wednesday, July 18, 2007 1:50 PM
28. Rice-Evans, C., Plant polyphenols: free radical scavengers or chain-breaking antiox-
idants? Biochem. Soc. Symp., 61, 103, 1995.
29. Dreosti, I. E., Antioxidant polyphenols in tea, cocoa, and wine, Nutrition, 16, 692,
2000.
30. Dragsted, L. O., Antioxidant actions of polyphenols in humans, Int. J. Vit. Nutr. Res.,
73, 112, 2003.
31. Das, D. K. et al., Cardioprotection of red wine: role of polyphenolic antioxidants,
Drugs Exp. Clin. Res., 25, 115, 1999.
32. Ferguson, L. R., Role of plant polyphenols in genomic stability, Mutat. Res., 475,
89, 2001.
33. Bianchini, F. and Vainio, H., Wine and resveratrol: mechanisms of cancer prevention?,
Eur. J. Cancer Prev., 12, 417, 2003.
34. Scalbert, A., Johnson, I. T., and Saltmarsh, M., Polyphenols: antioxidants and beyond,
Am. J. Clin. Nutr., 81, 215S, 2005.
35. Williams, R. J., Spencer, J. P., and Rice-Evans, C., Flavonoids: antioxidants or
signalling molecules? Free Radical Biol. Med., 36, 838, 2004.
36. Kundu, J. K. and Surh, Y. -J., Breaking the relay in deregulated cellular signal
transduction as a rationale for chemoprevention with anti-inflammatory phytochem-
icals, Mutat. Res., 591, 123, 2005.
37. Wang, S. Y. et al., Antioxidant activity in lingonberries (Vaccinium vitis-idaea L.)
and its inhibitory effect on activator protein-1, nuclear factor-kappaB, and mitogen-
activated protein kinases activation, J. Agric. Food Chem., 53, 3156, 2005.
38. Philpott, M. et al., Enhanced coloration reveals high antioxidant potential in new
sweet potato cultivars, J. Sci. Food Agric., 83, 1076, 2003.
39. Walsh, S. and Aisen, P. S., Inflammatory processes and Alzheimers disease, Expert
Rev. Neurother., 4, 793, 2004.
40. Karunasinghe, N. et al., DNA stability and serum selenium levels in a high-risk group
for prostate cancer, Cancer Epidemiol. Biomarkers Prev., 13, 391, 2004.
9281_C005.fm Page 60 Wednesday, July 18, 2007 1:50 PM
9281_C006.fm Page 61 Wednesday, July 18, 2007 1:57 PM
6 Personalized Prevention
of Type 2 Diabetes
Hans-Georg Joost
CONTENTS
61
9281_C006.fm Page 62 Wednesday, July 18, 2007 1:57 PM
is associated with morbid obesity and insulin resistance, and reflects a progressive
failure of the pancreatic -cells. The genetic basis of the diabetes can be dissected into
adipogenic and diabetogenic (i.e., causing hyperglycemia) alleles [1012]. Diabetes
requires the presence of both types of predisposing alleles (diabesity). Consequently,
in the absence of diabetogenic alleles, morbid obesity does not produce diabetes (e.g.,
in the ob/ob-mouse or the fa/fa-rat). Lean mouse lines (e.g., NON, SJL, or C57Ks/J)
may carry diabetogenic alleles; these will become active as a diabetogenic background
when the mice become obese either by a single obesity gene (e.g., in the db/db strain)
or after crossbreeding with a polygenic obesity strain such as NZO [1012].
6.2.2 NUTRITION
Most mouse strains are sensitive to a high-fat diet in that they develop obesity and
insulin resistance associated with impaired glucose tolerance [13]. Furthermore, in
diabetes-susceptible mouse strains such as NZO, the development of diabetes can be
accelerated by a high-fat diet [12]. Because diabetes in these mouse models reflects
failure and destruction of insulin-secreting cells, the finding seems to indicate that the
-cell is sensitive against fatty acids and incorporated triglycerides (lipotoxicity; see
later text). However, -cell destruction appears to also depend on the dietary carbo-
hydrates, because substitution of protein [14, 15] or fat [16] for carbohydrates delayed
or prevented the diabetes in db/db or NZO mice, respectively. Interestingly, the effect
of the high-fat diet on the development of diabetes can depend on certain genotypes.
In NZO mice, the effect of a diabetogenic allele on chromosome 5 (tentatively desig-
nated Nob1) depend on the fat content of the diet, whereas the effect of a second QTL
(Nidd/SJL) did not [12]. Such data provide proof of principle for the concept that gene
variants may determine the quality of the response to nutrients. Identification of these
variants in humans will allow the establishment of genotype-specific nutritional rec-
ommendations with regard to dietary fat and diabetes risk.
The association of diabetes with obesity, and the diabetogenic effect of a high-fat
diet, has lent support to the hypothesis that -cells incorporating excess triglycerides
undergo apoptosis [17]. In mouse models of diabetes and insulin resistance, enhanced
hepatic lipogenesis is a key pathophysiological feature [18,19]. When lipids accu-
mulate in nonadipose tissues during overnutrition, fatty acids enter deleterious
pathways such as formation of ceramide. Ceramide is probably the most damaging
lipid and is a cause of lipoapoptosis [17]. Alternatively, it has been postulated that
elevation of postprandial and postabsorptive blood glucose levels damage the pan-
creatic -cell (glucotoxicity). According to this hypothesis, the -cell is very sensitive
to oxidative damage, or is sensitized by a low antioxidative capacity [20]. It was
suggested that elevated blood glucose concentrations lead to formation of reactive
oxygen radicals [21], which cause loss of essential transcription factors such as
MafA and irreversible cell damage [22]. This hypothesis is supported by numerous
experimental findings: Long-term culture of -cells in the presence of high glucose
concentrations affects their function [23]. In db/db [14,15] or NZO mice [16],
9281_C006.fm Page 64 Wednesday, July 18, 2007 1:57 PM
TABLE 6.1
Anthropometric, Nutritional, Lifestyle, and Genetic Risk Factors
for the Development of Type 2 Diabetes
Risk Factors for the Development of Type 2 Diabetes
Anthropometry Age
Waist circumference
Height
Nutrition Whole grain bread (>50 g/d)
Red meat (>100 g/d)
Sugar-containing beverages (>300 ml/d)
Coffee (150 ml/d)
Moderate alcohol consumption (<40 g/d)
Lifestyle Exercise (sport, hiking, biking, gardening)
Smoking (>20/d)
Gene variants Transcription factor PPAR
Potassium channel KIR 6.2
Transcription factor TCF7L2
Transcription factor HHEX
Zn transporter SLC 30A8
CDKAL1
CDKN2
IGF2BP2
FTO
Note: Arrows indicate increase or decrease of risk with the indicated factors or the rare allele of the
indicated gene. A score based on these factors can be used to assess the individual diabetes risk, and
can guide personalized prevention [61]. Interaction between a genotype and a nutritional variable has
been suggested for the Pro12Ala polymorphism of PPAR and the dietary fat content [62]. Details of
nongenetic risk factors are described in Reference 34, and diabetogenic alleles are reviewed and
described in references 26, 27, 31, and 64.
9281_C006.fm Page 65 Wednesday, July 18, 2007 1:57 PM
Considerable efforts have been made to identify genes that cause type 2 diabetes.
In genomewide searches, chromosomal segments (quantitative trait loci, QTL) har-
boring alleles associated with hyperglycemia or other traits related with the disease
were identified. In one of these QTL, positional cloning of the gene responsible for
the effect, the calcium-dependent protease calpain-10, was successful [28]. In the
original cohort, the diabetogenic haplotype of calpain-10 was associated with a
threefold higher diabetes risk. Subsequent studies observed smaller effects, and a
meta-analysis of 26 studies determined a mean effect of the allele of only 20% [29].
In addition, several gene variants associated with an elevated diabetes risk were
identified in numerous case control studies by a candidate gene approach (e.g., the
PPAR Pro12Ala polymorphism and KIR6.2). These studies indicated that the con-
tribution of each variant to the overall diabetes risk is small (20 to 30%), although
statistically significant [26,27,30]. Thus, a single gene can predict only small
increases of the risk for type 2 diabetes. More recently, the transcription factor
TCF7L2 has been identified by positional cloning as a major diabetes gene in an
Icelandic study population, giving rise to a 1.5- to 2.5-fold disease risk [31]. In
addition, six other susceptibility genes (Table 6.1) have recently been identified
in genome-wide association studies [64]. However, it appears reasonable to assume
that many susceptibility genes have not been identified so far. Furthermore, it remains
to be investigated whether the effect of the known susceptibility genes depends on
nutritional or other environmental parameters, as has been demonstrated in mouse
models.
is associated with a higher diabetes risk. In addition, the diabetes risk is increased
by the consumption of sugar-sweetened soft drinks [40].
In addition to dietary carbohydrates, dietary fat seems to represent a major factor
that determines diabetes risk. The consumption of saturated triglycerides and trans-
fatty acids is associated with an increased, and the consumption of polyunsaturated
fatty acids with a reduced risk [38]. These epidemiological associations may reflect
direct effects of the different types of fat and fatty acids on the pancreatic -cell, or
indirect effects on insulin sensitivity, glucose tolerance and, consequently, on the
postprandial blood glucose excursions. Thus, the data are consistent with a scenario
in which blood glucose excursions, in particular under conditions of insulin resis-
tance and a genetically determined susceptibility of the pancreatic -cell, play a
crucial role in the pathogenesis of diabetes.
In an attempt to identify nutritional patterns rather than single nutrients and food
components associated with an increased diabetes risk, a prudent and a western
dietary pattern were compared [41]. The prudent dietary pattern characterized by a
higher consumption of vegetables, fruit, fish, poultry, and whole grains was associ-
ated with a lower diabetes risk, whereas the Western pattern (higher consumption
of red meat, processed meat, French fries, high-fat dairy products, and refined grains)
was associated with an increased risk (OR 1.6). More recently, the method of reduced
rank regression (RRR) was applied for the first time to identify dietary patterns
associated with a disease risk [42,43]. Using the risk factors serum adiponectin,
HbA1c, HDL, and CRP, a nutritional pattern was identified in incident diabetics
from the EPIC-Potsdam cohort that produced a maximal reduction of these risk
factors. This nutritional pattern consists of a high consumption of fruits, a low
consumption of meat, beer, soft drinks, and white bread, and is associated with an
approximately 80% reduction of the diabetes risk [43]. Similar results were obtained
independently in a Finnish cohort [44].
Moderate alcohol consumption, with the exception of beer [43], has consistently
been observed to lower the diabetes risk [34]. This effect might reflect the inhibitory
effect of alcohol on gluconeogenesis and hepatic glucose output, which could lead
to a reduction of fasting blood glucose levels. A similar effect was observed in a
study with the oral antidiabetic drug metformin, which exerts a blood-sugar-lowering
effect by inhibition of hepatic glucose output [8]. Furthermore, the beneficial effect
of blood sugar control on the incidence of progression of impaired glucose tolerance
to overt diabetes (fasting hyperglycemia) has been shown with the glucosidase
inhibitor acarbose, which blunts postprandial blood glucose excursions [9]. In the
case of beer, the beneficial effect of the alcohol seems to be outweighed by its high
carbohydrate content. Thus, the epidemiologic data as well as the results of different
pharmacological intervention studies are consistent with a scenario in which blood
glucose excursions play a major role in the pathogenesis of -cell failure.
disease than in healthy controls. Men who watched TV for more than 40 h per week
had an approximately threefold higher diabetes risk than those who spent less than
1 h per week watching TV [46]. Furthermore, a protective effect of exercise as part
of a weight reduction program including diet was observed in an intervention study
[7]. As the intervention resulted in a minor weight reduction (approximately 4 kg)
but a substantial (60%) reduction of diabetes risk, the authors suggested that exercise
exerted an effect that was independent of the weight reduction. This effect may be
due to an increased insulin sensitivity of muscle in response to exercise [47]. It
should be noted, however, that at present there are no data from intervention studies
directly comparing the effect of exercise with that of dietary intervention.
The majority of epidemiologic studies testing the effect of smoking have detected
a moderate increase of the diabetes risk that was reversed after cessation of smoking
[34]. It is noteworthy that the beneficial effect of smoking cessation apparently
outweighs the adverse effect on weight gain. The pathophysiological mechanism of
the effect of smoking is unclear. Nicotine stimulates sympathetic nervous activity
and may therefore stimulate transient elevations of blood glucose; such an effect
would be consistent with the concept that blood sugar excursions play a major role
in the pathophysiology of the disease.
During the last few years, several serum factors have been identified that may indicate
an increased risk of diabetes years before the actual onset of the disease. A small
but significant increase of the glycosylated hemoglobin (HbA1c) has consistently
been found to be associated with an increased risk of developing overt diabetes [48].
Type 2 diabetes is preceded by a prediabetic period in which glucose tolerance is
impaired (IGT, impaired glucose tolerance). Fasting blood glucose is still normal,
but increased postprandial blood glucose excursions are responsible for a small
increase in HbA1c levels. There is a probability of 15 to 20% that IGT is converted
to fasting hyperglycemia within the next year. In addition to HbA1c, it has been
shown that a moderate elevation of fasting blood glucose levels predicts an increased
risk of developing diabetes within a period of 6 years [49].
Serum parameters reflecting insulin resistance such as characteristic alterations
of the lipoprotein profile (low HDL cholesterol and elevated triglycerides) and
reduced levels of the cytokine adiponectin are associated with an increased diabetes
risk [50]. Adiponectin is secreted from adipose tissue, and enhances insulin sensi-
tivity of liver, muscle, and adipocyte [51]; its serum levels are reciprocal to body
mass index and intrahepatic triglycerides [52].
Obesity is associated with a massive infiltration of adipose tissue with immune
cells such as macrophages, and it has been suggested that the pathogenesis of type
2 diabetes has an immunologic component in that cytokines released from adipose
tissue play a causal role in the insulin resistance and -cell failure [53]. Consequently,
serum levels of inflammatory cytokines or other biomarkers of inflammatory pro-
cesses such as IL-6 and CRP are elevated long before the onset of overt type 2
diabetes [54,55].
9281_C006.fm Page 68 Wednesday, July 18, 2007 1:57 PM
KEY READINGS
Diamond, J., The double puzzle of diabetes, Nature, 423, 599, 2003.
Hansen, L. and Pedersen, O., Genetics of type 2 diabetes mellitus: status and perspectives,
Diabetes Obes. Metab., 7,122, 2005.
Schulze, M.B. and Hu, F.B., Primary prevention of diabetes: what can be done and how much
can be prevented? Annu. Rev. Public Health, 26, 445, 2005.
Stevenson, C., Barroso, I., and Wareham, N., The genetics of type 2 diabetes. In Nutritional
Genomics, Eds. Brigelius-Floh, R. and Joost, H.-G., Wiley-VCH, Weinheim, 223, 2006.
9281_C006.fm Page 70 Wednesday, July 18, 2007 1:57 PM
REFERENCES
1. Diamond, J., The double puzzle of diabetes, Nature, 423, 599, 2003.
2. Hanefeld, M. and Kohler, C., Screening, prevention and early therapy of type 2
diabetics, MMW Fortschr. Med., 146, 28, 31, 2004.
3. Wabitsch, M., Hauner, H., Hertrampf, M., Muche, R., Hay, B., Mayer, H., Kratzer, W.,
Debatin, K.M., and Heinze, E., Type II diabetes mellitus and impaired glucose
regulation in Caucasian children and adolescents with obesity living in Germany, Int.
J. Obes. Relat. Metab. Disord., 28, 307, 2004.
4. Wolf, A.M. and Colditz, G.A., Current estimates of the economic cost of obesity in
the United States, Obes. Res., 6, 97, 1998.
5. Hauner, H., Landgraf, R., Schulze, J., Spranger, J., and Standl, E., Nationales Aktion-
sforum Diabetes mellitus, Prvention des Typ-2-Diabetes mellitus, Positionspapier
des Nationalen Aktionsforum Diabetes Mellitus, Dtsch. Med. Wochenschr., 130, 1053,
2005.
6. Manuel, D.G. and Schultz, S.E., Health-related quality of life and health-adjusted
life expectancy of people with diabetes in Ontario, Canada, 19961997, Diabetes
Care, 27, 407, 2004.
7. Tuomilehto, J., Lindstrom, J., Eriksson, J.G., Valle, T.T., Hamalainen, H., Ilanne-
Parikka, P., Keinanen-Kiukaanniemi, S., Laakso, M., Louheranta, A., Rastas, M.,
Salminen, V., and Uusitupa, M., Finnish Diabetes Prevention Study Group. Prevention
of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired
glucose tolerance, N. Engl. J. Med., 344, 1343, 2001.
8. Knowler, W.C., Barrett-Connor, E., Fowler, S.E., Hamman, R.F., Lachin, J.M.,
Walker, E.A., and Nathan, D.M., Diabetes Prevention Program Research Group.
Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin,
N. Engl. J. Med., 346, 393, 2002.
9. Chiasson, J.L., Josse, R.G., Gomis, R., Hanefeld, M., Karasik, A., and Laakso, M.,
STOP-NIDDM Trial Research Group. Acarbose for prevention of type 2 diabetes
mellitus: the STOP-NIDDM randomised trial, Lancet, 359, 2072, 2002.
10. Leiter, E.H., Reifsnyder, P.C., Flurkey, K., Partke, H.J., Junger, E., and Herberg, L.,
NIDDM genes in mice: deleterious synergism by both parental genomes contributes
to diabetogenic thresholds, Diabetes, 47, 1287, 1998.
11. Plum, L., Kluge, R., Giesen, K., Altmuller, J., Ortlepp, J.R., and Joost, H.G., Type 2
diabetes-like hyperglycemia in a backcross model of NZO and SJL mice: charact-
erization of a susceptibility locus on chromosome 4 and its relation with obesity,
Diabetes, 49, 1590, 2000.
12. Plum, L., Giesen, K., Kluge, R., Junger, E., Linnartz, K., Schurmann, A., Becker, W.,
and Joost, H.G., Characterisation of the mouse diabetes susceptibility locus Nidd/SJL:
islet cell destruction, interaction with the obesity QTL Nob1, and effect of dietary
fat, Diabetologia, 45, 823, 2002.
13. West, D.B., Boozer, C.N., Moody, D.L., and Atkinson, R.L., Dietary obesity in nine
inbred mouse strains, Am. J. Physiol., 262, R1025, 1992.
14. Leiter, E.H., Coleman, D.L., Eisenstein, A.B., and Strack, I., Dietary control of
pathogenesis in C57BL/KsJ db/db diabetes mice, Metabolism, 30, 554, 1981.
15. Leiter, E.H., Coleman, D.L., Ingram, D.K., and Reynolds, M.A., Influence of dietary
carbohydrate on the induction of diabetes in C57BL/KsJ-db/db diabetes mice, J. Nutr.,
113, 184, 1983.
16. Jrgens, H., Schmolz, K., Neschen, S., Ortmann, S., Blher, M., Klaus, S., Tschp,
M., Joost, H.-G., and Schrmann, A., Development of diabetes in morbidly obese
9281_C006.fm Page 71 Wednesday, July 18, 2007 1:57 PM
Magnusson, K.P., Walters, G.B., Palsdottir, E., Jonsdottir, T., Gudmundsdottir, T.,
Gylfason, A., Saemundsdottir, J., Wilensky, R.L., Reilly, M.P., Rader, D.J., Bagger,
Y., Christiansen, C., Gudnason, V., Sigurdsson, G., Thorsteinsdottir, U., Gulcher, J.R.,
Kong, A., and Stefansson, K., Variant of transcription factor 7-like 2 (TCF7L2) gene
confers risk of type 2 diabetes, Nat. Genet., 38, 320, 2006.
32. Colditz, G.A., Willett, W.C., Rotnitzky, A., and Manson, J.E., Weight gain as a risk
factor for clinical diabetes mellitus in women, Ann. Intern. Med., 122, 481, 1995.
33. Wang, Y., Rimm, E.B., Stampfer, M.J., Willett, W.C., and Hu, F.B., Comparison of
abdominal adiposity and overall obesity in predicting risk of type 2 diabetes among
men, Am. J. Clin. Nutr., 81, 555, 2005.
34. Schulze, M.B., and Hu, F.B., Primary prevention of diabetes: What can be done and
how much can be prevented? Annu. Rev. Public Health, 26, 445, 2005.
35. Bajaj, M., Suraamornkul, S., Pratipanawatr, T., Hardies, L.J., Pratipanawatr, W., Glass,
L., Cersosimo, E., Miyazaki, Y., and DeFronzo, R.A., Pioglitazone reduces hepatic
fat content and augments splanchnic glucose uptake in patients with type 2 diabetes.
Diabetes, 52, 1364, 2003.
36. Stumvoll, M., Control of glycaemia: from molecules to men. Minkowski Lecture
2003. Diabetologia, 47, 770, 2004.
37. Petersen, K.F., Dufour, S., Befroy, D., Lehrke, M., Hendler, R.E., and Shulman, G.I.,
Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hypergly-
cemia by moderate weight reduction in patients with type 2 diabetes, Diabetes, 54,
603, 2005.
38. Hu, F.B., Manson, J.E., Stampfer, M.J., Colditz, G., Liu, S., Solomon, C.G., and
Willett, W.C., Diet, lifestyle, and the risk of type 2 diabetes mellitus in women, N.
Engl. J. Med., 345, 790, 2001.
39. Willett, W., Manson, J., and Liu, S., Glycemic index, glycemic load, and risk of type 2
diabetes, Am. J. Clin. Nutr., 76, 274S, 2002.
40. Schulze, M.B., Manson, J.E., Ludwig, D.S., Colditz, G.A., Stampfer, M.J, Willett,
W.C, and Hu, F.B., Sugar-sweetened beverages, weight gain, and incidence of type 2
diabetes in young and middle-aged women, JAMA, 292, 927, 2004.
41. van Dam, R.M., Rimm, E.B., Willett, W.C., Stampfer, M.J., and Hu, F.B., Dietary
patterns and risk for type 2 diabetes mellitus in U.S. men, Ann. Intern. Med., 5, 136,
201, 2002.
42. Hoffmann, K., Schulze, M.B., Schienkiewitz, A., Nothlings, U., and Boeing, H.. Appli-
cation of a new statistical method to derive dietary patterns in nutritional epidemiology,
Am. J. Epidemiol., 159, 935, 2004.
43. Heidemann, C., Hoffmann, K., Spranger, J., Klipstein-Grobusch, K., Mohlig, M.,
Pfeiffer, A.F., and Boeing, H., A dietary pattern protective against type 2 diabetes in
the European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam
Study cohort, Diabetologia, 48, 1126, 2005.
44. Montonen, J., Knekt, P., Harkanen, T., Jarvinen, R., Heliovaara, M., Aromaa, A., and
Reunanen, A., Dietary patterns and the incidence of type 2 diabetes, Am. J. Epidemiol.,
161, 219, 2005.
45. Sigal, R.J., Kenny, G.P., Wasserman, D.H., and Castaneda-Sceppa, C., Physical activity/
exercise and type 2 diabetes, Diabetes Care, 27, 18, 2004.
46. Hu, F.B., Leitzmann, M.F., Stampfer, M.J., Colditz, G.A., Willett, W.C., and Rimm,
E.B., Physical activity and television watching in relation to risk for type 2 diabetes
mellitus in men. Arch. Intern. Med., 161, 1542, 2001.
9281_C006.fm Page 73 Wednesday, July 18, 2007 1:57 PM
47. Hawley, J.A., Exercise as a therapeutic intervention for the prevention and treatment
of insulin resistance, Diabetes Metab. Res. Rev., 20, 383, 2004.
48. Khaw, K.T., Wareham, N., Luben, R., Bingham, S., Oakes, S., Welch, A., and Day,
N., Glycated haemoglobin, diabetes, and mortality in men in Norfolk cohort of
European prospective investigation of cancer and nutrition (EPIC-Norfolk), Br. Med.
J., 322, 15, 2001.
49. Lyssenko, V., Almgren, P., Anevski, D., Perfekt, R., Lahti, K., Nissen, M., Isomaa,
B., Forsen, B., Homstrom, N., Saloranta, C., Taskinen, M.R., Groop, L., and Tuomi,
T., Botnia study group, Predictors of and longitudinal changes in insulin sensitivity
and secretion preceding onset of type 2 diabetes, Diabetes, 54, 166, 2005.
50. Spranger, J., Kroke, A., Mhlig, M., Bergmann, M.M., Ristow. M., Boeing, H., and
Pfeiffer, A.F., Adiponectin and protection against type 2 diabetes mellitus, Lancet,
361, 226, 2003, Erratum in: Lancet, 361, 1060, 2003.
51. Trujillo, M.E. and Scherer, P.E., Adiponectin journey from an adipocyte secretory
protein to biomarker of the metabolic syndrome, J. Intern. Med., 257, 167, 2005.
52. Bajaj, M., Suraamornkul, S., Piper, P., Hardies, L.J., Glass, L., Cersosimo, E.,
Pratipanawatr, T., Miyazaki, Y., and DeFronzo, R.A., Decreased plasma adiponectin
concentrations are closely related to hepatic fat content and hepatic insulin resis-
tance in pioglitazone-treated type 2 diabetic patients. J. Clin. Endocrinol. Metab.,
89, 200, 2004.
53. Kolb, H. and Mandrup-Poulsen, T., An immune origin of type 2 diabetes? Diabeto-
logia, 48, 1038, 2005.
54. Spranger, J., Kroke, A., Mohlig, M., Hoffmann, K., Bergmann, M.M., Ristow,
M., Boeing, H., and Pfeiffer, A.F., Inflammatory cytokines and the risk to develop
type 2 diabetes: results of the prospective population-based European Prospective
Investigation into Cancer and Nutrition (EPIC)-Potsdam Study, Diabetes, 52, 812,
2003.
55. Sjoholm, A. and Nystrom, T., Inflammation and the etiology of type 2 diabetes,
Diabetes Metab. Res. Rev., 22, 4, 2006.
56. Torgerson, J.S., Hauptman, J., Boldrin, M.N., and Sjostrom, L., XENical in the
prevention of diabetes in obese subjects (XENDOS) study: a randomized study of
orlistat as an adjunct to lifestyle changes for the prevention of type 2 diabetes in
obese patients, Diabetes Care, 27, 155, 2004.
57. Sugerman, H.J., Wolfe, L.G., Sica, D.A., and Clore, J.N., Diabetes and hypertension
in severe obesity and effects of gastric bypass-induced weight loss, Ann. Surg., 237,
751, 2003.
58. Costa, A., Conget, I., and Gomis, R., Impaired glucose tolerance: is there a case for
pharmacologic intervention?, Treat. Endocrinol., 205, 2002.
59. Lindstrom, J. and Tuomilehto, J., The diabetes risk score: a practical tool to predict
type 2 diabetes risk, Diabetes Care, 26, 725, 2003.
60. Rathmann, W., Martin, S., Haastert, B., Icks, A., Holle, R., Lowel, H., and Giani,
G., KORA Study Group. Performance of screening questionnaires and risk scores
for undiagnosed diabetes: the KORA Survey 2000, Arch. Intern. Med., 165, 436,
2005.
61. Schulze, M.B., Hoffmann, K., Boeing, H., Linseisen, J., Rohrmann, S., Mhlig, M.,
Pfeiffer, A.F.H., Spranger, J., Thamer, C., Hring, H.-U., Fritsche, A., and Joost,
H.-G., An accurate risk score based on anthropometric, dietary and lifestyle factors
to predict the development of type 2 diabetes, Diabetes Care, 30, 510, 2007.
9281_C006.fm Page 74 Wednesday, July 18, 2007 1:57 PM
62. Lindi, V.I., Uusitupa, M.I., Lindstrom, J., Louheranta, A., Eriksson, J.G., Valle, T.T.,
Hamalainen, H., Ilanne-Parikka, P., Keinanen-Kiukaanniemi, S., Laakso, M., and
Tuomilehto, J.; Finnish Diabetes Prevention Study. Association of the Pro12Ala
polymorphism in the PPAR-gamma2 gene with 3-year incidence of type 2 diabetes
and body weight change in the Finnish Diabetes Prevention Study, Diabetes, 51,
2581, 2002.
63. U.K. Prospective Diabetes Study (UKPDS) Group: effect of intensive blood-glucose
control with metformin on complications in overweight patients with type 2 diabetes
(UKPDS 34), Lancet, 352, 854, 1998.
64. Zeggini, E., Weedon, M.N., Lindgren, C.M., Frayling, T.M., Elliott, K.S., Lango, H.,
Timpson, N.J., Perry, J.R., Rayner, N.W., Freathy, R.M., Barrett, J.C., Shields, B.,
Morris, A.P., Ellard, S., Groves, C.J., Harries, L.W., Marchini, J.L., Owen, K.R.,
Knight, B., Cardon, L.R., Walker, M., Hitman, G.A., Morris, A.D., Doney, A.S.,
McCarthy, M.L., and Hattersley, A.T., Replication of genome-wide association signals
in U.K. samples reveals risk loci for type 2 diabetes, Science, in press, 2007.
9281_C007.fm Page 75 Wednesday, July 18, 2007 1:58 PM
7 Toward Personalized
Nutrition for the
Prevention and Treatment
of Cancer
John C. Mathers
CONTENTS
7.1 INTRODUCTION
Cancers develop because of unrepaired genomic damage, causing aberrant gene expres-
sion that gives the tumor cell and its progeny a competitive advantage. For the majority
of cancers, this genomic damage results from environmental influences, which supports
the idea that if these influences were better understood, a large proportion of the significant,
and growing, global burden of cancer is potentially preventable. There is strong support
from epidemiologic studies that dietary (and lifestyle) factors play a substantial role in
modifying risk for a number of the major cancers including breast, colon, and prostate.
The exciting new information emerging from recent research on what determines personal
cancer risk includes evidence for the influence of (1) individual genotype, (2) dietary
factors on the acquisition and repair of genomic damage, and (3) interactions between
75
9281_C007.fm Page 76 Wednesday, July 18, 2007 1:58 PM
polymorphisms in specific genes and food components. It also seems likely that diet will
affect both quality of life and survival after cancer diagnosis. At present, the evidence base
is too limited to justify personalized dietary advice based on knowledge of genotype and
of dietgene interactions in an attempt to reduce cancer risk. However, the limited evidence
available to date is sufficiently promising to encourage research funders and researchers
to adopt this area as a high priority for sustained research. It will also be important to
undertake translational research to discover how best to deliver personalized dietary advice,
products, or other services to elicit the desired behavioral changes.
In the year 2000, 6.2 million people died from cancer, and 4.7 million women
and 5.3 million men were diagnosed with a malignant tumor [1]. Although mortality
rates from cancer are twice as high in developed as in developing countries, the
latter are catching up as the smoking epidemic spreads [1]. By 2020, it is anticipated
that the global burden of cancer will have increased by 50% because of the prevalence
of smoking and other unhealthy lifestyle choices, which are exemplified by the rising
incidence of obesity [1,2]. Better screening and earlier diagnosis combined with
more effective treatments and palliative care will all be important in management
of the growing global cancer burden, but a strong case can be made for a greater
focus on cancer prevention [1,3]. The evidence base for cancer prevention policies
will include understanding of the biology of the disease and of the factors that
contribute to risk for both populations and individuals.
Environmental
Factors DNA
Repair
Aberrant
DNA Gene
Damage Expression
Endogenous Apoptosis
Factors
FIGURE 7.1 Tumors arise in (stem) cells that have acquired genomic damage resulting in
abnormal gene expression. Genomic damage can be repaired or damaged cells can be removed
by apoptosis but, when these systems fail, cells that have a competitive advantage (in a
Darwinian sense) may initiate tumorigenesis.
9281_C007.fm Page 77 Wednesday, July 18, 2007 1:58 PM
cell, and its progeny, with a selective advantage. In a Darwinian fashion, further
advantageous genetic and epigenetic events enable the emerging neoplastic clone to
grow through enhanced proliferation, evasion of apoptosis, disregard of differentiation
signals, angiogenesis, and invasion [5]. Mutations of nuclear DNA, which result in
expression of a disabled protein or no protein, are the best-known types of genomic
damage, but mitochondrial DNA mutations, chromosomal abnormalities (loss, rear-
rangement, or duplication), loss of telomere function (which can lead to chromosomal
instability), and aberrant epigenetic marking are also important features of tumors.
Epigenetics describes changes to the genome that result in altered gene expression,
are heritable from one cell generation to the next, but do not involve changes to the
primary sequence. The main epigenetic markings of the genome include (1) posttrans-
lational modifications of the octet of histone proteins around which nuclear DNA is
wrapped, causing alterations in chromatin structure and (2) the pattern of methylation
of cytosine residues in CpG dinucleotides in DNA. Together, these epigenetic marks
are believed to constitute an epigenetic code that regulates gene expression in particular
cells under specific circumstances [6]. Over 20 years ago, Feinberg and Vogelstein
provided early evidence that epigenetic events might be important in carcinogenesis
when they reported that tumors had a lower level of DNA methylation than normal
tissues [7]. It is now clear that epigenetic events play a critical role in most, if not all,
cancers, and that they may be a mechanism through which environmental factors
(including diet) influence stem cell biology and, thus, cancer risk [8,9]. New onco-
genes and tumor suppressor genes whose expression is regulated by epigenetic events
are being discovered, and it has been proposed that epigenetic changes might addict
cancer cells to altered signal-transduction pathways early in tumorigenesis [10,11].
This evidence makes epigenetic markings an increasingly attractive target for identi-
fication of those at enhanced cancer risk and for chemoprevention [9,12,13].
TABLE 7.1
Estimates of the Variance in Cancer Risk Accounted for by Heritable
and Environmental Factors from Studies of Swedish, Danish,
and Finnish Twins
Shared Environmental Nonshared Environmental
Cancer Site Heritable Factors Factors Factors
They found significant associations in 16 of the 50 studies, but only 3 of these associations
were reported in more than 1 study. After pooling data from a number of studies, they
concluded that significant associations were seen for polymorphisms in three genes
(Table 7.2) [20]. For the I307K polymorphism in APC (which is relatively common in
the Ashkenazi population) and for the variable number tandem repeat polymorphism in
HRAS1, those carrying the unusual versions of the gene had increased incidence of bowel
cancer (Table 7.2). Conversely, those with TT at position 677 in the MTHFR gene had,
on average, about 25% lower risk of bowel cancer. A more recent review by Sharp and
Little of 10 studies involving more than 4000 bowel cancer cases showed that most
studies suggested a lower, but not significantly so, risk for TT individuals [21]. Indeed,
the relative risk of bowel cancer was clearly lower (0.45, 95% confidence interval 0.24
to 0.86) in TT compared with CC individuals only in the study of male physicians
participating in the Physicians Health Study [21]. The authors concluded that the effect
of the MTHFR C677T polymorphism per se is modest and emphasized the lack of
understanding of genegene and of geneenvironment interactions. [21] The MTHFR
gene encodes the enzyme methylenetetrahydrofolate reductase, which catalyzes the rem-
ethylation of homocysteine to methionine. The TT version of the gene results in a protein
with a valine rather than an alanine at amino acid position 222 and which has only about
one third of the normal catalytic activity, because the mutant protein does not bind the
cofactor FAD as strongly as does the wild-type protein [22]. As a consequence, individ-
uals carrying the TT version of MTHFR require higher intakes of folate to maintain
normal circulating concentrations of homocysteine.
Major mapping exercises such as those carried out by the International SNP
Map Working Group and by the International HapMap Consortium have described
the scale and nature of human genetic variability and provide tools for more extensive
genetic association studies [23,24]. As an example, the Cancer Genetic Markers of
Susceptibility Project (http://cgems.cancer.gov/index.asp), which began in 2005 and
is expected to run for 3 years at a cost of $14 million, will use ultra-high-throughput
genotyping to identify genetic variants that increase susceptibility to prostate and
breast cancer. It is possible that such genomewide scans will identify either large
numbers of gene variants each of which makes quite a modest contribution to risk
or a much smaller number of larger-effect variants that, to date, have escaped
detection. Some have urged caution about the potential benefits of such studies,
arguing that (1) they overlook the importance of environmental factors, e.g., diet
and lifestyle and that (2) even if the anticipated genetic variants are discovered, it
is not clear how that knowledge can be translated into clinical benefit [25].
TABLE 7.2
Polymorphisms Associated with Altered Risk of Bowel Cancer
Gene Polymorphism Odds Ratio 95% Confidence Interval
Source: From Houlston, R.S. and Tomlinson, I.P.M., Gastroenterology, 121, 282, 2001. With permission.
9281_C007.fm Page 80 Wednesday, July 18, 2007 1:58 PM
TABLE 7.3
Apparent Interactions between MTHFR C677T Genotype and Folate Intake
on Odds Ratio for Breast Cancer in Chinese Women
Genotype Q4a Q3 Q2 Q1 P for trend
Susceptibility Protective
SNP1 SNP1
SNP2 SNP2
Poor Good
Nutrition Nutrition
in utero in utero
Health
Food Food Pendulum
Component 1 Component 2
Physical
Smoking Activity
Time
Health Disease
FIGURE 7.2 The health pendulum. A simple model illustrating how key etiological factors
influence cancer risk. For a given individual, the inherited collection of susceptibility and
protective genes determines the position over the healthdisease continuum from which the
pendulum is suspended. Nutrition in utero and postnatal lifestyle factors interact with genetic
makeup to further modify risk. For most common cancers, risk increases steeply with age.
(Adapted from Mathers, J.C., Br J Nutr, 88 [Suppl. 3], S273, 2002.)
9281_C007.fm Page 82 Wednesday, July 18, 2007 1:58 PM
consortium of susceptibility and resistance genes that are inherited from the
individuals parents. Throughout life, the pendulum is able to move being pushed
to the right (further in the direction of cancer) or the left (better health) by a range
of pre- and postnatal factors; i.e., a range of dietary and other factors interact with
genotype to determine risk. A higher-risk lifestyle could include lack of physical
activity, exposure to environmental mutagens, or a poor diet. For most common
cancers, age is a potent carcinogen; i.e., risk rises steeply in old age.
the potential of genomic profiling to promote a healthy lifestyle, Haga et al. saw little
reason for optimism concerning the potential for genetic test results to motivate behavior
change [51]. The best evidence for the utility of such personalized advice will come
from properly designed, powered, and conducted randomized controlled intervention
trials. However, to my knowledge, no such trials have yet been attempted; such trials
will present formidable design, ethical, and implementation issues.
ACKNOWLEDGMENTS
Research in my laboratory on cancer prevention is funded by the Medical Research
Council (G0100496), the Biotechnology and Biological Sciences Research Council
(D20173), and by the Food Standards Agency (N12015, N12016).
KEY READINGS
Feinberg, A.P., Ohlsson, R., and Henikoff, S, The epigenetic progenitor origin of human
cancer, Nat Rev Genet, 7, 21, 2006.
Joost, H.G., Gibney, M.J., Cashman, K.D., Gorman, U., Hesketh, J.E., Mueller, M., van
Ommen, B., Williams, C.M., and Mathers, J.C., Personalized nutrition, Br J Nutr
[in press], 2007.
9281_C007.fm Page 85 Wednesday, July 18, 2007 1:58 PM
Mathers, J.C., Nutrition and cancer prevention: diet-gene interactions, Proc Nutr Soc, 62,
605, 2003.
Rock, C.L. et al., Plasma carotenoids and recurrence-free survival in women with a history
of breast cancer, J Clin Oncol, 23, 6631, 2005.
Stewart, B.W. and Kleihues, P., World Cancer Report, IARC Press, Lyon, 2003.
REFERENCES
1. Stewart, B.W. and Kleihues, P., World Cancer Report, IARC Press, Lyon, 2003.
2. Calle, E.E. and Thun, M.J., Obesity and cancer, Oncogene, 23, 6365.
3. Mathers, J.C., Nutrition and cancer prevention: diet-gene interactions, Proc Nutr Soc,
62, 605, 2003.
4. Hanahan, D. and Weinberg, R.A., The hallmarks of cancer, Cell, 100, 57, 2000.
5. Ponder, B.A.J., Cancer genetics, Nature, 411, 336, 2001.
6. Jenuwein, T. and Allis, C.D., Translating the histone code, Science 293, 1074, 2001.
7. Feinberg, A.P. and Vogelstein, B., Hypomethylation distinguishes genes of some
Human cancers from their normal counterparts, Nature, 301, 89, 1983.
8. Jones, P.A. and Baylin, S.B., The fundamental role of epigenetic events in cancer,
Nat Rev Genet, 3, 418, 2002.
9. Feinberg, A.P., Ohlsson, R., and Henikoff, S, The epigenetic progenitor origin of
human cancer, Nat Rev Genet, 7, 21, 2006.
10. Esteller, M., Epigenetics provides a new generation of oncogenes and tumour-
suppressor genes, Br J Cancer, 94, 179, 2006.
11. Baylin, S.B. and Ohm, J.E., Epigenetic gene silencing in cancer a mechanism for
early oncogenic pathway addiction?, Nat Rev Cancer, 6, 107, 2006.
12. Belshaw, N.J. et al., Use of DNA from stools to detect aberrant CpG island methylation of
genes implicated in colorectal cancer. Cancer Epidemiol Biomark Control, 13, 1495, 2004.
13. Mathers, J.C., Reversal of DNA hypomethylation by folic acid supplements: possible
role in colorectal cancer prevention, Gut, 54, 579, 2005.
14. Lichtenstein, P. et al., Environmental and heritable factors in the causation of cancer,
N Engl J Med, 343, 78, 2000.
15. Doll, R. and Peto, R., The causes of cancer: quantitative estimates of avoidable risks
in the USA today, J Natl Cancer Inst, 66, 1191, 1981.
16. World Cancer Research Fund/American Institute of Cancer Research, Food, Nutrition
and the Prevention of Cancer: A Global Perspective, World Cancer Research Fund/
American Institute of Cancer Research, Washington, D.C., 1997.
17. Department of Health, Nutritional Aspects of the Development of Cancer, Report on
Health and Social Subjects No. 48, The Stationery Office, London, 1998.
18. Garber, J.E. and Offit, K., Hereditary cancer predisposition syndromes, J Clin Oncol,
23, 276, 2005.
19. Kinzler, K.W. and Vogelstein, B., Lessons from hereditary colorectal cancer, Cell,
87, 159, 1996.
20. Houlston, R.S. and Tomlinson, I.P.M., Polymorphisms and colorectal cancer risk,
Gastroenterology, 121, 282, 2001.
21. Sharp, L. and Little, J., Polymorphisms in genes involved in folate metabolism and
colorectal neoplasia: a HuGE review, Am J Epidemiol, 159, 423, 2004.
22. Guenther, B.D. et al., The structure and properties of methylenetetrahydrofolate
reductase from Escherichia coli suggest how folate ameliorates human hyperho-
mocysteinemia, Nat Struct Biol, 6, 359, 1999.
9281_C007.fm Page 86 Wednesday, July 18, 2007 1:58 PM
45. Romond, E.H. et al., Trastuzumab plus adjuvant chemotherapy for operable HER2-
positive breast cancer, N Engl J Med, 353, 1673, 2005.
46. Ordovas, J.M., Genetic interactions with diet influence the risk of cardiovascular
disease, Am J Clin Nutr, 83, 443S, 2006.
47. Mathers, J.C., Food and cancer prevention: human intervention studies. In Dietary
Anticarcinogens and Antimutagens, Johnson, I.T. and G.R. Fenwick, G.R., Eds., Royal
Society of Chemistry, Cambridge, 2000, chap. 8.1.
48. Sanderson, P. et al., Emerging diet-related surrogate end points for colorectal cancer:
U.K. Food Standards Agency diet and colonic health workshop report, Brit J Nutr,
91, 315, 2004.
49. Marteau, T.M. and Lerman, C., Genetic risk and behavioural change, Br Med J, 322,
1056, 2001.
50. Adamson, A.J. and Mathers, J.C., Effecting dietary change, Proc Nutr Soc, 63, 537,
2004.
51. Haga, S.B., Khoury, M.J., and Burke, W., Genomic profiling to promote a healthy
lifestyle: not ready for prime time, Nat Genet, 34, 347, 2003.
52 Mathers, J.C., Pulses and carcinogenesis: potential for the prevention of colon, breast
and other cancers, Br J Nutr, 88(Suppl. 3), S273, 2002.
53. Joost, H.G., Gibney, M.J., Cashman, K.D., Gorman, U., Hesketh, J.E., Mueller, M.,
van Ommen, B., Williams, C.M., and Mathers, J.C., Personalized nutrition, Br J Nutr
[in press], 2007.
9281_C007.fm Page 88 Wednesday, July 18, 2007 1:58 PM
9281_C008.fm Page 89 Wednesday, July 18, 2007 1:59 PM
8 Nutrigenomics
and Angiogenesis
in Obesity
Aldona Dembinska-Kiec
CONTENTS
8.1 INTRODUCTION
Development and maturation/plasticity of adipose tissue vascularity is critical for
the function of this metabolic and the endocrine organ. The proangiogenic factors,
as well as the endogenous inhibitors of angiogenesis, are synthesized locally in
adipose tissue. It has been proved recently that treatment of animals with antiangio-
genic factors (such as anti-VEGF or its receptor antibody) dose dependently, in a
reversible manner, decreases adipose tissue depot and body weight. The involvement
of basal nutrient (such as fatty acids, amino acids, carbohydrates, and polyphenols)
availability and energy supply signals (i.e., caloric restriction) on the nutrient
sensors such as the mammalian target of rapamycin (mTOR) or CHOP-10/gadd
153 and the other pathways regulating adipose tissue morphogenesis are reviewed
on the basis of recent knowledge. Because transcriptomics applied to obesity, as
well as caloric restriction studies in humans, point to the important changes in pro-
and anti-inflammatory status, the essential role of personalized nutrition in regulation
of the angiogenesis-driven remodeling of the stromal vascular fraction of adipose
tissue is discussed.
89
9281_C008.fm Page 90 Wednesday, July 18, 2007 1:59 PM
Without appropriate blood supply from the capillary network, tissue cannot
survive; the circulatory system is essential for oxygen and nutrient distribution
between tissues and for the removal of by-products of metabolism. Vasculogenesis
and angiogenesis play an essential role in a number of physiologic and pathologic
events, such as fetal development, vascular and tissue remodeling in ischemia,
inflammation, proliferative diabetic retinopathy, as well as in promotion of solid
tumor growth and invasiveness (1,2).
The development and maturation/plasticity of adipose tissue vascularity is crit-
ical for the function of adipose tissue as the metabolic and the endocrine organ. It
has also been demonstrated that fetal adipocyte development is spatially and tem-
porally related to arteriolar development (3). Recent studies have evidenced that
human endothelial progenitors and preadipocytes express similar patterns of genes
and proteins [cellular markers and the proangiogenic factors such as CD34, AC133,
vascular endothelial growth factor (VEGF) receptors, integrins, serine proteases, and
others], which suggests a possible common origin from the stromal vascular progenitor
cell fraction (SVF) (3,4,5).
In adolescence, the adipose tissue SVF releases only 6% of the main proangio-
genic VEGF amount released by the differentiated adipocytes. Under the stimulus
of insulin, the differentiated adipocytes rather than the stromal vascular fraction are
a source of VEGF released by visceral adipose tissue (6), pointing to the stimulation
of angiogenesis by the growing mass of tissue.
Inammation Dierentiation
Cytokines: IL-6
TNF- FFA/RA PPAR-//RAR/RXR
Macrophages IL-1 Insulin
MCP-1 cAMP
Others Others
ROS
Adipocyte
Ca++
VSMC SVF
MMPs
VEGF Adipokines:
Angiopoietin Adiponectin, TPA, PAI, TSP
Tie1/2 Leptin, A-II
Others
NO
Angiogenesis
EC VEGF
PDGF EC and VSMC
Angiopoietin
Ephrins
Others
(MAPKs) and p70S6 kinase (S6K), migration of ECs by the stress-activated protein
kinase 2/p38 pathway, and survival (antiapoptotic effect) by Akt activation (7).
VEGF-A induces genes of the main metalloproteinases: MMP-2 and MMP-9, as
well as integrins (VE-cadherin or 3). The IL-8-induced activation of CXCR-2
receptor is necessary for matrix remodeling, adhesion, and EC differentiation during
tubulogenesis. The VEGF gene expression is induced by several factors such as
hypoxia (by the involvement of the hypoxia-inducible-factor, HIF-1), insulin, insulin
growth factors (IGFs), cytokines, as well as by nitric oxide (NO) (1,2,7).
Angiopoietin (Ang)-1 and Ang-2 with their receptors, Tie-1 and Tie-2, are
considered to be the most important partners of VEGF in the maturation of capillary
networks, especially in branching of the sprouts and modifying apoptosis to be not
redundant for capillary-sprouting cells. VEGF signaling is initiated through the
assembly of a multicomponent complex composed of the vascular endothelial (VE)-
cadherin, -catenin, VEGF-R2, and PI3-kinase. This complex then stimulates Akt,
leading to the downstream activation and nuclear translocation of the transcription
factor NF-B, antiapoptotic Bcl-2 protein synthesis, induction of IL-8, and monocyte
chemoattractant protein-1 (MCP-1) gene expression. This is followed by infiltration
of macrophages and tubulogenesis of the growing capillary network (1,7,8). Ephrin
B (EphB2) and its receptor EphB4 expressed on endothelial cell membranes deter-
mine arteriovenous shifting of capillary differentiation (8). Transforming growth
factor (TGF) with tumor necrosis factor (TNF), or platelet-derived growth factor
(PDGF) are induced in ischemic tissue or are released from activated macrophages
or platelets. They establish the lengthening of capillaries by regulating endothelial
cell quiescence, cell-to-cell interactions, and differentiation of fibroblast-like
9281_C008.fm Page 92 Wednesday, July 18, 2007 1:59 PM
progenitors to pericytes or vascular smooth muscle cells (9). Nitric oxide (NO)
synthesized by the endothelial nitric oxide synthase (eNOS) is induced by several
proangiogenic factors (including VEGF, TNF, angiotensin II (AII), insulin, leptin,
adiponectin, and others). Nitric oxide plays an essential role in angiogenesis. It
protects endothelial cells against apoptosis by stimulation of Akt, and induction of
Bcl-2 and other antiapoptotic proteins (IAPs). By inhibition of platelet aggregation
and adhesion of inflammatory cells (macrophages, leukocytes, and lymphocytes),
NO inhibits penetration of cells generating pro- and antiangiogenic factors (VEGF,
PDGF, TNF, AII, metalloproteinases, IL-8, IL-1, INF-, etc.). By inhibition of
vascular wall smooth muscle cell (VSMC) proliferation and basement membrane
protein synthesis, NO is responsible for the maturation of the capillary network and
inhibition of pathological remodeling of the vessel wall (1,79).
Simultaneously with the proangiogenic factors, the endogenous inhibitors of
angiogenesis are synthesized locally. Examples are thrombospondins (TSP-1 and
TSP-2) with their receptor CD36, which is also the long-chain fatty acid scavenger-
type receptor, or the other inhibitory products deriving from the protein digestion
such as the angiostatin, endostatin, maspin, and others (1,2,9).
may serve as the early preadipocytes, and the adipocyte tissue stromal vascular
fraction (SVF) secrete several factors necessary for angiogenesis and modulation of
the capillary network (4,5,12). Thus, during the postnatal period, VEGF expression
and resulting angiogenesis may precipitate adipogenesis (3). VEGF (as well as leptin)
are induced by ischemia (mediated by HIF-1) or insulin, and the exposure to cold
induces VEGF expression in the brown adipose tissue (1,6,8).
Using several experimental models of genetically modified mice (knockout or
transgenic), it has been demonstrated that the manipulation of the tPA, uPA/PAI-1,
or plasminogen gene expression deeply influences adipose tissue mass and weight
gain of mice fed a high-fat diet. Remodeling of extracellular matrix proteins by
endothelial, adipocyte, or macrophage-derived MMP-2 and MMP-9 is necessary for
angiogenesis and adipogenesis (1,3), and the TIMP-3 deficient mice exhibit
increased adipogenicity during mammary gland involution.
It has been proved recently that treatment of animals with antiangiogenic factors,
(such as anti-VEGF or its receptor antibody) dose dependently, in a reversible
manner, decreases adipose tissue deposition and body weight (13).
higher compared to those consuming a traditional Western diet. Thus, the beneficial
effect may be related also to the ethnic diet.
Interestingly, ferrets receiving the high dose of beta-carotene (the source of
retinoic acid) gained more weight than control animals, which is consistent with the
proadipogenic PPAR/RXR/RAR-induced pathway. No such effect was seen with the
low dose of beta-carotene (27). The size of the subcutaneousinguinal fat depot in
animals treated with the high dose of beta-carotene was significantly higher than
that of animals treated with the low dose and slightly higher than that of controls.
This study also showed that chronic treatment with beta-carotene induced a dose-dependent
hypertrophy of white adipocytes and increased neoangiogenesis in subcutaneous
WAT in all treated ferrets. It is in accordance with the suggested proangiogenic
activity of beta-carotene observed by the gene expression in endothelial cells measured
by microarray (26,27).
Beta-carotene treatment reduced the UCP1 protein levels in the interscapular
BAT as well as in the inguinal and retroperitonal WAT depots, pointing to the
accumulation of WAT-type tissue. Interestingly, rodents with knockout ,-carotene-
15,15-mono-oxygenase (Bcmo1-/-) gene accumulate beta-carotene in adipose tissue
and have increased body fat mass, which is consistent with the previously described
possible proangiogenic and proadipogenic effect of beta-carotene (28).
KEY READINGS
Carmeliet, P. Angiogenesis in health and disease. Nat Med 9(2003), 65360.
Hausman, G.J. and Richardson, R.L. Adipose tissue angiogenesis. J Anim Sci 2004; 82,
925934.
Rupnick, M.A., Panigrahy, D., Zhang, C.Y., Dallabrida, S.M., Lowell, B.B., Langer, L., and
Folkman, M.J. Adipose tissue mass can be regulated through the vasculature. Proc
Natl Acad Sci USA 2002; 99, 1073010735.
9281_C008.fm Page 98 Wednesday, July 18, 2007 1:59 PM
Voros, G., Maguoi, E., Demeulemeester, D., Clerx, N., Collen, D., and Lijen, H.R. Modulation
of angiogenesis during adipose tissue development in murine models of obesity.
Endocrinology 2005; 146, 45454554.
Wang, S., Yehya, N., Schardt, E.E., Wang, H., Drake, T.A., and Lusis, A.J. Genetic and
genomic analysis of a fat mass trait with complex inheritance reveals marked sex
specificity. PLoS Genetics /www.plosgenetix.org; 2006; 2, 01480153.
REFERENCES
1. Carmeliet, P. Angiogenesis in health and disease. Nat Med 9(2003), 65360.
2. Folkman, J., Merler, E., Abernathy, C., and Williams, G. Isolation of a tumor factor
responsible of angiogenesis. J Exp Med 1971; 133, 275288.
3. Hausman, G.J. and Richardson, R.L. Adipose tissue angiogenesis. J Anim Sci 2004;
82, 925934.
4. Rodriguez, A.M., Elabd, C., Amri, Ez-Z., Alhaud, G., and Dani, C. The human adipose
tissue is a source of multipotent stem cells. Biochemistry 2005; 87, 125128.
5. Prunet-Marcassus, B., Cousin, B., Caton, D., Andre, M., Penicaud, L., and Casteilla,
L. From heterogeneity to plasticity in adipose tissues: site-specific differences. Exp
Cell Res 2006; 312, 727736.
6. Mick, G.J., Wang, X., and McCornick, K. White adipocyte vascular endothelial
growth factor; regulation by insulin. Endocrinology 2002; 143, 948953.
7. Dembinska-Kiec, A., Dulak, J., Partyka, Huk, I., and Malinski, T. VEGF-nitric oxide
reciprocal regulation. Nat Med 1997; 3, 1177.
8. Josko, J. and Mazurek, M. Transcription factors having impact on vascular endothelial
growth factor (VEGF) gene expression in angiogenesis. Med Sci Monit 2004; 10, RA8998.
9. Crandall, D.L., Hausman, G.J., and Kral, J.G. A review on the microcirculation of
adipose tissue: anatomic, metabolic and angiogenic perspectives. Microcirculation
1997; 4, 211232.
10. Park, H.Y., Kwon, H.M., Lim, H.J., Hong, B.K., Lee, J.Y., Park, B.E., Jang, Y., Cho,
S.Y., and Kim, H.S. Potential role of leptin in angiogenesis; Leptin induces endothelial
cell proliferation and expression of metaloproteinases in vivo and in vitro. Exp Mol
Med 2001; 33, 95102.
11. Planat-Benard, V., Silvestre, J.S., Cousin, B., Andre, M., Nibbelink, M., Tamarat, R.,
Clerque, M., Manneville, C., Saillan-Barreau, C., Duriez, M., Tedgui, A., Levy, B.,
Penicaud, L., and Casteilla, L. Plasticity of human adipose lineage cells toward endo-
thelial cell: physiological and therapeutic perspectives. Circulation 2004; 109, 656663.
12. Rechman, J., Tractuev, D., Li, J., Merfeld-Clauss, S., Temm-Grove, C.J., Bovenkerk,
J.E., Pell, C.L., Johnstone, B.H., Considine, R.V., and March, K.L., Secretion of
angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation
2004; 109, 12921298.
13. Rupnick, M.A., Panigrahy, D., Zhang, C.Y., Dallabrida, S.M., Lowell, B.B., Langer,
L., and Folkman, M.J. Adipose tissue mass can be regulated through the vasculature.
Proc Natl Acad Sci USA 2002; 99, 1073010735.
14. Moloney, F., Noone, E., Loscher, C., Gibney, M.J., and Roche, H.M. Cis-9, trans 11
conjugated linoleic acid improves metabolic and molecular markers of insulin sen-
sitivity in adipose tissue and liver. Proc Nutr Soc 2004; 64,58A.
15. Voros, G., Maguoi, E., Demeulemeester, D., Clerx, N., Collen, D., and Lijen, H.R.
Modulation of angiogenesis during adipose tissue development in murine models of
obesity. Endocrinology 2005; 146, 45454554.
9281_C008.fm Page 99 Wednesday, July 18, 2007 1:59 PM
16. Rosen, ED. and Spiegelman, B.M. Molecular regulation of adipogenesis. Annu Rev
Cell Dev Biol 2000; 16, 145171.
17. Fukumura, D., Ushiyama, A., Duda, D.D., Xu, L., Tam, J., Chatterjee, V.K.K.,
Garkavtsev, I., and Jain, R.K. Paracrine regulation of angiogenesis and adipocyte
differentiation during in vivo adipogenesis. Circ Res 2003; 93, e88e97.
18. Kim, J.E. and Chen, J. Regulation of peroxisome proliferator-activated receptor-
activity by mammalian target of rapamycin and amino acids in adipogenesis. Diabetes
2004; 53, 27482756.
19. Viguerie, N., Poitou, C., Cancello, R., Stich, V., Clement, K., and Langin, D.
Transcriptomics applied to obesity and caloric restriction. Biochemie 2005; 87,
117123.
20. Novakofski, J. Adipogenesis: usefulness of in vitro and in vivo experimental models.
J Anim Sci 2004; 82, 905915.
21. Dulloo, A.G., Duret, C., Rohrer, D., Girardier, L., Mensi, N., Fathi, M., Chantre, P.,
and Vandermander, J. Efficacy of a green tea extract rich in catechin polyphenols and
caffeine in increasing 24-h energy expenditure and fat oxidation in humans. Am J
Clin Nutr 1999; 70, 10401045.
22. Stoclet, J.C., Chataigneau, T., Ndiaye, M., Oak, M.H., Bedoui, J.E., Chataigneau,
M., and Shini-Kert, V.B. Vascular protection by dietary polyphenols. Eur J Pharmacol
2004; 500, 299313.
23. Bagli, E., Stefaniotou, M., Morbidelli, L., Ziche, M., Psillas, K., Murphy, C., and
Fosis, T. Luteolin inhibits vascular endothelial growth factor-induced angiogenesis;
inhibition of endothelial cell survival and proliferation by targeting phosphatidi-
linositol 3-kinase activity. Cancer Res 2004; 64, 79367946.
24. Cao, Y. and Cao, R. Brakenhielm Antiangiogenic mechanism of diet-derived polyphe-
nols. J Nutr Biochem 2002; 13, 380390.
25. Mennen, L.I., Walker, R., Bennetau-Pelissero, C., and Scalbert, A. Risk and safety
of polyphenol consumption. Am J Clin Nutr 2005; 81, 326S329S.
26. Dembinska-Kiec, A., Polus, A., Kiec-Wilk, B., Grzybowska, J., Mikolajczyk, M.,
Hartwich, J., Razny, U., Szumilas, K., Banas, A., Bodzioch, M., Stachura, J., Dyduch,
G., Laidler, P., Zagajewski, J., Langman, T., and Schmitz, G. Proangiogenic activity
of beta-carotene is coupled with the activation of endothelial cell chemotaxis. Biochim
Biophys Acta May 30, 2005; 1740(2): 22239.
27. Luisa Bonet Murano, I., Morroni, M., Zingaretti, M.C., Oliver, P., Sanchez, J., Fuster,
A., Pico, C., Palou, A., and Cinti, S. Morphology of ferret subcutaneous adipose
tissue after 6-month daily supplementation with oral beta-carotene. Biochim Biophys
Acta May 30, 2005; 1740(2): 30512. Epub November 17, 2004.
28. von Lintig, J., Hessel, S., Eichnger, A., Isken, A., Oberhauser, V., Vogt, K., Goralczyk,
R., and Wyss, A. Analysis of Bcmo1 knock-out mouse model uncovers a role of -
carotene for the regulation of lipid metabolism. In Oxidants and Antioxidants in
Biology. Abstract book of the Oxygen Club of California. Santa Barbara; March
1518, 2006, p 21.
29. Balasubramanian, S.P., Brown, N.J., and Reed, M.W.R. Role of genetic polymor-
phisms in tumor angiogenesis. Br J Cancer 2002; 87, 10571065.
30. Wang, S., Yehya, N., Schardt, E.E., Wang, H., Drake, T.A., and Lusis, A.J. Genetic
and genomic analysis of a fat mass trait with complex inheritance reveals marked
sex specificity. PLoS Genetics /www.plosgenetix.org; 2006; 2, 01480153.
9281_C008.fm Page 100 Wednesday, July 18, 2007 1:59 PM
9281_C009.fm Page 101 Wednesday, July 18, 2007 2:01 PM
9 Metabolic Programming
during Pregnancy:
Implications for
Personalized Nutrition
Simon C. Langley-Evans
CONTENTS
9.1 INTRODUCTION
This chapter will introduce the concept that nutritional factors before birth impact
disease risk in later life. Evidence from epidemiologic and experimental studies
demonstrates that variation in nutrition during fetal development exerts a program-
ming effect on tissue development and metabolic/physiological status in adult life.
These programming effects have profound implications for the attainment of meta-
bolic phenotypes that promote the development of the metabolic syndrome and
associated disease outcomes. There is growing evidence of interactions between the
genotype and indicators of fetal growth and nutrition in determining this disease
risk phenotype. For example, a well-characterized association between low birth
weight and risk of type 2 diabetes in adult life is dependent on the pro12ala
polymorphism of the peroxisome proliferators activated receptor 2. Similarly, the
established association between the Bsm1 polymorphism of the vitamin D receptor
101
9281_C009.fm Page 102 Wednesday, July 18, 2007 2:01 PM
(Table 9.1). Low birth weight and disproportion at birth are essentially simple
markers of constrained growth in utero. Such growth impairment is therefore pro-
posed to be central to programming of disease risk, but the effects of intrauterine
growth restriction are also modulated by postnatal growth rates. Thus, the individual
at greatest risk of metabolic syndrome is likely to have been born small but undergone
rapid catch-up growth in infancy, attaining an increased body mass index in adolescence
(Eriksson et al., 2003a).
The relationship between risk of coronary heart disease, non-insulin-dependent
diabetes, and birth weight has been attributed to the effects of variation in maternal
nutritional status on fetal growth. However, this explanation at first glance appears
tenuous as in human populations the association between maternal nutrient intake and
birth weight is difficult to demonstrate (McMillen and Robinson, 2005). It is estimated
that maternal nutrient intake only explains approximately 6% of the variation in birth
weight. In extreme situations, such as the wartime Dutch Hunger Winter and siege of
Leningrad, starvation had only minor effects on birth weight. In well-nourished pop-
ulations, although isolated studies show that intakes of animal protein or sucrose are
related to birth weight and placental weight, the majority of studies find no significant
relationships between maternal diet and weights of either baby or placenta.
Although the lack of clear associations between maternal nutrient intake and
birth weight, or proportions at birth, appears to undermine the nutritional program-
ming hypothesis, there are important studies that directly demonstrate associations
between maternal nutritional status in pregnancy and disease-risk markers in the
resulting offspring. A Jamaican study showed that higher blood pressure in prepu-
bescent boys was predicted by low maternal hemoglobin, weight gain, and triceps
skin fold thicknesses in pregnancy (Godfrey et al., 1994). A study of men born in
Aberdeen in the 1950s found that blood pressure was related to maternal intakes of
animal protein and of carbohydrate (Campbell et al., 1996). Data from the ongoing
TABLE 9.1
Associations between Anthropometric Indices at Birth and Markers
of Disease Risk in Human Populations
Birth Characteristic Associated Disease State Associated Risk Factor
Low birth weight Coronary heart disease, type 2 Obesity, hypertension, insulin
diabetes, asthma, COPD, resistance, poor response to
osteoporosis, depression, vaccination, metabolic syndrome,
schizophrenia, stroke, cataracts raised clotting factors, raised LDL-
cholesterol
Low ponderal indexa Type 2 diabetes, schizophrenia Insulin resistance, hypertension,
hypercortisolism
Reduced abdominal Hypertension, raised LDL-
circumference cholesterol, hyperinsulinemia
Large head circumferenceb Asthma, eczema Raised IgE
a Ponderal index is a marker of relative thinness at birth (birth weight, kg/length, m3).
b Corrected for crown-heel length. COPD = chronic obstructive pulmonary disease.
9281_C009.fm Page 104 Wednesday, July 18, 2007 2:01 PM
Project Viva study in the U.S. also appear to support the concept of nutritional
programming. For example, higher maternal calcium intakes in pregnancy result in
lower blood pressure in 6-month-old babies (Gillman et al., 2004).
The inconsistencies in the literature relating to maternal diet and birth weight
almost certainly arise because of the lack of appropriate methodology to assess the
nutrient supply the fetus actually gets. This is partly determined by maternal intake,
but will also depend on maternal stores going in to pregnancy, the metabolic demands
of the mother, and the perfusion of the placenta. These are all problems that are best
addressed by experimental animal models. These have been widely used to investi-
gate the plausibility of the programming concept and to consider the mechanistic
basis of nutritional programming.
140
120
Systolic BP (mm Hg)
100
80
Male
Female
60
40
20
0
Control LP d0-7 LP d8-14 LP d15-22 LP d0-22
Maternal diet
FIGURE 9.1 Systolic blood pressure in 4-week-old rats exposed to low-protein diet during
specific periods of pregnancy. Undernutrition at any stage of gestation increases later blood
pressure, and these effects persist into adulthood. Figure reproduced from data published in
Langley-Evans et al. (1996). LP = low-protein diet. Full rat gestation is 22 d (d 0 to 7 represents
targeting of protein restriction to early gestation, d 8 to 14 to midgestation, d 15 to 22 to late
gestation). * denotes P < 0.05 compared to control group.
9281_C009.fm Page 105 Wednesday, July 18, 2007 2:01 PM
observations in large animal species, such as the sheep, suggest that programming
of cardiovascular function occurs in all mammals.
A broad spectrum of nutritionally programmed disorders have been reported
from animal studies, but in general such experiments uncover blood pressure, glucose
intolerance, insulin resistance, and increased adiposity as common endpoints of
nutritional interventions targeted at pregnancy, or specific periods in pregnancy
(Table 9.2). It is becoming increasingly apparent that in addition to programming
disease states or their direct risk factors (glucose intolerance, adiposity, and hyper-
tension), alterations to the quality or quantity of rodent diets in pregnancy determine
behavioral antecedents of metabolic risk. For example, food restriction and protein
TABLE 9.2
Programming Effects of Maternal Undernutrition in Animal Models
Observed Phenotype Maternal Diet Manipulation
in Offspring in Pregnancy Species
Hypertension Food restriction Rat, sheep, and guinea pig
Low-protein diet Rat and mouse
Uterine artery ligation Rat
High saturated fat Rat
Iron deficiency Rat
Calcium deficiency Rat
restriction in rat pregnancy have both been shown to impact feeding and locomotor
behavior in the offspring (Vickers et al., 2003).
Working with species that have relatively short life span and gestation allows
evaluation of intergenerational effects and consideration of interactions between
dietary exposures at different developmental stages in pre- and postnatal life. Using
this approach, it has been possible to show that although the combination of under-
nutrition before birth combined with overnutrition in adulthood does not increase
risk of obesity, the introduction of overnutrition in the early postnatal (suckling and
weaning stages) following prenatal restriction can induce extreme central adiposity
(Langley-Evans et al., 2005). Such animal models, although varying in the detailed
approaches adopted, importantly underpin the nutritional programming hypothesis,
demonstrate that programming of cardiovascular changes can occur even in the
absence of fetal growth retardation, and provide useful tools with which to study
the mechanisms involved in nutritional programming.
PPAR genotype
25
Homa-IR index
20
15
10
5 PP
0 PA or AA
<3.0 3.03.5 >3.5
Birthweight (kg)
FIGURE 9.2 The interaction of genotype for PPAR with birth weight in determining adult
insulin sensitivity. Subjects genotyped according to the Pro12Ala polymorphism in the PPAR
gene were grouped according to weight at birth. The Pro12Pro (PP) variant was associated with
raised HOMA-IR only in subjects in the two lowest birth weight groups (P < 0.005 comparing
PP and PA or AA genotype for birth weight less than 3.0 kg, P = 0.03 for birth weight 3.0 to
3.5 kg). Birth weight was only related to adult HOMA-IR (P = 0.002) in the population subset
with the PP genotype. Figure produced from data reported by Eriksson et al., 2002.
allele to have higher blood pressure and to require hypertensive medication, but once
again this detrimental effect of the genotype was observed only in individuals who
had been of lower weight or shorter stature at birth (Yliharsa et al., 2004).
Overall, these studies of the Pro12Ala polymorphism in PPAR suggest that a
single genotype can give rise to different phenotypes because of variation in early
life experience. This evidence of programmed modulation of the effects of disease-
related alleles is of great importance when considering strategies for personalized
nutrition.
The same considerations are clearly indicated by studies of the interaction between
early life characteristics and the effects of the K121Q polymorphism of plasma cell
glycoprotein 1 (PC1). This is a strong candidate gene for non-insulin-dependent
diabetes, as PC1 is an inhibitor of insulin signaling downstream from the insulin
receptor. The K121Q polymorphism in exon 4 of this gene is associated with
variation in the inhibitory effects of PC1. The 121K variant lowers the inhibitory
action, whereas 121Q enhances the inhibition and is consequently associated with
insulin resistance, raised plasma glucose, and hyperinsulinemia. In the study of
Kubaszek et al. (2004) no variation in insulin concentrations or HOMA-IR index
was noted between individuals with a K121K genotype and those with at least one
121Q allele. However, once grouped by weight at birth, it was apparent that the
121Q allele had adverse effects. Adults that had the lowest weights at birth had
greater insulin concentrations and HOMA-IR index if they carried the 121Q variant.
Most importantly, although there was no variation in prevalence of diabetes or
hypertension that could be attributed solely to PC1 genotype, in individuals who
were of short stature at birth (birth length below 49 cm), the 121Q allele increased
prevalence of hypertension by threefold and type 2 diabetes by twofold. In this study
there was no apparent effect of the genotype until the interaction with a proxy of
fetal growth was considered in the analysis. Thus, without anthropometric data from
9281_C009.fm Page 110 Wednesday, July 18, 2007 2:01 PM
birth, genotyping for the PC1 polymorphism would be of little use in the application
of personalized nutritional advice.
Human angiotensin converting enzyme (ACE) has an insertion/deletion (I/D)
polymorphism that appears to impact both circulating and tissue activities of the
enzyme. The insertion is a 287 bp fragment, the presence of which lowers ACE
activity and apparently increases risk of vascular injury in diabetic subjects (Kajantie
et al., 2004). However, the effects of this very common allele (the ID form pre-
dominates in populations, whereas II is the rarest variant) are inconsistent and, as
with the PPAR Pro12Ala polymorphism, this is probably explained by geneenvi-
ronment factors. Kajantie et al. (2004) found that the DD variant was associated
with lower weight, smaller head circumference, and shorter length at birth, imply-
ing that this variant of ACE plays a role in regulation of fetal growth. In adult
life, subjects with the II variant exhibited greater insulin responses to a glucose
tolerance test than those with DD, but this difference was only observed in those
of lower weight at birth. In this case the polymorphic allele may confound the
association between birth weight and disease, because the I allele is itself associ-
ated with shorter gestation and increased weight at birth. However, this study is
a further example of how the effects of a polymorphism on a diabetes risk marker
are modulated by early growth.
9.3.2 OSTEOPOROSIS
Mapping of the genetics of bone metabolism is well advanced, and a number of
putative risk-determining genes have been identified, including the vitamin D receptor
(VDR), the estrogen receptors, and the type 1 collagen A1 gene (Walker-Bone
et al., 2002). Polymorphisms at these loci have been identified, including 22 separate
polymorphisms of VDR, but population studies suggest that these explain only a
small proportion of variation in bone mass. For example, it is estimated that the BB
variant of the Bsm I restriction site in VDR can reduce site-specific bone mineral
density by at most 2%.
The influence of genotype on bone health is clearly modulated by lifestyle and
environmental factors. As with diabetes, any consideration of such factors has to
include some assessment of proxies for early life nutrition and growth. Several
studies have suggested that growth in utero and in the first year of infancy determine
osteoporosis and osteoarthritis (Walker-Bone et al., 2002). In a study of the Bsm 1
polymorphism of VDR, Jordan and colleagues (2005) reported that the B allele
increased severity of osteophytosis (the outgrowth of immature bony processes,
reflecting the presence of degenerative disease) in the lumbar region. In men, but
not women, there was an interaction of the VDR genotype with weight at birth in
determining the severity of the lumbar degeneration. Thus, fetal factors modified
the risk associated with this particular VDR genotype.
The tt variant of the Taq1 polymorphism of VDR is also associated with disease
risk. Individuals with a tt genotype have lower bone mineral density and greater
prevalence of fractures. Adult bone mineral density is strongly a function of the peak
bone mass attained in the third decade of life, and this to some extent depends on
earlier rates of growth. Keen et al. (1997) reported that the tt variant of VDR was
9281_C009.fm Page 111 Wednesday, July 18, 2007 2:01 PM
associated with weight at 1 year, suggesting that patterns of skeletal growth even in
the very early years are determined by the VDR genotype. The early environment
may therefore interact with genotype to determine rates of skeletal growth, and the
two factors together program adult bone characteristics.
SNPs have also been identified in the growth hormone gene cluster, which in
addition to the growth hormone (GH1) gene, includes placental lactogen, placental
growth hormone, and the chorionic somatomammotrophin genes. Polymorphisms
of GH1 are associated with variability in bone mineralization. Dennison et al. (2004)
found that over a 4 year period, the loss of bone from the lumbar spine and proximal
femur was greatest in individuals with the 2 allele of the GHV1 and GHV4 loci.
Individuals with these unfavorable allele variants also exhibited blunted growth
hormone secretory profiles. However, these effects depended entirely on an interac-
tion with weight at age 1 year. Individuals in the lowest tertile of infant weight
showed a strong association between GHV1 genotype and rates of bone loss, whereas
in adults who had been heavier infants, GHV1 genotype had no effect on bone loss.
These data show that the environment encountered in infancy can modulate the effect
of GH1 genotype on bone health up to seven decades later.
FIGURE 9.3 Disease risk in adult life is a product of cumulative experience across the life
course. Geneenvironment interactions occur at all stages of life. The product of this interaction
in early life will determine the strength and nature of such responses in adulthood.
To date the emphasis of the research agenda in the field of metabolic program-
ming has been on demonstrating the biological principle and defining the mechanistic
basis of programming. The time has come to consider the possible nutritional
interventions that might reverse adverse programming effects. To construct appro-
priate personalized nutrition interventions, we require better-quality information
about the characteristics of a programmed disease-risk phenotype. More studies need
to utilize nutrigenomic tools, particularly proteomics and metabolomics, to focus on
the cellular and metabolic consequences of an adverse nutritional environment in
utero or during early infancy. One of the major challenges of applying these
approaches, at least in human studies, will be to separate the effects of early life
programming and the interaction of early life events with the genome from the
effects of lifestyle factors in adult life.
KEY READINGS
Barker, D.J.P. (1998). Mothers, babies and health in later life. Churchill Livingstone. London.
Gluckman, P.D. and Hanson, M.A. (2006). Developmental origins of health and disease.
Cambridge University Press. Cambridge, U.K.
Langley-Evans, S.C. (2004). Fetal Nutrition and Adult Disease: Programming of chronic
disease through fetal exposure to undernutrition. CABI. Wallingford, U.K.
9281_C009.fm Page 113 Wednesday, July 18, 2007 2:01 PM
REFERENCES
Barker, D.J.P. (2004). The developmental origins of adult disease. J Am Coll Nutr. 23(6
Suppl.), 588S595S.
Campbell, D.M., Hall, M.H., Barker, D.J., Cross, J., Shiell, A.W., and Godfrey, K.M. (1996).
Diet in pregnancy and the offsprings blood pressure 40 years later. Br J Obstet
Gynaecol. 103, 273280.
Dennison, E.M., Syddall, H.E., Rodriguez, S., Voropanov, A., Day, I.N., and Cooper, C.
(2004). Polymorphism in the growth hormone gene, weight in infancy, and adult bone
mass. J Clin Endocrinol Metab. 89, 48984903.
Eriksson, J.G., Lindi, V., Uusitupa, M., Forsen, T.J., Laakso, M., Osmond, C., and Barker,
D.J. (2002). The effects of the Pro12Ala polymorphism of the peroxisome proliferator-
activated receptor-gamma2 gene on insulin sensitivity and insulin metabolism interact
with size at birth. Diabetes 51, 23212324.
Eriksson, J.G., Forsen, T.J., Osmond, C., and Barker, D.J. (2003a) Pathways of infant and
childhood growth that lead to type 2 diabetes. Diabetes Care. 26, 30063010.
Eriksson, J., Lindi, V., Uusitupa, M., Forsen, T., Laakso, M., Osmond, C., and Barker, D. (2003b).
The effects of the Pro12Ala polymorphism of the PPARgamma-2 gene on lipid metab-
olism interact with body size at birth. Clin Genet. 64, 366370.
Gillman, M.W., Rifas-Shiman, S.L., Kleinman, K.P., Rich-Edwards, J.W., and Lipshultz, S.E.
(2004). Maternal calcium intake and offspring blood pressure. Circulation 110,
19901995.
Gluckman, P.D. and Hanson, M.A. (2004). Living with the past: evolution, development, and
patterns of disease. Science 305, 17331736.
Godfrey, K.M., Forrester, T., Barker, D.J., Jackson, A.A., Landman, J.P., Hall, J.S., Cox, V.,
and Osmond, C. (1994). Maternal nutritional status in pregnancy and blood pressure
in childhood. Br J Obstet Gynaecol. 101, 398403.
Jordan, K.M., Syddall, H., Dennison, E.M., Cooper, C., and Arden, N.K. (2005). Birthweight,
vitamin D receptor gene polymorphism, and risk of lumbar spine osteoarthritis. J
Rheumatol. 32, 678683.
Kajantie, E., Rautanen, A., Kere, J., Andersson, S., Yliharsila, H., Osmond, C., Barker, D.J.,
Forsen, T., and Eriksson, J. (2004). The effects of the ACE gene insertion/deletion
polymorphism on glucose tolerance and insulin secretion in elderly people are modified
by birth weight. J Clin Endocrinol Metab. 89, 57385741.
Keen, R.W., Egger, P., Fall, C., Major, P.J., Lanchbury, J.S., Spector, T.D., and Cooper, C.
(1997). Polymorphisms of the vitamin D receptor, infant growth, and adult bone mass.
Calcif Tissue Int. 60, 233235.
Kubaszek, A., Markkanen, A., Eriksson, J.G., Forsen, T., Osmond, C., Barker, D.J., and
Laakso, M. (2004). The association of the K121Q polymorphism of the plasma cell
glycoprotein-1 gene with type 2 diabetes and hypertension depends on size at birth.
J Clin Endocrinol Metab. 89, 20442047.
Langley-Evans, S.C., Welham, S.J.M., Sherman, R.C., and Jackson, A.A. (1996) Weanling
rats exposed to maternal low protein diets during discrete periods of gestation exhibit
differing severity of hypertension. Clin Sci. 91, 607615.
Langley-Evans, S.C., Bellinger, L., and McMullen, S. (2005). Animal models of program-
ming. Maternal Child Nutr 1, 142148.
Langley-Evans, S.C. (2004). Fetal programming of adult disease: an overview. In Fetal
nutrition and adult disease: Programming of chronic disease through fetal exposure
to undernutrition. Ed. S.C. Langley-Evans. CABI, Wallingford, pp. 120.
9281_C009.fm Page 114 Wednesday, July 18, 2007 2:01 PM
Langley-Evans, S.C. (2006). Developmental programming of health and disease. Proc Nutr
Soc. 65, 97105.
Lillycrop, K.A., Phillips, E.S., Jackson, A.A., Hanson, M.A., and Burdge, G.C. (2005). Dietary
protein restriction of pregnant rats induces and folic Acid supplementation prevents
epigenetic modification of hepatic gene expression in the offspring. J Nutr. 135,
382386.
McMillen, I.C. and Robinson, J.S. (2005). Developmental origins of the metabolic syndrome:
prediction, plasticity, and programming. Physiol Rev. 85, 571633.
Seckl, J.R. and Meaney, M.J. (2004). Glucocorticoid programming. Ann NY Acad Sci. 1032,
6384.
Vickers, M.H., Breier, B.H., McCarthy, D., and Gluckman, P.D. (2003). Sedentary behavior
during postnatal life is determined by the prenatal environment and exacerbated by
postnatal hypercaloric nutrition. Am J Physiol Regul Integr Comp Physiol. 285,
R271273.
Walker-Bone, K., Walter, G., and Cooper, C. (2002). Recent developments in the epidemiology
of osteoporosis. Curr Opin Rheumatol.14, 411415.
Waterland, R.A. and Jirtle, R.L. (2003). Transposable elements: targets for early nutritional
effects on epigenetic gene regulation. Mol Cell Biol. 23, 5293300.
Weaver, I.C., Cervoni, N., Champagne, F.A., DAlessio, A.C., Sharma, S., Seckl, J.R., Dymov,
S., Szyf, M., and Meaney, M.J. (2004). Epigenetic programming by maternal behavior.
Nat Neurosci. 7, 847854.
Yliharsila, H., Eriksson, J.G., Forsen, T., Laakso, M., Uusitupa, M., Osmond, C., and Barker,
D.J. (2004). Interactions between peroxisome proliferator-activated receptor-gamma
2 gene polymorphisms and size at birth on blood pressure and the use of antihyper-
tensive medication. J Hypertens. 22, 12831287.
9281_C010.fm Page 115 Thursday, July 26, 2007 7:07 PM
CONTENTS
10.1 INTRODUCTION
Human food preferences and intake are determined by a complex mix of innate,
acquired, and contemporary societal factors. Understanding the individual drivers
that determine what food is consumed and in which quantities, how it is prepared,
and when it is eaten should provide avenues for successful dietary advice and
interventions aimed at decreasing disease risk and achieving healthier aging. This
chapter reviews the current knowledge regarding the genetics of taste with a focus
on bitter taste perception and discusses how genetic variation in taste may be linked
115
9281_C010.fm Page 116 Thursday, July 26, 2007 7:07 PM
to the consumption of foods associated with disease protection and increased well-
being, as well as avoidance of habits associated with increased risk of disease. Better
understanding of the role of taste in dietary behavior may lead to the identification
of genetic markers that can be utilized for the development of personalized dietary
recommendations based on individual taste preferences. Such personalized guide-
lines may improve adherence to dietary recommendations and provide better
preventive strategies for chronic diseases.
In many countries, chronic diseases such as cardiovascular disease and cancer
are the leading causes of mortality. Moreover, the prevalence of conditions such as
metabolic syndrome and obesity are on the rise worldwide. Although these diseases
have complex etiologies, diet is thought to be a major contributing factor. One of
the greatest public health challenges is to alter eating behavior in populations as a
means of preventing chronic diseases. Adherence to dietary recommendations such
as increasing fruit and vegetable consumption proves to be difficult for many people.
Although there are various environmental and physiological factors that determine
individual food selection, taste has been suggested as an important factor in consumer
food choice. There is a wide variety of taste preferences in the population, and part
of this variability is genetically determined [1]. It is of interest to elucidate whether
there are genetic markers of taste that predispose a person to consume a certain type
of diet. Such information would be useful to identify people who are at risk for
diseases based on their taste genotype. The genetic markers may provide the
foundations for tailored recommendations based on their preferences.
are some naturally bitter compounds in foods that have been shown to have beneficial
effects on health. Some of these compounds include phenols (found in tea, citrus fruits,
wine, and soy), triterpenes (citrus fruits), and organosulfur compounds (cruciferous
vegetables) [3]. These phytochemicals have been investigated for their putative
anticarcinogenic and antioxidant properties. Various epidemiologic studies have
shown that increased intake of fruit and vegetables is associated with decreased risk
of chronic diseases such as heart disease and cancer. Public health efforts to increase
fruits and vegetable intake have been challenging, and the resistance to increased
consumption may be in part due to the bitter tastes of these foods [4]. Indeed, many
of the healthy vegetables such as cruciferous vegetables are often disliked for their
bitter taste, especially by children [3].
For this reason, it has been hypothesized that variability in bitter taste perception
may influence diet-related health outcomes. Much information investigating the
association between bitter-taste sensitivity and food preferences has centered on the
ability to taste a thiourea compound, phenylthiocarbamide (PTC). The N-C=S group
in PTC is responsible for its bitter taste [5]. Although PTC is not found in nature,
there are chemically related compounds such as isothiocyanates in cruciferous veg-
etables [6]. These bitter compounds often play a protective role for the plants. For
example, glucosinolates are natural pesticides found in cruciferous vegetable and,
when the plant gets damaged, they are hydrolyzed to goitrin, an isothiocyanate [3].
The abundance of bitter compounds in our food supply makes PTC sensitivity an
ideal marker for the study of taste, food preferences, and dietary behaviors.
146F, 136M Food preference and Twenty-five items that Tasting of PTC saturated No Mattes
(1722 yr) consumption inhibit thyroidal iodine filter paper 1989 [26]
questionnaire uptake, and six bitter foods
17F, 17M Verbal food preference Eight tasted foods, order- Forced choice threshold Sensory: Tasters disliked cheese and liked Anliker
(57 yr) questionnaire and sensory by-choice and 60 food detection and whole milk 1991 [17]
perception checklist suprathreshold method
9281_C010.fm Page 119 Thursday, July 26, 2007 7:07 PM
53F Sensory rating Tasted soy products and Forced choice threshold Sensory: Tasters perceived green tea to be Gayathri
(2045 yr) different dilutions of detection and more bitter and dislike plain tofu, 1997 [15]
green tea suprathreshold method preferred vanilla flavor soymilk
45F, 30M Sensory rating Fats (10 vs. 40% salad Suprathreshold method Sensory: Tasters reported more oral burn Tepper
Taste as the Gatekeeper of Personalized Nutrition
(1851 yr) dressing) from capsaicin and distinguished fat 1997 [19]
capsaicin content
123F Food preference Rated fruits and juices Suprathreshold method Sensory: Tasters disliked naringin and Drewnowski
(2060 yr) questionnaire Sensory testing of naringin grapefruit juice 1997 [18]
Sensory rating solution
118F Sensory and hedonic rating Fifteen Fat/sugar mixtures Forced choice detection Sensory: No Drewnowski
(2040 yr) threshold suprathreshold 1998 [23]
method
159F Food preference checklist One hundred and seventy- Suprathreshold method Sensory: Tasters had lower preference for Drewnowski
(2060 yr) 3-d food record (86 one items from various brussels sprouts, cabbage, spinach, and 1999 [10]
subjects) food categories coffee
Intake: No
(continued)
119
120
50F Sensory rating Six bitter foods tasted Suprathreshold method Sensory: Taste perception is associated Kaminski
(2040 yr) FFQ Twenty-two bitter food Tasting PROP saturated with bitterness of food 2000 [14]
items questionnaire paper Intake: No
24F, 22M Food preference survey Eighty-three items Forced choice threshold Sensory: In women sweet and high-fat Duffy
(1840 yr) detection and foods negatively correlated with bitter 2000 [21]
suprathreshold method sensitivity: in men a positive correlation
9281_C010.fm Page 120 Thursday, July 26, 2007 7:07 PM
was found
326F Food preference checklist One hundred and seventy- Suprathreshold method Sensory: Tasters dislike cruciferous, green, Drewnowski
(mean 47.651.7) one food items raw vegetables 2000 [16]
54F Sensory rating Sucrose, caffeine, Suprathreshold method Sensory: Tasters perceived caffeine as Ly
(1830 yr) chocolate Tasting PROP saturated being more bitter and disliked 2001 [12]
paper
92F, 55M Sensory rating Fattiness, saltiness, Suprathreshold method Sensory: No Yackinous
(1736 yr) sweetness of four foods 2001 [22]
85F, 38M Food questionnaire for Forty-three food items Taste recognition threshold Sensory: Tasters rated foods higher than Pasquet
(1959 yr) flavor, cost, effect on representative of Tunisian Suprathreshold method nontasters overall; no differences in bitter 2002 [13]
health diet foods except green tea, which tasters
disliked
33F, 34M Sensory test Tasted ten common Single concentration of Sensory: Tasters disliked broccoli, full-fat Keller
(45 yr) FFQ (child and parent) foods/drinks that are bitter PROP (yes/no) milk, American cheese 2002 [20]
or varying in fat 114 items Taster girls disliked full-fat milk
(FFQ) Intake: Girl tasters had lower intake of
discretionary fat
Personalized Nutrition: Principles and Applications
114F, 69M FFQ Ninety-five items Suprathreshold method Intake: No difference in men Yachinous
(1736 yr) Women tasters obtained lower percentage 2002 [28]
of energy from protein and fruit servings,
higher from fat
Supertasters consumed less green salad
compared to T, MT*
22F,14M Food intake Ad libitum intake of high- Suprathreshold method Intake: Taster had higher fat intake, lower Kamphuis
(mean age 31) fat, high-carb, or mixed CHO intake ad libitum of mixed meal 2003 [27]
meal No difference for high-fat or high-carb
meal
71F, 39M FFQ, food sampling Block 98.1 Suprathreshold method Sensory: Tasters rated sampled bitter Dinehart
(1860 yr) vegetables as most bitter (brussels 2006 [25]
sprouts, asparagus, kale). No association
9281_C010.fm Page 121 Thursday, July 26, 2007 7:07 PM
and unpleasant [30]. Taste status has also been linked with smoking habits [31]. In
these studies the proportion of nontasters was greater in smokers compared to
nonsmokers. It has been hypothesized that nontasters have less aversion to the
bitterness of cigarettes and are thus at greater risk to smoke. Together, these data
suggest that PTC/PROP sensitivity may mitigate the development of addictive behav-
iors such as smoking and alcoholism.
sequence analysis of TAS2R16 shows that the K172N polymorphism in the gene is
under positive selection based on the observation of high frequency of the evolu-
tionary derived variants in populations worldwide [44]. This result suggests that this
receptor has adapted according to unique human environments. The K172N poly-
morphism lies in the third extracellular loop and functional analysis shows that the
ancestral K172 allele has weaker response to ligands such as salicin, arbitun, and
amygdalin than the evolutionary derived N172 allele. Consumption of foods con-
taining these cyanogenic glycosides has been hypothesized to be protective against
malaria [44]. Consistent with this hypothesis, the K172 allele has been found at
higher frequencies where malaria is endemic. In addition, the K172 has been asso-
ciated with increased risk for alcohol dependence [45]. Because TAS2R38 has also
been associated with alcohol consumption and both TAS2R16 and TAS2R38 are
located on neighboring regions on chromosome 7, it would be interesting to examine
both of these genes in relation to alcohol intake.
In an effort to identify genes associated with myocardial infarction (MI), an associ-
ation study examining 11,053 polymorphisms representing 6,891 genes was conducted
[46]. To identify the variants significantly associated with MI, three case control studies
were analyzed sequentially. In the final analysis, five genes were significantly asso-
ciated with MI, one being TAS2R50 with odds ratio of 1.58 for the susceptible allele.
Interestingly, another gene identified in this study was one of the olfactory receptors,
OR13G1. The authors hypothesized that the association observed for both TAS2R50
and OR13G1 may be through dietary choices. Further studies are needed to confirm
this hypothesis.
With the exception of the known alleles of TAS2R38 and the sensitivity to the
bitterness of PTC and related compounds, there are no known phenotypegenotype
relationships between alleles of the other bitter receptors and the taste perception
of their bitter ligands. However, there are many examples of a wide range of
perceptual sensitivity by human populations to other bitter stimuli. Although PTC
and TAS2R38 are the first example of the effect of genetics on taste perception, the
complement of taste receptors and their many alleles will probably reveal numerous
relationships with bitter taste perception and the motivation to consume bitter foods.
binding of the sweetener to the receptor, which is composed of two subunits, TAS1R2
and TAS1R3 (for a review of the discovery and biology of this receptor, see Reference
36). A second subunit (TAS1R1) when paired with TAS1R3, the common partner,
is responsible for the transduction of umami (savory) taste. Alleles of TAS1R3 are
strongly associated with sweet intake in the mouse, and it has been hypothesized
that alleles of human T1Rs may explain part of the variability of the intake of sugars.
These phenotypegenotype studies have yet to be conducted in humans, although
there are many known alleles in these taste receptors, especially the subunit special-
ized to sweet ligands. The umami-specific subunit of the receptor, TAS1R1, also
contains many polymorphisms, and might be related to the sensory blindness to the
savory taste of MSG experienced by a subset of people [47].
10.4.2 SOUR
Individual differences in the ability to perceive sourness exist, but the genetic basis
of this taste quality is unknown. The recent discovery of a key protein in sour taste
in mice provides the foundations to investigate the mechanisms of sour taste trans-
duction and the role of this taste modality on human dietary behavior and health
[48]. Unripe fruit, types of milk and cheese and some vegetables are sour, and people
often confuse sour and bitter, which contributes to the perception of a sour food as
undesirable. Because many nutritious foods are slightly sour, its perception is an
aspect of food selection that is understudied relative to its importance to human
health.
10.4.3 SALT
The perception of saltiness differs from the other taste qualities because differences
among people in sodium threshold and intensity ratings of saltiness are marked, but
are related to immediate food intake rather than to genotype. Although some aspects
of sodium transduction are understood, the search for the full complement of proteins
that participate in this process is still on. It is possible that genetic differences among
people are important, but it could also be that salt perception is labile, and the genetic
differences would only be apparent if all subjects were fed diets equivalent in
saltiness.
10.4.4 FAT
In addition to the basic tastes and their receptor genes, examination of unconventional
taste qualities will likely be important in understanding human diet and health.
Because fat is important to the sensory experience of food, differences in fat taste
perception may influence dietary behavior. Several lines of evidence suggest there
might be genetic determinants of oral fat perception, the most convincing of which
is the observation that fat intake in humans is heritable [1]. There is debate about
whether animals and humans have a sensory system that is specifically tuned to oral
fats, largely because the receptor and transduction mechanisms are not known.
Chewing or tasting fatty foods without swallowing is sufficient to stimulate digestion,
which is evidence that fats are sensed on the tongue [49]. Whether fat perception is
9281_C010.fm Page 127 Thursday, July 26, 2007 7:07 PM
and 20q13.3 for total energy intake, and 12q14.1 for total fat intake. In African
Americans, suggestive linkage found at 1q43-44 for sucrose intake, and regions
1p22.3, 6q22.31, and 12q24.21 for fat intake. No specific candidate genes were
examined in the study; therefore, the genes underlying the linkage are yet to be
identified. It is noteworthy that the linkage observed in the studies were specific to
each ethnic group. This may be due to the differences in the methodologies for
dietary assessment or linkage analyses used by the two studies. Or it may be an
indication of different genetic architecture of dietary behavior by ethnicity. These
studies provide clues regarding the regions of the genome that may contribute to
macronutrient intake and may be the initial steps toward identifying novel genes
involved in dietary behavior.
ACKNOWLEDGMENTS
We thank Dr. Carol Christensen and Dr. Marcia Levin Pelchat for their generous
contributions in the preparation of this manuscript.
9281_C010.fm Page 130 Thursday, July 26, 2007 7:07 PM
KEY READINGS
Herness, M. S. and Gilbertson, T. A., Cellular mechanisms of taste transduction, Annu Rev
Physiol, 61, 873900, 1999.
Mennella, J. A. et al., Genetic and environmental determinants of bitter perception and sweet
preferences, Pediatrics, 115, e21622, 2005.
Rankinen, T. and Bouchard, C., Genetics of food intake and eating behavior phenotypes in
humans, Annu Rev Nutr, 26, 41334, 2006.
Reed, D. R. et al., Diverse tastes: Genetics of sweet and bitter perception, Physiol Behav, 88,
21526, 2006.
Tepper, B. J., 6-n-Propylthiouracil: a genetic marker for taste, with implications for food
preference and dietary habits, Am J Hum Genet, 63, 12716, 1998.
REFERENCES
1. Reed, D. R. et al., Heritable variation in food preferences and their contribution to
obesity, Behav Genet, 27, 37387, 1997.
2. Herness, M. S. and Gilbertson, T. A., Cellular mechanisms of taste transduction, Annu
Rev Physiol, 61, 873900, 1999.
3. Drewnowski, A. and Gomez-Carneros, C., Bitter taste, phytonutrients, and the con-
sumer: a review, Am J Clin Nutr, 72, 142435, 2000.
4. Drewnowski, A. and Rock, C. L., The influence of genetic taste markers on food
acceptance, Am J Clin Nutr, 62, 50611, 1995.
5. Guo, S. W. and Reed, D. R., The genetics of phenylthiocarbamide perception, Ann
Hum Biol, 28, 11142, 2001.
6. Tepper, B. J., 6-n-Propylthiouracil: a genetic marker for taste, with implications for
food preference and dietary habits, Am J Hum Genet, 63, 12716, 1998.
7. Bartoshuk, L. M., Comparing sensory experiences across individuals: recent psycho-
physical advances illuminate genetic variation in taste perception, Chem Senses, 25,
44760, 2000.
8. Reed, D. R. et al., Propylthiouracil tasting: determination of underlying threshold
distributions using maximum likelihood, Chem Senses, 20, 52933, 1995.
9. Fox, A. L., The relationship between chemical constitution and taste., Proc Natl Acad
Sci, 18, 115120, 1932.
10. Drewnowski, A. et al., Taste and food preferences as predictors of dietary practices
in young women, Public Health Nutr, 2, 5139, 1999.
11. Reed, D., Behavioral modulation of the path from genotype to obesity phenotype,
John Libbey & Company, Paris, 1999, pp. 199208.
12. Ly, A. and Drewnowski, A., PROP (6-n-Propylthiouracil) tasting and sensory responses
to caffeine, sucrose, neohesperidin dihydrochalcone and chocolate, Chem Senses, 26,
417, 2001.
13. Pasquet, P. et al., Relationships between threshold-based PROP sensitivity and food
preferences of Tunisians, Appetite, 39, 16773, 2002.
14. Kaminski, L. C. et al., Young womens food preferences and taste responsiveness to
6-n-propylthiouracil (PROP), Physiol Behav, 68, 6917, 2000.
15. Gayathri Devi, A. et al., Sensory acceptance of Japanese green tea and soy products
is linked to genetic sensitivity to 6-n-propylthiouracil, Nutr Cancer, 29, 14651, 1997.
16. Drewnowski, A. et al., Genetic taste markers and preferences for vegetables and fruit
of female breast care patients, J Am Diet Assoc, 100, 1917, 2000.
9281_C010.fm Page 131 Thursday, July 26, 2007 7:07 PM
17. Anliker, J. A. et al., Childrens food preferences and genetic sensitivity to the bitter
taste of 6-n-propylthiouracil (PROP), Am J Clin Nutr, 54, 31620, 1991.
18. Drewnowski, A. et al., Taste responses to naringin, a flavonoid, and the acceptance
of grapefruit juice are related to genetic sensitivity to 6-n-propylthiouracil, Am J Clin
Nutr, 66, 3917, 1997.
19. Tepper, B. J. and Nurse, R. J., Fat perception is related to PROP taster status, Physiol
Behav, 61, 94954, 1997.
20. Keller, K. L. et al., Genetic taste sensitivity to 6-n-propylthiouracil influences food
preference and reported intake in preschool children, Appetite, 38, 312, 2002.
21. Duffy, V. B. and Bartoshuk, L. M., Food acceptance and genetic variation in taste, J
Am Diet Assoc, 100, 64755, 2000.
22. Yackinous, C. and Guinard, J. X., Relation between PROP taster status and fat
perception, touch, and olfaction, Physiol Behav, 72, 42737, 2001.
23. Drewnowski, A. et al., Genetic sensitivity to 6-n-propylthiouracil and sensory
responses to sugar and fat mixtures, Physiol Behav, 63, 7717, 1998.
24. Jerzsa-Latta, M. et al., Use and perceived attributes of cruciferous vegetables in terms
of genetically-mediated taste sensitivity, Appetite, 15, 12734, 1990.
25. Dinehart, M. E. et al., Bitter taste markers explain variability in vegetable sweetness,
bitterness, and intake, Physiol Behav, 87, 30413, 2006.
26. Mattes, R. and Labov, J., Bitter taste responses to phenylthiocarbamide are not related
to dietary goitrogen intake in human beings, J Am Diet Assoc, 89, 6924, 1989.
27. Kamphuis, M. M. and Westerterp-Plantenga, M. S., PROP sensitivity affects macro-
nutrient selection, Physiol Behav, 79, 16772, 2003.
28. Yackinous, C. A. and Guinard, J. X., Relation between PROP (6-n-propylthiouracil)
taster status, taste anatomy and dietary intake measures for young men and women,
Appetite, 38, 2019, 2002.
29. Pelchat, M. L. and Danowski, S., A possible genetic association between PROP-
tasting and alcoholism, Physiol Behav, 51, 12616, 1992.
30. Intranuovo, L. R. and Powers, A. S., The perceived bitterness of beer and 6-n-
propylthiouracil (PROP) taste sensitivity, Ann NY Acad Sci, 855, 8135, 1998.
31. Enoch, M. A. et al., Does a reduced sensitivity to bitter taste increase the risk of
becoming nicotine addicted?, Addict Behav, 26, 399404, 2001.
32. Reed, D. R. et al., Localization of a gene for bitter-taste perception to human chro-
mosome 5p15, Am J Hum Genet, 64, 147880, 1999.
33. Drayna, D. et al., Genetic analysis of a complex trait in the Utah Genetic Reference
Project: a major locus for PTC taste ability on chromosome 7q and a secondary locus
on chromosome 16p, Hum Genet, 112, 56772, 2003.
34. Adler, E. et al., A novel family of mammalian taste receptors, Cell, 100, 693702, 2000.
35. Matsunami, H. et al., A family of candidate taste receptors in human and mouse,
Nature, 404, 6014, 2000.
36. Reed, D. R. et al., Diverse tastes: Genetics of sweet and bitter perception, Physiol
Behav, 88, 21526, 2006.
37. Kim, U. K. et al., Positional cloning of the human quantitative trait locus underlying
taste sensitivity to phenylthiocarbamide, Science, 299, 12215, 2003.
38. Bufe, B. et al., The molecular basis of individual differences in phenylthiocarbamide
and propylthiouracil bitterness perception, Curr Biol, 15, 3227, 2005.
39. Wooding, S. et al., Independent evolution of bitter-taste sensitivity in humans and
chimpanzees, Nature, 440, 9304, 2006.
40. Wooding, S. et al., Natural selection and molecular evolution in PTC, a bitter-taste
receptor gene, Am J Hum Genet, 74, 63746, 2004.
9281_C010.fm Page 132 Thursday, July 26, 2007 7:07 PM
11 Personalized Nutrition
and Public Health
Pieter vant Veer, Edith J.M. Feskens,
and Ellen Kampman
CONTENTS
11.1 INTRODUCTION
Personalized nutrition (PN) addresses the idea that optimal nutrition differs between
individuals as a result of their genetic makeup. One hundred years after Mendel and
50 years after Watson and Crick, the current deciphering of the human genome holds
the promise that knowledge of the genetic makeup and the omic technologies will
133
9281_C11.fm Page 134 Wednesday, July 18, 2007 3:03 PM
enable us to provide highly specific dietary advice fitting the nutritional requirements
of individuals.1
Nutritional requirements are traditionally based on physiological needs, e.g., for
growth and development, to maintain nutritional status, to minimize disease risk, or
to optimize life expectancy. Based on this biomedical approach, nutritional require-
ments and recommendations are provided for energy intake and nutrient needs for
specific populations according to age, sex, physical activity, health status, life cycle,
etc. Beyond these variations in requirements at the group level, variability in nutri-
tional requirements has been denoted as natural biological variation, stemming
from genetic variation, uncontrolled environmental factors, and measurement error.
Thus, effects of dietary factors may at least in part depend on inherited
individual susceptibility, allowing the strength of the diethealth equation to differ
between subjects. In this framework, biological variation in classical risk factors
such as blood pressure and blood lipids serve as phenotypical intermediates of
dietgene interactions.
When we apply this biomedical approach of PN to disease prevention and health
promotion, it enters the domain of public health, defined as the science and art of
preventing disease, prolonging life and promoting health through organized efforts
of society.2 To enhance public health, nutritional requirements and recommenda-
tions are translated into food patterns, which, at least implicitly, incorporate national
and local traditions and cultural issues, consumer acceptance, economic interests,
and feasibility. These dietary guidelines complete the sequence from requirements
to recommendations and, subsequently, to guidelines, and the process provides a
basis for evidence-based nutritional policy.
In the context of public health, PN can fulfill a role in effective implementation
of dietary guidelines at the individual level, i.e., in dietetics and health promotion.
Here, the challenges are similar to those of clinical nutritionists and dieticians
treating specific distortions in blood pressure, lipid metabolism, etc.; both in a
clinical and in a public health context, population nutrition recommendations
have to be translated to an individualized dietary optimum in everyday food
choice matching the lifestyle of the individual.
Thus, although the biomedical concept of PN is scientifically challenging and
may help to further uncover and define physiological requirements, it also has an
independent meaning in the domain of public health, where existing and future
nutritional requirements are applied to populations (public health) and individuals
(PN) aiming at disease prevention and health care.
In this contribution, we will delineate the link between PN and public health.
We will do this from the viewpoint of epidemiology. We will first address the role
of diet (and lifestyle) in chronic disease epidemiology at the global level and then
clarify the basic concept of dietgene interaction. Subsequently, developments in
the status quo on research in nutrient/dietgene interactions will be summarized for
colorectal carcinogenesis and diabetes, representing two pathological processes of
major importance to population health. Then, the so-called prevention paradox and
its implications for PN will be illustrated and knowledge gaps will be touched upon.
Finally, elements for a discussion framework on PN are proposed, and future pros-
pects are delineated.
9281_C11.fm Page 135 Wednesday, July 18, 2007 3:03 PM
ratio is high, the opposite is seen.14 In fact, this would suggest that on a low dietary
P:S ratio diabetes risk is larger in Ala carriers, and on a high P:S ratio diabetes risk
is larger in Pro/Pro homozygotes. As fatty acids are natural ligands for PPAR-gamma,
these findings await confirmation and further study; also, a lack of power and other
methodological issues make interpretation difficult.
6
GSTM1 GSTT1 EPHX
7%
5
RR
2
40% 17% 54%
46% 4%
15%
1
27% 23% 20% 35%
12%
0
Present Null Present Null Wild-type Mutant
Genotype Frequency
FIGURE 11.1 Graphical representation of population attributable risk [PAR (%), surface
area] as a function of relative risk (RR) and genotype frequency. Data were derived from a
case control study on liver cancer as related to aflatoxin-contaminated peanut butter and
genotypes of GSTM1, GSTT1, and EPHX, conducted in Sudan.
9281_C11.fm Page 141 Wednesday, July 18, 2007 3:03 PM
proportion of the population harboring the genotypes considered. Thus, for each
genotype, the case load is graphically represented by the surface given by the product
of the prevalence Pij and the RR of the nutritional exposure, here aflatoxin-
contaminated peanut butter (this is the case because we have used a median split
for aflatoxin exposure). For the three genes considered, aflatoxin reduction in the
whole population would prevent about 60% of all cases, independent of genotype
(we neglect the fact that the PARs for exposure to aflatoxin are not identical because
of weak associations between genotype and exposure and rounding errors). For the
highly prevalent GSTM1 and GSTT1 null polymorphisms, a substantial part of
the case load derives from the exposed and genetically nonsusceptible subgroup.
For the variant of the EPHX gene, only one tenth of all exposure-related cases are
derived from the category of the genetically susceptible.
This example illustrates that any single screening test, even when the mutant
genotype is highly prevalent (GSTs), tends to provide relatively small PARs for
exposure, whereas major opportunities to prevent disease in large segments of the
population are not seized. The RRs do not differ greatly according to genotype, and
the rare frequency of the high-risk genotype renders the case load due to the
interaction relatively small. As illustrated in the previous section, published
dietgene interactions in obesity, diabetes, and cardiovascular and malignant disease
all suggest that this example of liver cancer reflects a general phenomenon.
resulting from exposure and the genetic factor coincide. In this specific and uncom-
mon case of geneenvironment interaction, dietary modification (if possible) can
reduce the risk among the genetically susceptible. When RRN exceeds 1, the case
load emerging from the relative large size of the less susceptible genetic subpop-
ulation does not justify an individualized approach that only targets this subgroup,
unless both the prevalence of the high-risk genotype is large and RRNG is large as
well.
The foregoing observations become highly relevant if results on different genes
are considered simultaneously. The study on liver cancer could provide a hypothetical
example: One could speculate that if the genes mentioned are independent and
if the RRs are multiplicative that the RR of liver cancer for exposure to aflatoxin
contaminated peanut butter could rise to RR = 2.7 * 2.3 * 7.7 = 48, which would
be applicable to a population segment of 0.44 * 0.41 * 0.04 = 0.007, resulting in a
very low PAR. Thus, when several genes are addressed simultaneously, the RRs and
individual benefits can be large. However, the higher the RR, the smaller the pop-
ulation segment it applies to. Finally, any of the data in this example should be used
with caution, both because of uncertainties due to sampling and because of uncer-
tainties with respect to independence of the polymorphisms and the biochemical
pathways involved.
Thus, for combinations of specific dietgene interactions, the biological inter-
action (synergism or antagonism) of genes and diet is superimposed on background
risk. Joint effects can be quantified using theoretical models, but we still have to
learn what type of model applies most commonly to geneenvironment interactions.
It is highly questionable whether elucidation of biochemical pathways can contribute
to valid inference on additivity, multiplicativity, or other synergistic/antagonistic
effects; at the same time, high-throughput mass approaches that simultaneously
include a large number of gene variants and dietary factors in epidemiologic research
are often incapable of yielding sufficient evidence for specific genenutrient inter-
actions. Uncertainty on individual risks is a necessary consequence and will limit
the possibilities for PN advice.
treatment), such individual advice can indeed lead to direct changes in dietary intake,
but demonstrating effects on habitual diet, nutritional status, risk factors of disease,
and long-term disease risk is increasingly difficult.
In contrast to effective individual treatment, an important issue in prevention is
effective communication. Mass-media approaches to low-risk populations are known
for poor efficacy, conceptually similar to poor compliance in drug treatment. Group
approaches (e.g., at schools) to communicate dietary recommendations are considered
more effective, and the idea is that tailored dietary advice based on individual
genomics data would be even more effective. This notion is questionable, however.
As we can see in the practice of clinical dietetics, dietary advice incorporates dietary
assessment, provides options for dietary change, helps to set priorities, is practical,
and monitors changes, provides feedback on dietary intake (and exposure/health
markers). For the individual, the trade-off is between lowering a high risk at the
expense of giving up a habitual diet. For PN the healthy individual will make a
trade-off between (unsure) future risk reduction and rewards of (positive) feedback
on the one hand and direct losses of well-being related to changing habitual patterns
of food preparation, psychological aspects, taste, and social aspects of eating. Direct
changes in quality of life must be balanced against uncertain future longevity.
(See Chapter 13 and Chapter 14.)
11.5.2 GENOMICS
Research on dietgene interactions and genomics research are in their infancy. So far,
the results of dietgene studies have not been equivocal. Several methodological issues
may account for this, such as the need for large study samples, a consistent and well-
measured phenotype, high-quality information on dietary exposures, etc. There
certainly is an enormous growth potential for research, and we are beginning to see
its potency in explaining variation of nutrient needs and risks between individuals.
In this chapter, we focused on genetic data; information on human disease risks
as related to transcriptomics, proteomics, and metabolomics is not available nowa-
days. The genetic blueprint can be regarded as a somewhat static trait that tells us
about the potential of our body, but differently so for different individuals and results
are still scarce. Whole-genome scans employing thousands of SNPs will soon
become available and indicate new relevant loci, with possibly surprising findings.
This may enable the search for underlying pathways and study of the simultaneous
effects of SNP-patterns and dietary modification. However, statistical methods and
common scientific principles are still being developed. For instance, the etiological
distance between SNPs and a clinical disease is long, and dilution occurs. Shortening
this distance by reducing the phenotype from individual to organ, tissue, cellular,
or molecular levels tends to increase the strength of scientific proof on specific
interactions; at the same time, however, zooming in on molecular laboratory research
excludes higher-level interactions and increases uncertainty due to other factors,
thereby tempering the generalizability to individual subjects and populations. In
addition to SNPs, future results of transcriptomics, proteomics, and metabolomics
may provide powerful noninvasive techniques to screen populations and monitor the
effect of dietary interventions. For instance, the role of folate in DNA methylation and
other reversible epigenetic processes may be examined using these techniques on blood
9281_C11.fm Page 144 Wednesday, July 18, 2007 3:03 PM
11.5.3 TAILORING
As mentioned earlier, balancing direct gains in well-being against future risk is now-
adays restricted to clinical settings with high and intermediate risks. However, ICT
technologies can make tailored advice available to a larger proportion of the population
in a cost-effective way. A personal balanced diet, however, should not be based on
nutrient composition alone, but it should also include dietary assessment (as the starting
point to identify options for change), energy balance (physical activity), lifestyle factors
(e.g., smoking and drinking), and medical risk factors (BMI, blood lipids, blood
pressure, etc.), whereas genomic markers might be added in the distant future.
Nevertheless, PN targets the individual, and remains a high-risk-group approach, rather
than the population-based approach advocated by Sir Geoffrey Rose.
The concept of PN also calls for defining health as more than the absence of
disease, also including personal well-being (as WHO does). If this is ignored, it
may lead to adding years to life rather than life to years. In a clinical high-risk
context, risk reductions are paid for not only economically, but also personally by
accepting side effects that adversely affect well-being in a physical (surgery), mental
(pain, suffering), or social sense (hospitalization). The price individuals want to pay
for reducing moderate or low risks are logically much less, but they are taken into
account in tailored advice to individual patients. If well informed, individuals can
decide their own personal balance of long-term risks (with attached values) and
direct benefits in the domain of well-being (with their attached values). In this view,
personal is based on individual values rather than genomic risks.
In practice, it appears that some people are willing to pay large amounts of
money for genetic testing and tailored advice, even though the advice provided is
quite similar to generic guidelines; however, individual risk cannot be addressed
9281_C11.fm Page 145 Wednesday, July 18, 2007 3:03 PM
with reasonable accuracy and precision, leaving the predicted risks associated with
high uncertainty. Thus, similar to the widespread use of supplements and the increas-
ing use of functional foods, the PN approach may preferentially reach the worried,
healthy, well-educated, and health-literate population segments that already have the
highest life expectancy. This way, PN may contribute to widening the socioeconomic
health gradient in populations. This calls for careful balancing of the population-
targeted policies against the PN approach.
ACKNOWLEDGMENT
The work underlying this chapter has been funded by a grant of the Wageningen
University Board (VIVRE project MyFood, 20042005).
9281_C11.fm Page 146 Wednesday, July 18, 2007 3:03 PM
KEY READINGS
Brennan, P., Gene-environment interaction and aetiology of cancer: what does it mean and
how can we measure it?, Carcinogenesis 23(3), 3817, 2002.
Mutch, D.M., Wahli, W., and Williamson, G., Nutrigenomics and nutrigenetics: the emerging
faces of nutrition, FASEB J. 19(12), 160216, 2005.
Ordovas, J.M. and Corella, D., Nutritional genomics, Annu Rev Genomics Hum Genet, 5,
71118, 2004.
Perera, F. P., Environment and cancer: who are susceptible?, Science 278(5340), 106873, 1997.
Rose, G., Sick individuals and sick populations, Int J Epidemiol 14(1), 328, 1985.
REFERENCES
1. Mutch, D.M., Wahli, W., and Williamson, G., Nutrigenomics and nutrigenetics: the
emerging faces of nutrition, FASEB J. 19(12), 160216, 2005.
2. Report of the committee of inquiry into the future development of the public health
function and community medicine. HMSO, London, 1988.
3. The World Health Report 2002 Reducing risks, promoting healthy lifestyle WHO,
2002.
4. MacMahon, B., Gene-environment interactions in human disease, J Psychiatr Res 6,
393242, 1968.
5. WCRF, Food, nutrition and the prevention of cancer: a global perspective WCRF/
American Institute for Cancer Research, Washington, D.C., 1997.
6. Lampe, J. W. and Peterson, S., Brassica, biotransformation and cancer risk: genetic
polymorphisms alter the preventive effects of cruciferous vegetables, J Nutr 132(10),
29914, 2002.
7. Brennan, P., Gene-environment interaction and aetiology of cancer: what does it mean
and how can we measure it?, Carcinogenesis 23(3), 3817, 2002.
8. Beck-Nielsen, H., Vaag, A., Poulsen, P., and Gaster, M., Metabolic and genetic
influence on glucose metabolism in type 2 diabetic subjects experiences from
relatives and twin studies, Best Pract Res Clin Endocrinol Metab 17(3), 44567, 2003.
9. McCarthy, M. I. and Zeggini, E., Genetics of type 2 diabetes, Curr Diab Rep 6(2),
14754, 2006.
10. Florez, J. C., Hirschhorn, J., and Altshuler, D., The inherited basis of diabetes mellitus:
implications for the genetic analysis of complex traits, Annu Rev Genomics Hum
Genet 4, 25791, 2003.
11. Sreenan, S. K., Zhou, Y. P., Otani, K., Hansen, P. A., Currie, K. P., Pan, C. Y., Lee,
J. P., Ostrega, D. M., Pugh, W., Horikawa, Y., Cox, N. J., Hanis, C. L., Burant, C. F.,
Fox, A. P., Bell, G. I., and Polonsky, K. S., Calpains play a role in insulin secretion
and action, Diabetes 50(9), 201320, 2001.
12. Grant, S. F., Thorleifsson, G., Reynisdottir, I., Benediktsson, R., Manolescu, A., Sainz,
J., Helgason, A., Stefansson, H., Emilsson, V., Helgadottir, A., Styrkarsdottir, U.,
Magnusson, K. P., Walters, G. B., Palsdottir, E., Jonsdottir, T., Gudmundsdottir, T.,
Gylfason, A., Saemundsdottir, J., Wilensky, R. L., Reilly, M. P., Rader, D. J., Bagger, Y.,
Christiansen, C., Gudnason, V., Sigurdsson, G., Thorsteinsdottir, U., Gulcher, J. R.,
Kong, A., and Stefansson, K., Variant of transcription factor 7-like 2 (TCF7L2) gene
confers risk of type 2 diabetes, Nat Genet 38(3), 3203, 2006.
9281_C11.fm Page 147 Wednesday, July 18, 2007 3:03 PM
13. Uusitupa, M., Gene-diet interaction in relation to the prevention of obesity and type
2 diabetes: evidence from the Finnish Diabetes Prevention Study, Nutr Metab
Cardiovasc Dis 15(3), 22533, 2005.
14. Luan, J., Browne, P. O., Harding, A. H., Halsall, D. J., ORahilly, S., Chatterjee, V. K.,
and Wareham, N. J., Evidence for gene-nutrient interaction at the PPARgamma locus,
Diabetes 50(3), 6869, 2001.
15. Rose, G., Sick individuals and sick populations, Int J Epidemiol 14(1), 328, 1985.
16. Smith, G.D, Behind the broad street pump: aetiology, epidemiology and prevention
of cholera in mid-19th century Britain, Int J Epidemiol 31(5), 920932, 2002.
17. van der Pal-de Bruin, K.M., de Walle, H.E., de Rover, C.M., Jeeninga, W., Cornel,
M.C., de Jong-van den Berg, L.T., Buitendijk, S.E., and Paulussen, T.G.. Influence
of educational level on determinants of folic acid use, Paediatr Perinat Epidemiol
17(3), 25663, 2003.
18. Perera, F. P., Environment and cancer: who are susceptible?, Science 278(5340),
106873, 1997.
19. Tiemersma, E. W., Omer, R. E., Bunschoten, A., vant Veer, P., Kok, F. J., Idris, M. O.,
Kadaru, A. M., Fedail, S. S., and Kampman, E., Role of genetic polymorphism of
glutathione-S-transferase T1 and microsomal epoxide hydrolase in aflatoxin-associated
hepatocellular carcinoma, Cancer Epidemiol Biomarkers Prev 10(7), 78591, 2001.
9281_C11.fm Page 148 Wednesday, July 18, 2007 3:03 PM
9281_S002.fm Page 149 Wednesday, July 18, 2007 4:32 PM
Section II
Personalized Nutrition and
Stakeholders in Society
9281_S002.fm Page 150 Wednesday, July 18, 2007 4:32 PM
9281_C012.fm Page 151 Wednesday, July 18, 2007 3:06 PM
12 Imminent Applications
of Nutrigenomics:
A Stakeholder Analysis
J.T. Winkler
CONTENTS
12.1 INTRODUCTION
The promise of nutrigenomics is grand; the practicalities are daunting. Many scien-
tists researching the frontiers of the field believe the complexities are such that useful
applications are decades away. But, for better or for worse, scientists do not always
151
9281_C012.fm Page 152 Wednesday, July 18, 2007 3:06 PM
determine what happens in the world. Other groups have interests in nutrigenomics.
For them, benefits are in prospect genetic understanding and personalized diets,
better health and enhanced capacities, improved foods and targeted marketing, lower
costs, and higher revenues. Rewards such as these may inspire a more optimistic
interpretation of the scientific evidence. And provide incentives to immediate action.
This chapter identifies probable actions of key interest groups whose interplay will
shape the future of nutrigenomics. The intent is description, not prescription. The
perspective is intentionally agnostic, to describe not what is desirable, but what is
likely, for good or for ill. The method is stakeholder analysis. Five groups will be
considered: scientists, food companies, consumers, competitive athletes, and health-
care providers. Their behavior will raise issues on which policy decisions will have
to be taken, sooner or later. For all these parties, the discovery of inherited suscep-
tibilities and capabilities is a prerequisite for whatever actions they want to take.
Hence, this chapter is as much about genetic testing as it is about nutritional
interventions.
12.2 SCIENTISTS
Most people active in nutrigenomics at present are scientists, conducting research
in genomics, transcriptomics, proteomics, metabolomics, and associated disci-
plines. In this field, as everywhere else, differing views coexist even among the
cognoscenti.
Nonetheless, the view of numerous leaders is that, because of the multiple genes,
multiple polymorphisms, and multiple metabolic pathways involved, unraveling
genetic influences on diet-related diseases will take a long time. We are in the stage
of basic science and likely to remain so for many years. With the present state of
knowledge, speculation regarding practical applications of nutrigenomics would be
premature.
Thus, for example, Ben van Ommen, Coordinator of the NuGO program: I
think it is way too early to give dietary advice based on your genes.1 Similarly,
Sian Astley, U.K. coordinator of NuGO, feels there is no way that nutrigenomics
is ready yet to give individual advice or help health policy.2 Not in our lifetimes,
says optimist Bruce German.3 Never, feels Larry Parnell, because it is technically
impossible to cover all possible genegene permutations.4 Prudence is most vividly
expressed by Jose Ordovas: What we are seeing now is the very, very beginning.
Nutrigenomics has established proof of concept, but thats it. To commercialize the
science at this point is almost like throwing darts in the dark.5
Even within the scientific community, however, others urge immediate use. Most
obvious are scientists in companies selling genetic tests, such as Sciona and Genelex.
Among clinical geneticists, Aubrey Milunsky condenses the positive case into the
title of his book, Your Genetic Destiny: Know your Genes, Secure your Health, Save
your Life.6 For others, moral as well as practical arguments encourage incremental
application of nutrigenomic knowledge as soon as it emerges. Diet-related diseases
are so widespread, responsible for so much mortality, morbidity and misery, that it
would be criminally negligent to continue prescribing ineffective general diets
when personalized nutrition is possible.7
9281_C012.fm Page 153 Wednesday, July 18, 2007 3:06 PM
a health premium. Some economists will agree with them, because that pricing
strategy creates economic problems.
Some people, made aware of their nutrigenetic susceptibilities, will become
anxious about their future. In distress, some will feel they need to grasp whatever
nutritional remedies are available, whatever the cost. In the language of economics,
they become price insensitive; they have inelastic demand for those targeted
foods that are relevant to their risks. This is the exploitation of anxiety. In many
countries, commercial appeals to fear are prohibited by advertising regulations. But
genetic susceptibility may be presented more subtly, fright disguised as fate (see
Chapter 15).
Not only is individual psychology at issue, but macro health economics. Diet-
related diseases are overwhelmingly diseases of poverty. With nutritional problems,
as with much else, the poor suffer more. If targeted foods are permanently positioned
at higher prices, then the people who need them most will be least able to acquire
them. The public health promise of nutrigenomics would then not be realized (see
Chapter 11).
But a different economic calculus leads to an intermediate position. Even for
poor people, absolute price is not always the concern; sometimes they seek value
for money. Targeted foods, if genuinely effective, would not only diminish disease
risks and symptoms, but also associated costs. These include outgoings on care, but
equally important, income foregone because of hindrances to employment. There
may be net benefits, even if prices are high. Whether targeted foods really represent
value for money, however, is conditional on another fundamental issue, namely, their
efficacy. Will personalized nutrition be effective in coping with inherited, diet-related
diseases? Even the most cautious researchers are optimistic on that point, or they
would not be working in the field. But decades of nutrigenomic research may still
not suffice, because this is as much a legal as a scientific issue.
may claim that foods reduce risk factors for disease. Companies may be required
at some stage to present the scientific evidence behind their health claims, a
surrogate assessment of efficacy. The U.S. has adopted a particularly complex variant
of this approach, with four grades of qualified claims, depending on the quality
of the evidence, with even preliminary and unpersuasive science allowed at the
bottom rung.
Nutrigenomics has the potential to end this hotchpotch. When mechanisms of
action are eventually proved, it will offer new and stronger forms of evidence, beyond
normal nutritional arguments, about foods beneficial actions against disease. That
should, logically, undermine the legal distinction between foods and drugs.
However, doctors and most other health professionals receive little training in
nutrition. They are often suspicious of dietary interventions, citing the absence of
randomized controlled trials. This is part rationality, part protective mechanism
against alternative therapies. Furthermore, interests have built up to defend the
status quo, especially in pharmaceutical companies. Changing the law will be a
lengthy, perhaps interminable, process. Legal pragmatism has already resisted sci-
ence for more than a century and may continue to do so.
In the interim, possibly a long interim, nutrigenomic tests will be offered to
people with varying degrees of completeness and certainty. Targeted foods will come
wrapped in imprecise statements about what they do and how well. To paraphrase
Eliot, this is the way nutrigenomics may begin, not with a bang but a whimper.11
But food companies will strive for a more explosive beginning. They will actively
promote both tests and products. Advertising may be as persuasive in implying the
efficacy of targeted foods as it has been about many other goods. Consumers may
respond more to these promises than to the scruples of scientists and lawyers.
12.5 CONSUMERS
Compared to the corporates, consumers have a more intense as well as a more
personal interest in nutrigenomics. For some, their life depends on it. For others,
only their quality of life hangs in the balance, but that is motivation enough for
most. At present, relatively few consumers know about nutrigenomics, so they do
not yet constitute a coherent interest group. But targeted commercial promotions
to the vulnerable will spread information quickly. And with it, hope.
Sick consumers sometimes employ a different rationality from scientists, defined
by their context. In the early days of HIV/AIDS drugs, sufferers refused to wait for
the conclusion of long randomized controlled trials. Their personal end might arrive
before scientists had proof. The rigor of three-stage clinical trials was seen, not
as good scientific practice, but as delay, or worse, denial of treatment.
Seriously ill patients are increasingly demanding access to experimental, unap-
proved drugs.12 The controversy over Herceptin for breast cancer is the latest example
of consumer impatience with scientific method and regulatory due process. Sellers
of remedies often encourage, support, and even finance, such demanding consumers.
Elected politicians may respond to their appeals. They did in both the cases cited.
Diet-related diseases are usually chronic, lacking the life-and-death drama of these
examples. But the popular perception of genes-as-destiny may lend nutrigenomics
9281_C012.fm Page 158 Wednesday, July 18, 2007 3:06 PM
an urgency that nutrition has seldom acquired, outside famines. Consumers are
already more accepting of nutritional remedies than doctors and regulators. Nutrige-
nomics will reinforce their preferences.
However, both the health and commercial benefits of targeted foods depend on
consumers willingness to share their nutrigenetic data with others, not just health
professionals, but also food companies. Who will have access to genetic profiles?
Nutrigenomic testing has two distinct attributes that make it exceptional, and
probably more acceptable. First, a positive response is always possible, changing
diet. Second, that response is under the individuals control. These are intrinsic
advantages of nutrigenomic testing, compared to other types of screening. Here,
within economic constraints, knowledge empowers individual action.
But extrinsic factors will also influence the acceptability of these tests. Those
who are not just at risk, but already ill with a diet-related disease, may be more
insistent than resistant about testing. As with the aforementioned drugs, some will
be more concerned about short-term relief from problems than long-term invasions
of privacy. With nutrigenomics, as with other subjects, the views of consumer
organizations are not always those of individual consumers.
The terms and conditions on which nutrigenomic tests are presented to the public
will further shape uptake. Tests subsidized by industry would certainly be cheaper
than those available individually from specialist testing companies. And perhaps
more accessible, too, if distributed through local outlets, even unconventional ones
such as pharmacies and supermarkets. Lower prices and easier availability would
widen the customer base and help counter the allegation that nutrigenomics tests
are merely a luxury for the worried well. They would become a resource for the
demonstrably sick as well as those at risk.
Moreover, the agreement of consumers to share results with third parties, includ-
ing food companies, might be a condition for providing subsidized tests. To those
worried about privacy, such requirements will seem extortionate. But concerned
consumers might be willing to share information with those they believe, rightly or
wrongly, could help them, especially as initial tests are likely to be specific to genes
for defined problems, rather than expensive whole-genome scans that could allow
later fishing expeditions for other information.
Decisions on sharing personal data will not be taken by individuals in isolation,
but within an evolving social context. Nutrigenomics is arriving at a time when
skepticism toward conventional medicine is growing. Alternative or complementary
therapies are increasingly popular, including dietary responses such as supplements,
functional foods, and herbal remedies, as well as simply healthier eating. Nutrige-
nomics again reinforces an existing trend.
But it is already clear that nutrigenomics will be the focus of a public debate in
its own right, amplified in popular media, about whether it is endangering or empow-
ering.13 The outcome is by no means certain. In Europe, the opponents of the new
genetics in all its forms have had considerable success. Other parts of the world
are more enthusiastic.
otherwise minister to them. More important for public health, this category also
includes the much larger numbers who aspire to become leading, or at least improved,
sportsmen and women, the millions who train for marathons, as well as the handful
who win them. Hope is as vulnerable to exploitation as fear.
being used now by the Department of Trade and Industry in the U.K. as the basis
for public consultations on the ethics of science. If enhancement for sport arrives,
purchasable packages for additional traits and talents will follow. They would put
prices on the qualities people value most. They would create a market in superiority.
Ethicists will see gene transfer as raising fundamental questions about humanity.
For the more worldly, it exacerbates eternal arguments about inequality. For others,
this is just a new technical option for giving people what they have always wanted,
but were previously unable to obtain.
Whatever the merits of arguments for and against, gene doping in popular sports
would rapidly increase public familiarity with genetic testing and beneficial
responses. In the long term, gene therapies for otherwise incurable diseases may be
the more compassionate face of genetic modifications.
The point here is not to endorse any of these arguments, pro or con. It is merely
to anticipate the imminent disputes, vicious but profound, about human enhancement
in all its forms, including the nutrigenomic. Gene doping at the Olympics may fire
the starting gun.
In the market-oriented U.S., the language is different, but the goal is the same.
The strategy is to shift the incentives to people having ownership of their own
health and therefore health care, as opposed to thinking some third party pays the
bills.27 In the misty past, when our proverbs were coined, sages spoke more plainly,
Patient, heal thyself. Self-care is the generic name for this approach. On balance,
over time, it may or may not prove better for patients. But its sprouting in the 21st
century has more to do with finance than therapeutics.
KEY READINGS
Miller, P. and Wilsdon, J., Better Humans? The politics of human enhancement and life
extension, Demos, London, 2006.
Milunsky, A., Your Genetic Destiny: Know Your Genes, Secure Your Health, Save Your Life,
Perseus, Cambridge, 2001.
Sweeney, H.L., Gene doping, Scientific American, July 2004, p. 37.
9281_C012.fm Page 167 Wednesday, July 18, 2007 3:06 PM
REFERENCES
1. Cited in, Pray, L.A., Dieting for the genome generation, The Scientist, 19, 14, 2005.
2. Astley, S., Nutrigenomics, presented at Demos Symposium, You eat what you are,
London, April 20, 2005.
3. German, B., Nutritional phenotypes in the age of metabalomics, presented at NuGO
Week Symposium, Pisa, September 1013, 2005.
4. Parnell, L., The complexity of nutritional disorders presents a bioinformatic challenge
to genotyping, presented at NuGo Week Symposium, Pisa, September 1013, 2005.
5. Cited in Pray, L.A., op cit.
6. Milunsky, A., Your Genetic Destiny: Know Your Genes, Secure Your Health, Save
Your Life, Perseus, Cambridge, 2001.
7. Gibney, M., Public communication at NuGO Week Symposium, Pisa, September
1013, 2005.
8. Kutz, G., Nutrigenetic testing: tests purchased from four web sites mislead consumer,
U.S. Government Accountability Office, GAO-06-977T, Washington, D.C. 2006.
9. Heart U.K., www.heartuk.org.uk.
10. Department of Health, Dietary Reference Values for Food Energy and Nutrients for
the United Kingdom, HMSO, London 1991.
11. Eliot, T.S., The hollow men, in Collected Poems 19091962, Harcourt Brace Jovanovich,
New York, 1991.
12. Groopman, J., The right to a trial, The New Yorker, December 18, 2006, p. 40.
13. Trivedi, B., Hungry genes?, New Scientist, 193, No. 2587, 34, 2006.
14. The true state of C.S.I. justice, New York Times, January 29, 2007.
15. Connor, S., Glaxo chief: our drugs do not work on most patients, The Independent,
December 8, 2003.
16. Miller, P. and Wilsdon, J. (Eds.), Better Humans? The politics of human enhancement
and life extension, Demos, London, 2006.
17. Sokolove, M., The lab animal, New York Times Magazine, January 18, 2004, p. 28.
18. Hamilton, B., Human enhancement technologies, uncorrected evidence, House of
Commons Science and Technology Committee, October 25, 2006.
19. World Anti-Doping Agency, www.wada-ama.org.
20. Yang, N. et al., ACTN3 genotype is associated with human elite athletic performance,
Am J Hum Genet, 73, 627, 2003.
21. Sears, B., The Zone, Regan Books, New York, 1995.
22. Miah, A., Genetically Modified Athletes: Biomedical Ethics, Gene Doping and Sport,
Routledge, London, 2004.
23. Wanless, D. Securing Our Future Health: Taking a Long Term View, HM Treasury,
London, 2002.
24. Wanless, D., Securing Good Health for the Whole Population, HM Treasury, London,
2004.
25. Cayton, H., Patients as entrepreneurs: who is in charge of change?, in Andersson, E.,
Tritter, J., and Wilson, R. (Eds.), Healthy Democracy, The National Centre for
Involvement, London, 2006.
9281_C012.fm Page 168 Wednesday, July 18, 2007 3:06 PM
26. Donnelly, L., PM calls for empowerment instead of nannying, Health Serv J, 12,
August 3, 2006.
27. McKinnell, H.A., cited in Berenson, A., An insiders critique of the health care system,
New York Times, June 4, 2005.
28. Food and Drug Administration, Pharmacogenomic data submissions, final guidance,
Issue 22, March 2005.
29. Milunsky, op cit.
30. Eckholm, E., Medicaid plan prods patients towards health, New York Times, December
1, 2006.
31. Steinbrook, R., Imposing personal responsibility for health, New Eng J Med, 355,
353, 2006.
32. Bishop, G. and Brodkey, A., Personal responsibility and physician responsibility:
West Virginias Medicaid plan, New Eng J Med, 355, 356.
9281_C013.fm Page 169 Wednesday, July 18, 2007 3:07 PM
CONTENTS
13.1 INTRODUCTION
Information that is personalized to take into account a targeted individuals char-
acteristics and situation is more effective in influencing that persons health behav-
ior than general information. The central factor for effectiveness is perceived
personal relevance. In most theoretical models of behavior change, concepts relat-
ing to this personal factor are included, such as perceived personal risk, effective-
ness of recommended actions, self-efficacy, and cost-benefit evaluation. In the field
of nutrition, communication can be tailored to an individuals food-related needs,
habits, preferences, and interests shaped by his or her physical and social
169
9281_C013.fm Page 170 Wednesday, July 18, 2007 3:07 PM
circumstances. When the messages are also delivered in a location and at a time
desired by the individual, we can speak of personalized nutrition communication.
The innovative field of nutritional genomics (nutrigenomics) is expected to lead to
more insights into the interaction between diet, genes, proteins and metabolites,
and health. This will hopefully lead to evidence-based strategies for the develop-
ment of stronger health messages that will influence both the perceived severity
and effectiveness of the actions recommended in nutrition communication. Nutrige-
nomics aims to use genetic testing to assess personal vulnerability to the devel-
opment of nutrition-related illnesses. Such a test could contribute to perceived
personal vulnerability and to the perceived effectiveness of recommended actions,
thereby influencing the perceived personal relevance of healthy eating. Next to a
personalized assessment, the availability of a personalized solution by means of
a diet, product, or nutrient that helps prevent nutrition-related diseases is a prereq-
uisite for the concept of personalized nutrition to become integrated in health
behavior change strategies.
Many causes of premature death and illness are preventable, or at least
postponable, at the level of individual behavior. As individuals, if we did not
smoke, exercised more, and ate less saturated fat and more fruits and vegetables,
we would probably be healthier. In the last decades, a lot of effort has been put
into improving dietary habits through nutrition communication. However, it has
not been effective in changing the behavior of populations or individuals: in most
European countries, actual consumption is not in line with basic recommendations
for healthy nutrition. Although consumers know what they should be doing, diets
still contain too much energy, saturated fat, sugar and salt, and insufficient veg-
etables, fruits, and fish. Dietary habits are important determinants of health
because unhealthy eating, coupled with poor lifestyle choices, increases the risk
of disease such as obesity, diabetes, cardiovascular disease, and cancer. The
growing incidence of diet-related diseases accentuates the need for innovative
approaches that motivate people to eat healthily [13]. A promising approach is
personalization of nutrition communication. Reviews on health interventions [4,5]
and research on the effect of personalization [68] have shown that information
that is personalized to a targeted individuals characteristics and situation is more
effective in influencing that persons health behavior than general information.
Central to this chapter is perceived personal relevance, because personalized
nutrition communication that is not perceived as being relevant to the individual
will not induce motivation to eat healthily in the long term. We discuss how
personal relevance is integrated in communication, fear arousing communication,
and health behavior change theory. In most theoretical models, concepts relating
to this personal factor are included, such as selective perception, perceived per-
sonal risk, effectiveness of recommended actions, and self-efficacy [914]. The
innovative field of nutritional genomics is expected to lead to more insight into
the interaction between diet, genes, protein, and metabolites, and health [15]. The
possible influence of this innovation on perceived personal relevance of nutrition
communication is discussed in Section 13.3. Finally, we discuss some of the issues
surrounding the personalization of nutrition communication and topics for future
research.
9281_C013.fm Page 171 Wednesday, July 18, 2007 3:07 PM
about their personal food intake. They rate their food intake as healthy and
therefore do not consider nutrition communication on healthy eating as being
personally relevant [2124].
In most communication about nutrition, the messages are about the consequences
of unhealthy eating. The theory of planned behavior, protection motivation theory,
and health belief model all rely on severity as the influencing factor for perceived
personal relevance. Fear appeals are often made to spell out the severity of
nutrition-related diseases such as diabetes and cardiovascular diseases, but the
fact that these are outside the experience of most people may explain why the
appeal is not effective. Gleicher and Petty [26] and Liberman and Chaiken [27]
state that fear arousal can induce two different coping strategies: either acting as
a motivator to induce intensive (and accurate) message processing or inducing
defense motivation, both temporarily. Defense motivation is most likely to occur
when a health threat is both severe and personally relevant because personal
beliefs are being threatened. According to the heuristic-systematic model [27],
the processing goal of defense-motivated people is to confirm the validity of a
particular attitudinal position (I am eating healthily) and to disconfirm the validity
of others (your eating choices place you at risk). Defense-motivated people will
process information selectively in the way that best supports their own beliefs
(see also: selective perception). Risk perception research has raised questions
about the assumptions of most models in preventive health behavior, which is,
as stated earlier, that people are able to adequately assess the risk to themselves
associated with their behavior. Risk assessment is a complex process influenced
by several factors that interfere with accurate assessment of personal risk. The
catastrophic effect, controllability, reversibility, and whether the risk is taken on
a voluntary basis or not influence risk perception and, thereby, fear arousal. For
instance, perceived risks of unhealthy lifestyles (voluntary) are known to be lower
than perceived risks of new technology (nonvoluntary) [28]. Thus, people tend
to have misperceptions about their personal behavior, depending on the context
in which the risk information is presented, the way the risk is being described,
and their personal and cultural characteristics [11]. Furthermore, estimation of
personal risks tends to be biased. Many people overestimate small probabilities
9281_C013.fm Page 173 Wednesday, July 18, 2007 3:07 PM
(plane crashes) and underestimate large probabilities (heart disease). Risks that
are cognitively available through personal experience or intense media coverage
tend to be overestimated. This bias process is related to Tversky and Kahnemans
[29] availability heuristic and refers to peoples tendency to judge an event as
more probable to the extent that it is more easily pictured or recalled. The lack
of knowledge about the specific relationship between food intake and individual
risk of disease may interfere with the perceived severity of nutrition communi-
cation and contribute to misperceptions of personal risk.
In protection motivation theory and the health belief model, perceived vulnera-
bility or susceptibility refers to the subjective risk of acquiring an illness if no
countermeasures are taken [30]. In combination with high perceived severity,
perceived vulnerability is known to build fear and increase the personal relevance
of messages. Research shows that people are quite aware of the relative risk of
specific activities or behavior, but this tends to change when personal risk needs
to be assessed. For instance, smokers accept the association between smoking
cigarettes and disease but do not believe themselves to be personally at risk [31].
This is referred to as unrealistic optimism from Weinsteins [32] paper that
focused on comparative risks in health risk perception. Van der Pligt [11]
describes six causes of unrealistic optimism that can lead to perceived personal
invulnerability, perceived control, egocentric bias, personal experience, stereo-
typical or prototypical judgment, self-esteem maintenance, and coping strategies.
Risks judged to be under personal control tend to induce feelings of optimism
[33]. People generally know more about their own protective behavior than about
that of others, causing an egocentric bias that can generate optimism. They also
tend to focus on their own risk-reducing behavior and are less aware of personal
behavior that increases risk. Personal experience with a risk tends to be relatively
vivid and can decrease unrealistic optimism. Stereotypical or prototypical judg-
ment is a relatively extreme image people have of high-risk groups, which is
unlikely to fit with their self-image, thereby increasing optimism. Generally,
people tend to rate their own actions, lifestyle, and personality as better than
that of others: this is known as self-esteem maintenance or enhancement. The
last factor that influences unrealistic optimism relates to coping strategies. Con-
ditions of high stress or threat can induce denial, thereby reducing emotional
distress but also reducing the likelihood of preventive actions or their success.
In general nutrition communication, vulnerability is addressed without reference
to the personal factor. Recent research on the stage model of processing of fear-
arousing communications developed by Das et al. [34] concluded that unless
individuals can be persuaded of their vulnerability to health risk, they are unlikely
to take protective action [35]. Through face-to-face consultation, vulnerability
to nutrition-related disease can be made more personally relevant, based on
assessment of the individuals lifestyle (e.g., calories), physical parameters
(e.g., blood pressure), and environmental circumstances (e.g., sedentary work).
As in relation to communication on severity, uncertainties about whether
9281_C013.fm Page 174 Wednesday, July 18, 2007 3:07 PM
In Banduras [14] social learning theory (later called cognitive learning theory),
the central concept relating to personal factors is self-efficacy. It describes a
cognitive state of taking control in which people believe they are capable of
carrying out the specific behavior and can help create and control their environment
in doing so. This concept of reciprocal determinism of behavior and environment
is associated with concepts of self-management and self-control and is influenced
by several processes, such as direct experience. It is also influenced by the storing
and processing of complex information in cognitive operations that facilitate
anticipation of the consequences of actions, represents goals, and weighs evidence
from different sources to assess personal capabilities. This leads to a situation-
specific self-appraisal that induces feelings of confidence or insecurity about
behavior in new, unpredictable, or stressful situations. Self-efficacy is, then, the
perception of ones own capacity to successfully organize and implement new
9281_C013.fm Page 175 Wednesday, July 18, 2007 3:07 PM
behavior largely based on experience with similar actions and situations encountered
or observed in the past, also called performance history [38]. Self-efficacy is also
influenced by indirect or vicarious learning experience gained by observing others
(modeling), such as a parent, teacher, or television personality who seems to enjoy
a specific behavior. It is assumed that people learn more from models that are
competent, attractive, likable, admired, and loved. Also, similarity to/empathy with
the observer is known to influence learning [30]. Another influence on self-efficacy
stemming from others is verbal persuasion. Strong persuasive messages from a
respected, trusted person, such as a dietician, can have a positive influence on
feelings of self-efficacy. Besides influencing behavior, self-efficacy affects thought
patterns and emotional reactions, thereby inducing or reducing feelings of anxiety
or coping ability. It is linked to specific skills and, by personalizing communica-
tion, attention can be paid to an individuals feelings of self-efficacy. Communi-
cation about what to eat can, for instance, be matched with an individuals level
of cooking skills. Perceived behavioral control in the theory of planned behavior
is closely related to self-efficacy and refers to the fact that people can have positive
attitudes toward certain behavior but simply lack the resources to carry it out. In
protection motivation theory, the coping appraisal will be positively influenced
by response efficacy, the equivalent of effectiveness of recommended action and
self-efficacy.
Stage 1: Unaware of
Awareness
Stage 2: Unengaged
Perceived personal
vulnerability
Stage 3: Deciding
about acting
Perceived severity
Perceived eectiveness of
the recommended behavior
Cost-benet evaluation
Stage 6: Acting
Stage 7: Maintenance
FIGURE 13.1 Personal factors in different stages of behavior change. (From Weinstein, N.D.,
Health Psychol., 7, 355, 1988. With permission.)
TABLE 13.1
Personal Factors Influencing Perceived Personal
Relevance
Concepts Reference
TABLE 13.2
Factors Relating to Personal Relevance and Interfering Concepts in Stages
of Behavior Change
Awareness Threat Appraisal Coping Appraisal
TABLE 13.3
Concepts Relating to Personal Factors in Discussed Theories
Concept Related to Personal
Theories Factors Reference
new science of nutrigenomics examines the response of our genes, proteins, and
metabolism to different foods. Nutrigenomics is expected to lead to evidence-based
dietary intervention strategies for maintaining, and perhaps restoring, health and fitness
and preventing diet-related disease. It is expected that, in the long term, nutrigenomic
technologies will be used to determine how our body responds to foods that affect our
long-term health [15]. Nutrigenomics is also targeting the assessment of personal
vulnerability to the development of nutrition-related illnesses through genetic testing.
Next to a personalized assessment, the availability of a personalized solution by
means of a diet, product, or nutrient that helps to prevent nutrition-related diseases is
a prerequisite for the concept of personalized nutrition to become integrated in health
behavior change strategies. The assumption is that individuals will be able to use this
information to reduce their risk of common diseases such as heart disease, diabetes,
and obesity or to improve overall health and well-being. But not much is known yet
about how individuals will actually use the information and whether it will contribute
more to behavior change than the information currently supplied [4043].
The most promising contribution to behavior change may lie in the reduction of
uncertainties on a general and personal level, thereby reducing the influence of the
interfering concept of probabilistic outcomes. The expected insights into the relation-
ship between genes, nutrition, and health may provide a stronger base for designing
clearer health messages about severity and effectiveness of the recommended actions.
On a personal level, advice based on genetic testing can provide insight into individual
vulnerability to nutrition-related illnesses and into the effectiveness of preventive
strategies, thereby strengthening messages targeted at perceived personal vulnerability
and perceived efficacy of recommended actions. Also, beliefs about the effectiveness
of a treatment recommendation based on genotypic information could be strengthened.
From research, it is known that tests offering great certainty of results (clinical validity),
with available treatment and prevention options, are more readily undertaken [44].
Another potentially positive influence on perceived personal relevance is the avoidance
of optimistic bias that leads to feelings of invulnerability. Uncertainties as to whether
or not an individual is at risk of developing nutrition-related disease can be influenced
by the results of genetic testing.
However, information on individual genetic makeup can also have undesired
effects on motivation to change behavior. It is known that, when fear appeals become
too strong, some people will react defensively; this leads to inaction. Also, higher
susceptibility to developing nutrition-related disease can induce feelings of fatalism,
thus decreasing motivation to change. Given the common perception that genetic
risks are immutable, motivation to change behavior may be decreased by weakening
beliefs that changing behavior will reduce risk. Perceived self-efficacy could also
be negatively influenced by weakening the belief in the ability to change behavior:
Its in my genes so I cant change it.
Further research has to be undertaken to gain more insight into how people will
include information on genetic makeup in the process of behavior change and
whether it will either enhance or decrease motivation. In Table 13.4, an overview is
presented of the possible contribution of innovations to perceived personal relevance
of nutrition communication in respect of creating awareness, the threat appraisal,
and the coping appraisal.
9281_C013.fm Page 179 Wednesday, July 18, 2007 3:07 PM
TABLE 13.4
Possible Contribution of Information on NutritionGenesHealth to
Perceived Personal Relevance of Nutrition Communication in Stages
of the Behavior Process
Awareness Threat Appraisal Coping Appraisal
I have the gene, so what can I do? I have the gene, so I eat healthily
FIGURE 13.2 What will be the effect of information on genes and health?
and education offices, for services, information, and social support; and industry,
for products. Views on how nutrigenomics-based personalized nutrition communi-
cation will impact individuals and society need to be exchanged among all actors
concerned to ensure its legitimate and successful introduction. As with other new
technologies, personalized nutrition will entail benefits and risks and may change
social structures, culture, norms, and values. These will best be addressed by the
people that will be confronted with personalized nutrition in their daily life. Early
involvement in the development and implementation process of innovations influ-
ences personal commitment to those innovations. The WHO also recently stated that
capacity building through partnerships is an important strategy to promote health.
Partnerships are important for bringing together diversity in expertise, skills, and
resources for more effective health outcomes [46]. However, partnerships can only
be successful if participants share visions about goals, leadership, and the necessary
investment of each participant. Most often, this does not reflect reality.
A first step toward an open dialogue to create partnerships on personalized nutrition
was taken at the round table discussion at the conference of the European Nutrige-
nomics Organisation in November 2005. The views of representatives from different
scientific disciplines, industry, and government were collected about who should be
involved in a dialogue and what topics should be on the agenda. Although the discus-
sion was very lively, it was clear that its content remained scattered, leaving many
topics touched upon unexplored in depth. The reactions of the participants were limited
to their own specific interest, and the discussion did not elaborate further on specific
9281_C013.fm Page 181 Wednesday, July 18, 2007 3:07 PM
topics. Further action is needed to facilitate extensive and fruitful dialogue about
relevant topic such as dissemination of knowledge, practical relevance of scientific
insights, and social-ethical issues such as expected high costs of applications.
KEY READINGS
Frosch, D.L., Mello, P., and Lerman C., Behavioral consequences of testing for obesity risk,
Cancer Epidemiol. Biomarkers. Prev., 14(6), 1485.
Khoury, M.J., Genetics and genomics in practice: the continuum from genetic disease to
genetic information in health and disease, Genet. Med., 5, 261.
Lerman, C. et. al., Genetic testing: psychosocial aspects and implications, J. Consult. Clin.
Psychol., 70(3), 784, 2002.
WHO (2005) The Bangkok Charter for Health Promotion in a Globalized World. Available
at http://www.who.int/healthpromotion.
REFERENCES
1. Department of Health, Choosing Health: making healthy choices easier, Public Health
White Paper; Command paper CM 6374, U.K., 2004.
2. Ministry of Health, Welfare and Sports, Living longer in good health also a
question of a healthy lifestyle. Dutch Ministry of Health, Welfare and Sports, The
Hague, 2004.
3. World Health Organization, The World Health Report 2002: reducing risks, promoting
healthy life, World Health Organization, Geneva, 2002.
4. Contento, I.R. et al., The effectiveness of nutrition education and implications for
nutrition education policy, programs and research: a review of research, J. Nutr. Educ.,
27, 277, 1995.
5. Contento, I.R., Randell, J.S., and Basch, C.E., Review and analysis of evaluation
measures used in nutrition education intervention research, J. Nutr. Educ. Behav., 34,
2, 2002.
6. Curry, S.J. et. al., A Randomized trial of self-help materials, personalized feedback,
and telephone counselling with non-volunteer smokers, J. Consult. Clin. Psychol.,
63(6), 1005, 2005.
7. Brug, J. et al., The impact of computer-tailored feedback on fat, fruit and vegetable
intake, Health Educ. Behav., 25, 517, 1998.
8. Kreuter, M.W. and Stretcher, V.J., Do tailored behaviour change messages enhance
the effectiveness of health risk appraisals? Results from a randomized trial, Health
Educ. Res., 11, 97, 1996.
9. Sears, D.O. and Freedman J.L., Selective exposure to information: a critical review,
in The Process and Effects of Mass Communication, Schramm, W. and Roberts, D.F.,
Eds., University of Illinois Press, Urbana, 1971, p. 209.
10. Petty, E.P. and Cacioppo, J.T., Communication and Persuasion: Central and Periph-
eral Routes to Attitude Change, Springer-Verlag, New York, 1986.
11. Van der Pligt, J., Risk perception and self-protective behaviour, Eur. Psychol., 1(1),
34, 1996.
12. Rogers, R.W., Cognitive and psychological processes in fear appeals and attitude
change: a revised theory of protection motivation, in Social Psychology: A Source
Book, Cacioppo, J.T. and Petty, R.E., Eds., Guilford Press, New York, 1983, p. 153.
9281_C013.fm Page 182 Wednesday, July 18, 2007 3:07 PM
13. Janz, N. and Becker, M.H., The health belief model: a decade later, Health Educ. Q.,
11, 1, 1984.
14. Bandura, A., Self-efficacy mechanism in human agency, Am. Psychol., 37,122, 1982.
15. Mller, M. and Kersten, S., Nutrigenomics: goals and strategies, Nat. Rev., 4, 315, 2003.
16. Weinstein, N.D., The precaution adoption process, Health Psychol., 7, 355, 1988.
17. Batra, R. and Ray, M.L., Advertising situations: the implications of differential
involvement and accompanying affect responses, in Information Processing Research
in Advertising, Harris, R.J., Ed., Lawrence Earlbaum Associates Publishers, London,
1983, p. 127.
18. McGuire, W.J., Attitudes and attitude change, in Handbook of Social Psychology,
Lindzey, G. and Aronson, E., Eds., Random House, New York, 1985, p. 233.
19. Sherif, C.W., Sherif, M., and Nebergall, R.E., Attitude and Attitude Change: The
Social Judgement-Involvement Approach. W.B. Saunders, Philadelphia, PA, 1965.
20. Festinger, L., A Theory of Cognitive Dissonance. Stanford University Press, Stanford,
CA,1957.
21. Brug, J., Hospers, H., and Kok, G.J., Differences in psychosocial factors and fat con-
sumption between stages of change for fat reduction, Psychol. Health, 12, 719, 1997.
22. Brug, J. et al., Self-rated dietary fat intake: association with objective assessment of
fat, psychosocial factors and intention to change, J. Nutr. Educ., 26, 218, 1994.
23. Glanz, K., Brug, J., and Van Assema, P., Are awareness of dietary fat intake and
actual fat consumption associated? A Dutch-American comparison, Eur. J. Clin. Nutr.,
51, 542, 1997.
24. Lechner, L., Brug, J., and De Vries, H., Misconceptions of fruit and vegetable
consumption: differences between objective and subjective estimation of intake, J.
Nutr. Educ., 29, 313, 1997.
25. Ajzen, I. and Madden, T.J., Prediction of goal directed behaviour: attitudes, intentions
and perceived behavioural control, J. Exp. Soc. Psychol., 22, 453, 1986.
26. Gleicher, F. and Petty, R.E., Expectations of reassurance influence the nature of fear-
stimulated attitude change, J. Exp. Soc. Psychol., 28, 86, 1992.
27. Liberman, A. and Chaiken, S., Defensive processing of personally relevant health
messages, Pers. Soc. Psychol. Bull., 18, 669, 1992.
28. Koelen, M.A. and Lyklema, S., (in Dutch) De consument en perceptie van voed-
selveiligheid, in Ons Eten Gemeten. van Kreijl, C.F. and Knaap, A.G.A.C., Eds.,
Bohn, Stafleu, Van Loghum, RIVM, Bilthoven, the Netherlands, 2004, chap. 8.
29. Tversky, A. and Kahneman, D., Judgement under uncertainty: heuristics and biases,
Science, 185, 1127, 1974.
30. Koelen, M.A. and Van den Ban, A., Health Education and Health Promotion,
Wageningen Academic Publishers, Wageningen, the Netherlands, 2004.
31. Pechacek, T. and Danaher, B., How and why people quit smoking: a cognitive-
behavioural analyses, in Cognitive Behavioural Intervention: Theory, Research and
Procedures, Kendall, P. and Hollon, S., Eds., Academic Press, New York, 1979, p. 389.
32. Weinstein, N.D., Unrealistic optimism about future life events, J. Pers. Soc. Psychol.,
39, 806, 1980.
33. Otten, W. and Van der Pligt, J., Risk and behaviour: the mediating role of risk
appraisal, Acta Psychol., 80, 325, 1992.
34. Das, E.H.H.J., de Wit, J.B.F., and Stroebe, W., Fear appeals motivate acceptance of
action recommendations: evidence for a positive bias in the processing of persuasive
messages, PSPB, 29, 650, 2003.
35. De Hoog, N., Stroebe, W., and de Wit, J.B.F., The impact of fear appeals on processing
and acceptance of action recommendations, PSPB, 31(1), 24, 2005.
9281_C013.fm Page 183 Wednesday, July 18, 2007 3:07 PM
36. Zimbardo, P.G. and Leippe, M.R., The Psychology of Attitude Change and Social
Influence, McGraw-Hill, Boston, MA, 1991.
37. Sutton, S.R., Fear arousing communications: a critical examination of theory and
research, in Social Psychology and Behavioural Medicine, Eiser, J.R., Ed., John Wiley
& Sons, Chichester, 1982, chap. 13.
38. Green, L.W. and Kreuter, M.W., Health Program Planning: An Educational and
Ecological Approach, 4th ed., McGraw-Hill, New York, 2005.
39. Darnton-Hill, I.I., Margetts, B., and Deckelbaum, R., Public health nutrition and
genetics: implications for nutrition policy and promotion, Proc. Nutr. Soc., 63, 173,
2004.
40. Marteau, T.M. and Lerman C., Genetic risk and behavioural change, BMJ, 322, 1056,
2001.
41. Haga, S.B., Khoury, M.J., and Burke, W., Genomic profiling to promote a healthy
lifestyle: not ready for prime time yet, Commentary, Nat. Genet., 34(4), 347, 2003.
42. McCain, M. and Schmid, G., From Nutrigenomics Science to Personalized Nutrition:
the market in 2010. Institute for the Future, Silicon Valley, California, 2003.
43. Massoud, M. et al., The Future of Nutrition: Consumers Engage with Science, Institute
for the Future, Silicon Valley, California, 2001.
44. Marteau, T.M. and Croyle, R.T., The new genetics: psychological responses to genetic
testing, BMJ, 316, 693, 1998.
45. Lerman, C. et. al., Genetic testing: psychosocial aspects and implications, J. Consult.
Clin. Psychol., 70(3), 784, 2002.
46. World Health Organisation, The Bangkok Charter for Health Promotion in a Globalized
World, Available at http://www.who.int/healthpromotion, 2005, accessed June 2006.
9281_C013.fm Page 184 Wednesday, July 18, 2007 3:07 PM
9281_C014.fm Page 185 Thursday, July 26, 2007 7:10 PM
CONTENTS
14.1 INTRODUCTION
Adjusting marketing offerings to the identified needs and wants of consumers is at
the heart of the marketing philosophy (e.g., Kotler and Keller, 2006). Essentially,
marketing aims to achieve an exchange of values for the mutual benefit of both the
customer and the supplier. What the customer receives in terms of need satisfaction
from products or services and gives in terms of monetary and nonmonetary sacrifices
to obtain the product is the mirror image of what the supply chain provides in terms
of products and services, and receives in terms of money to satisfy its financial
objectives of the value exchange. This process benefits both parties: the customer
benefits from superior need satisfaction through consumption, whereas the suppliers
benefits are financial means for growth and business continuity. When there are
many customers and multiple suppliers, the processes of segmenting, targeting, and
positioning become crucial. In other words, companies attempt to understand
185
9281_C014.fm Page 186 Thursday, July 26, 2007 7:10 PM
field of personalized nutrition. The Institute of the Future defines personalized nutrition
as the application by individuals of their knowledge of nutrigenomics to their everyday
decisions about nutrition (Cain and Schmid, 2003: 2). However, we believe that
personalization goes beyond the field of genetics and includes other information related
to personal preferences as well, such as perceived state of health, tastes, values, and
other relevant segmentation variables.
Personalization in the field of human nutrition provides great potential to all
stakeholders, including the consumer, the firm, and those concerned with public health.
The process of personalization provides consumers with the opportunity to find an
offer that is better tailored to their specific needs. This might be in the form of
nutritional information, personal advice, and even customized food products. In this
way, personalization may also help consumers to reduce their search and evaluation
costs in product choice behavior. For the firm, personalization may help to distinguish
it from competitors, build customer relationships, and increase customer loyalty. If
well executed and substantiated in nutritional science, personalized nutrition may
contribute to the improvement of public health. However, there are benefits and costs
associated with personalization both to the individual consumer and to the company.
In this chapter we will address personalization from a consumer and marketing
perspective. First, we will delineate the definition of personalization and distinguish
it from customization. Then, we will review process models of the personalization
process and use these models to identify and discuss the costs and benefits involved
in personalization. Next, we will focus on consumer evaluation of personalization,
before we turn to the operational dimensions of personalized nutrition. Lastly, we
will discuss critical success factors from a marketing perspective, turn to conclusions,
and look at the outlook for personalized nutrition.
TABLE 14.1
Definitions of Personalization
Reference Definition
Peppers and Rogers One-to-one marketing based on the idea of an enterprise knowing its
(www.1to1.com) customer. Through interactions with a customer, the enterprise can learn
how a customer wants to be treated and can then treat that customer
differently from other customers.
Hagen (1999) The ability to provide content and services tailored to individuals based
on knowledge about their preferences and behavior.
Personalization consortium The use of technology and consumer information to tailor electronic
(2005) commerce interactions between a business and each individual customer.
Using information either previously obtained or provided in real time
about the customer, the exchange between the parties is altered to fit
that customers stated needs, as well as the need perceived by the
business based on the available customer information.
Dyche (2002) The capacity to customize customer communication based on knowledge
preferences and behaviors at the time of interaction with the customer.
Peppers and Rogers (1997) The process of using a customers information to deliver a targeted solution
to that consumer is known as personalization or one-to-one marketing.
Riecken (2000) Personalization is about building customer loyalty by building a
meaningful one-to-one relationship; by understanding the needs of each
individual and helping satisfy a goal that efficiently and knowledgeably
addresses each individuals needs in a given context.
Hanson (2000) A specialized form of product differentiation in which a solution is
tailored to a specific individual.
Peppers, Rogers, and Dorf Customizing some feature of a product or service so that the customer
(1999) enjoys more convenience, lower cost, and some other benefit.
Allen, et al. (2001) Company-driven individualization of customer Web experience.
Imhoff, Loftis, and Geiger Personalization is the ability of a company to recognize and treat its
(2001) customers as individuals through personal messaging, targeted banner
ads, special offers on bills, or other personal transactions.
Wind and Rangaswamy Personalization can be initiated by the customer (i.e., customizing the
(2001) look and contents of a Web site) or by the firm (i.e., individualized
offering, greeting customer by name).
Coner (2003) Personalization is performed by the company and is based on a match of
categorized content to profiled users.
Roberts (2003) The process of preparing and individualizing communication for a
specific person based on stated or implied preferences.
Riemer and Totz (2003) To match one objects nature to one subjects needs. More precisely: to
customize products, services, content, communication, etc., to the needs
of single consumers or customer groups.
Bonnett (2001) Personalization involves a process of gathering user information during
interaction with the user, which is then used to provide appropriate
assistance or services, tailor-made to the users needs.
Source: Adapted from Vesanen, J. and Raulas, M., Journal of Interactive Marketing, 20(1), 520, 2006
and Adomavicius, G. and Tuzhilin, A., Communications of the ACM, 48(10), 8390, 2005.
9281_C014.fm Page 189 Thursday, July 26, 2007 7:10 PM
Personalization thus requires knowledge about the consumer and his or her specific
needs, and adjusting offerings to fit those needs. Some offerings, such as services
(which are by nature often developed in interaction with the consumer) and infor-
mation can be adjusted to consumer needs relatively easily. However, adjusting
physical products to individual consumer needs entails substantially increasing the
complexity of the supply chain, with potential additional costs as a consequence.
This is the field of customization.
Sometimes referred to as product personalization (Riemer and Totz, 2003),
mass customization is the field of research that focuses on using flexible manufac-
turing processes to customize physical products to the needs and preferences of
consumers with mass production (or near mass production) efficiency (Piller and
Mller, 2004). Personalization and mass customization are different but closely
related terms. Wind and Rangaswamy (2001) differentiate between the two terms,
arguing that personalization is the process located on the consumer side of the
marketing spectrum, whereas mass customization is the process on the operational
product side. Much of the mass customization literature originates from the literature
on flexible manufacturing. In customization, consumers can alter or even create
products that contain precisely those attributes that the individual consumer specifies
(Godek, 2002). In other words, the marketing offerings are being adjusted by, or
for, the user very close to the moment of purchase or consumption. An example
would be companies such as Dell and Gateway, which have adopted flexible and
responsive manufacturing systems that create products to meet the needs of individ-
ual consumers upon their request (Pine, 1993). In contrast, personalization does not
presuppose on-the-spot personalized production. Usually based on directly elicited
or indirectly inferred consumer preferences, the firm recommends those products
from an existing range that provides the best fit with those preferences (Godek,
Yates, and Yoon, 2002). An example of personalization would be Amazon.com,
which recommends alternatives on the basis of consumer-expressed preferences
(customers who bought this item also bought ). In conclusion, customization
goes one step beyond personalization, in that the customer is an active codesigner
of the product (Bonnett, 2001). Mass customization in the context of nutrition
involves a large number of complex supply chain issues that are beyond the scope
of this chapter. We will only discuss its important implications for the marketing of
personalized nutrition (see Section 14.7).
Personalization implies an interaction between the customer and the supply chain.
Simonson (2005) discusses this interactive process and emphasizes that customer
9281_C014.fm Page 190 Thursday, July 26, 2007 7:10 PM
satisfaction is not only enhanced through delivering products with superior fit to the
customers individual preferences, but also that through self-assessment, the interactive
process itself can help shape the customer preferences, particularly when customers
do not yet have stable preferences. In this way, personalization may be a means to
offer customers what they want, often even before they know that they want it (i.e.,
latent needs). Others (Fotheringham and Owen, 2000) have argued that although
interaction is important, personalization is not restricted to synchronous interaction.
Personalization may be based on previously collected consumer information. Also,
personalization issues are dominant in but not restricted to online interactions
(Murthi and Sarkar, 2004) but can be realized at all user interfaces (Riecken, 2000).
TABLE 14.2
Several Proposed Process Models for Personalization
Adomavicius and Murthi and Sarkar Vesanen and
Tuzhilin (2005) (2003) Pierrakos et al. (2003) Raulas (2006)
Source: Summarized in Vesanen, J. and Raulas, M., Journal of Interactive Marketing, 20(1), 520, 2006.
With permission.
9281_C014.fm Page 191 Thursday, July 26, 2007 7:10 PM
(Steckel et al., 2005; Piller, 2005) have argued for more research on consumer
evaluation, as successful personalization depends on an enduring learning relation-
ship between the customer and the firm. Hence, the existing process models as included
in Table 14.2 should be conceived of as cycles rather than linear processes, with their
success depending on the ability to generate repeated interaction with the customer.
Process models by Murthi and Sarkar (2003) and Adomavivius and Tuzhilin
(2005) separate the personalization process into three stages: (1) understanding and
learning about customers preferences, (2) delivering personalized offerings to cus-
tomers, and (3) evaluating the learning and matching process. Pierrakos et al. (2003)
describe the personalization process as having four basic features, namely: (1) the
two-way nature of the communication system, (2) the level of response control that
each party has in the communication process, (3) the personalization in the com-
munication process, and (4) the use and involvement of database technology. How-
ever, the specific tasks they distinguish in the personalization process again relate
to the understanding (data collection), personalization (data preprocessing, pattern
discovery, knowledge postprocessing), and evaluation (reports) substages.
Vesanen and Raulas (2006) synthesize these process models by distinguishing
between four objects (customer, customer data, customer profile, and marketing
output) and four operations (interactions, processing of information, customization,
and delivery) as the key variables that together define the marketing process with
individual consumers.
The Vesanen and Raulas (2006) model defines the personalization process as a
loop, with the customer as the starting point. Through interactions with a customer,
relevant data are collected, such as expressed preferences (e.g., through question-
naires), Web site/purchasing behavior, or other more objective measurements such
as blood parameters and genetic typing. This customer information is processed
into relevant customer profiles that serve as a basis for the differentiation and
segmentation of customers. Sophisticated techniques such as data mining and neural
networks are increasingly being used for this purpose. The customer profiles are
used to generate personalized marketing output, such as tailored communication
material, personal advice, or personalized products. The delivery stage describes
how (e.g., through which channel) the personalized marketing offerings reach the
customer and will bring about a response from the customer as a new interaction,
resulting in new customer data. This closes the loop in the Vesanen and Raulas
(2006) model, as this new customer information will be processed into customer
profiles as a new input into the customization process.
At a more general level, the proposed stages can be classified in terms of three
responsibility domains: (1) consumer, (2) consumerfirm interaction, and (3) firm.
The consumer domain refers to those stages that involve consumer actions, whereas
the consumerfirm interaction domain comprises the processes by which the supply
chain and the consumer interact through various interfaces. Finally, the firm domain
includes the value-creation process undertaken by the supply chain on the basis of
the consumers personal information.
Ronteltap et al. (2006) have developed a process model specifically for the area
of personalized nutrition, which defines specific stages in each of the three respon-
sibility domains within the personalization process: the consumer, the interaction,
9281_C014.fm Page 192 Thursday, July 26, 2007 7:10 PM
7. Usage 3. Data
handling
6. Delivery
8. Evaluation 4. Design
2. Communication
1. Information 5. Production
sharing
and the firm. The model (see Figure 14.1) describes the personalization process in
eight different stages (see also Steckel et al., 2005; Wendel et al., 2006). During the
first stage of the exchange process, consumers make available certain personal, and
possibly sensitive, information (e.g., current health condition) to the supply chain
(stage one: information sharing). In the next stage, this information passes through
a physical interface (e.g., service desk), a digital interface (e.g., electronic question-
naires), or a combination (stage two: communication). Stage three of the exchange
process (data handling) pertains to the receiver of the information (i.e., the supply
chain) that will transform the personal information into a personalized solution on
the basis of a decision model (stage four: design). Subsequently, the supply chain
will create or select personalized recommendations (information or lifestyle or
product advice) that address the needs of a particular client (stage five: production).
In stage six (delivery), this personalized advice, involving different types of infor-
mation possibly combined with products or services, is communicated and distrib-
uted via a user interface (e.g., e-mail) and received by the consumer. After acting
upon the recommendation (stage seven: usage), the consumer may evaluate the
added value of the recommendations and assess any personal benefit obtained from
the interaction (stage eight: evaluation). This evaluation will then serve as input to
the next cycle and the decision about whether or not to repeat the interaction.
By emphasizing the three domains, the model illustrates the joint responsibility
of the consumer and the firm in establishing the personalized interaction. Whether
such interactions occur depends largely on the perceived costs and benefits from the
personalization process, which we will review next. In Section 14.5 and Section 14.6
we will specifically focus on the consumer evaluation of personalized nutrition.
decision to engage in the personalized advice process (on nutrition or another matter)
is basically the result of a simple equation: if the (expected) returns exceed the
(expected) costs, the likelihood that customers will engage in this option will increase.
The decision of suppliers to provide personalized advice follows the same logic: only
if the expected returns exceed the expected cost will the firm offer personalized advice.
The costs to the company are the costs of collecting personalized consumer information
and differentiation (e.g., delivering personalized advice and the costs of resources
required to do so). The returns are the price premium that can be charged for individ-
ualized options, the relationship and customer loyalty that individualization may build,
and the enhancement of corporate or brand image. The key question is how to define
the costs and returns at the consumer level. Although the firms costs and benefits can
be expressed in monetary units, the consumer value is more complex and involves
psychological and ethical factors, too (Karat et al., 2003). At the consumer level, costs
and benefits are usually (e.g., Piller and Mller, 2004; Simonson, 2005; Karat et al.,
2003) differentiated into those emerging from the outcomes of the personalization
process and those emerging from the interactive process itself.
However, the interactive process also involves perceived costs on the part of
the consumer. These costs primarily constitute the cost of disclosing personal
information (Karat et al., 2003); here, privacy issues become pertinent. This is
particularly problematic in the case of genetic information, as the information is
hard-wired and stable over time, may be relevant to many different parties (e.g.,
insurance companies), and it is uncertain what applications the information may
yield in the future (Chadwick, 2004). As a result, consumers may be reluctant to
accede to their genetic information being stored, as in future it may be used against
them (Chadwick, 2004; Economist, 2005). As a psychological cost, consumers
may experience a lack of freedom and may feel that marketers are trespassing on
their personal preferences; or they may interpret personalization as attempts to
persuade and manipulate (Simonson, 2005). Also, the active involvement in the
codesign task may result in the consumer feeling psychologically at a disadvantage,
because the manufacturer possesses much more information. The consumer has
few options available, other than trusting that the manufacturer will propose the
best personalized option.
TABLE 14.3
Factors and Factor Levels Applied
in the Scenario-Rating Tasks
1. Information sharing
a. Blood composition
b. DNA/genetic makeup
c. Food consumption habits
2. Communication
a. Through fitness club
b. Through hospital
c. Through family practitioner
3. Data handling
a. Fully anonymous
b. Patient and family practitioner
c. Available to commercial food company
4. Design
a. Commercial company
b. Insurance company
c. Government nutrition center
5. Production
a. At ingredient level
b. Food product groups
c. Special branded products
6. Delivery
a. Through e-mail
b. Through fitness club
c. Through family practitioner
7. Usage
a. Within existing meal patterns
b. Add special products to diet
c. Prepare own adjusted meals
8. Evaluation
a. No feedback for verification
b. Feedback for verification optional
c. Feedback for verification mandatory
9281_C014.fm Page 197 Thursday, July 26, 2007 7:10 PM
As an illustration of the conjoint task results, in Box 14.1 we show the two scenarios
from the Ronteltap and Van Trijp (2006) study that consumers thought were the
most (italics) and least (between brackets) desirable scenarios for personalized
nutritional advice.
Overall, the results as presented in Box 14.1 show that there are still a number
of hurdles to be overcome before personalized nutrition can be applied commercially
in marketing. In the next section, we will discuss the critical success factors from
a marketing point of view.
14.8 CONCLUSION
Personalized nutrition based on insights from nutrigenomics is a relatively new
concept. Building on marketing literature about personalization and effective market
segmentation, in this chapter we have reviewed the marketing potential of person-
alized nutrition. Although personalization has revolutionized the marketing approach
in many fields (see Wind, 2001; Wind and Rangaswamy, 2001 and Riemer and Totz,
2003 for more details), we have specifically focused on the potential of personalization
in the food domain. We have argued that personalized nutrition based on nutrige-
netics may provide potential for personalized nutritional advice with the purpose of
assisting consumers to make better choices from the existing product supply. In
terms of personalized food products, we have argued that currently there is limited
marketing potential for the mainstream food industry, because the consumer seg-
ments are likely to be small and the additional costs involved in production, distri-
bution, and marketing are large. However, niche markets of sufficient size and
purchasing power might develop that can profitably be catered to by smaller and
specialized food companies. Another potentially successful application of nutrige-
nomics knowledge in the short term may be foods personalized on the basis of the
absence of certain ingredients and nutrients.
KEY READINGS
Adomavicius, G. and Tuzhilin, A., Personalisation technologies: a process-oriented perspec-
tive, Communications of the ACM, 48(10), 8390, 2005.
Murthi, B.P.S. and Sarkar, S., The role of the management sciences in research on personal-
ization. Management Science, 49(10), 13441362, 2003.
Piller, F.T., Mass customization: reflections on the state of the concept. The International
Journal of Flexible Manufacturing Systems 16: 313334, 2005.
Vesanen, J. and Raulas, M., Building bridges for personalization: a process model for mar-
keting. Journal of Interactive Marketing, 20(1), 520, 2006.
Wind, J. and Rangaswamy, A., Customerization: the next revolution in mass customization.
Journal of Interactive Marketing, 15(1), 1332, 2001.
REFERENCES
Adomavicius, G. and Tuzhilin, A., Personalisation technologies: a process-oriented perspec-
tive, Communications of the ACM, 48(10), 8390, 2005.
Bonnett, M., Personalization of Web services: opportunities and challenges. Ariadne 28 (June),
www.ariadne.ac.uk, 2001.
Bouwman, L.I., Hiddink, G.J., Koelen, M.A., Korthals, M., Vant Veer, P., and Van Woerkum,
D., Personalized nutrition communication through ICT application: how to overcome
the gap between potential effectiveness and reality. European Journal of Clinical
Nutrition 59(Suppl. 1), S108S116, 2005.
Brug, J., Campbell, M., and Van Assema, P., The application and impact of computer-generated
personalized nutrition education: a review of the literature. Patient Education and
Counseling 36, 145156, 1999.
Brug, J., Oenema, A., and Campbell, M., Past, present and future of computer-tailored nutrition
education. American Journal of Clinical Nutrition 77(Suppl.), 1028S1034S, 2003.
Cain, M. and Schmid, G. From nutrigenomic science to personalize nutrition. The market in
2010. Palo Alto (CA): Institute for the Future. New Consumer New Genetics Program,
report SR793 (www.ifth.org), 2003.
Chadwick, R., Nutrigenomics, individualism and public health. Proceedings of the Nutritional
Society 6, 161166, 2004.
Csikszentmilhalyi, M., Flow: The Psychology of Optimal Experience. New York: Harper and
Row, 1990.
9281_C014.fm Page 203 Thursday, July 26, 2007 7:10 PM
Rogers, E.M., Diffusion of innovations, 5th ed. New York: Free Press, 2003.
Ronteltap, A. and Van Trijp, H.C.M. (2007). Consumer preferences for personalised nutritional
advice: a conjoint analysis study. Wageningen University; Marketing and Consumer
Behaviour Group. Working Paper (in preparation).
Ronteltap, A., Wendel, B.S., Van Trijp, H.C.M., and Dellaert, B.G.C., Marketing van producten
die passen bij de genetische structuur van het individu [Marketing of products that
fit the genetic predisposition of the individual]. Jaarboek van de Nederlandse Verenig-
ing van Marketing Onderzoekers, 2006, pp. 127141.
Simonson, I., Determinants of customers responses to customized offers: conceptual frame-
work and research propositions. Journal of Marketing 69(January), 3245, 2005.
Sheth, J., Sisodia, R., and Sharma, A., The antecedents and consequences of customer-centric
marketing, Journal of the Academy of Marketing Sciences, 28(1), 5566, 2000.
Steckel, J.H., Winer, R.S., Bucklin, R., Delleart, B.G.C., Dreze, X., Haubl, G., Jap, S.D.,
Little, J.D.C., Meyvis, T., Montgomery, A.L., and Rangaswamy, A., Choice in inter-
active environments. Marketing Letters 16(3/4), 309320, 2005.
Tornatzky, L.G. and Klein, K.J., Innovation characteristics and innovation-adoption imple-
mentation: a meta-analysis of findings. IEEE Transactions on Engineering Manage-
ment 29(1), 2845, 1982.
Treacy, M. and Wiersema, F. (1993), Customer intimacy and other value disciplines. Harvard
Business Review 71(1), 8493.
Venkatesh, V., Morris, M.G., Davis, G.B., and Davis, F.D., User acceptance of information
technology: toward a unified view. MIS Quarterly 27(3), 425478, 2003.
Vesanen, J. and Raulas, M., Building bridges for personalization: a process model for market-
ing. Journal of Interactive Marketing, 20(1), 520, 2006.
Wendel, S., Ronteltap, A., Dellaert, B.G.C., and Van Trijp, H.C.M., CustomerFirm Interaction
in the creation and delivery of personalized recommendations. Working paper, 2006.
Wind, J., The challenge of customerization in financial services. Communications of the ACM,
44(6), 3944, 2001.
Wind, J. and Rangaswamy, A., Customerization: the next revolution in mass customization.
Journal of Interactive Marketing, 15(1), 1332, 2001.
9281_C015.fm Page 205 Wednesday, July 18, 2007 3:11 PM
CONTENTS
15.1 Methods........................................................................................................205
15.2 Results ..........................................................................................................206
15.2.1 Genomics: General Attitudes and Perceptions ................................206
15.2.1.1 Awareness and Interest Regarding Genomics..................206
15.3 The Opportunity for Personalized Nutrition ...............................................209
15.3.1 Key Findings on Consumers Perceptions
Regarding Health and Nutrition ......................................................209
15.3.1.1 Interest in and Favorability toward
Personalized Nutrition ......................................................209
15.3.1.2 Applications for Personalized Nutrition ............................210
15.4 Discussion ....................................................................................................212
15.4.1 Awareness, Interest, and Barriers Regarding Genomics .................212
15.4.2 Awareness, Interest, and Applications
for Personalized Nutrition................................................................215
15.5 Conclusion and Principles for Effective Communication
Related to Personalized Nutrition................................................................216
15.6 Future Perspectives ......................................................................................217
Key Readings.........................................................................................................218
References..............................................................................................................219
15.1 METHODS
Since 2004, the Cogent Syndicated Genomics Attitudes and Trends (CGAT) survey
has been annually taking the publics pulse regarding the issue of genomics. CGAT
explores public awareness, understanding, favorability, and interest in genomics and
its applications, including nutrigenomics and pharmacogenomics. This chapter
205
9281_C015.fm Page 206 Wednesday, July 18, 2007 3:11 PM
describes data from the CGAT survey, collected in January 2006, via a Web-based
205-question survey of more than 1000 Americans. Using both balancing and
weighting techniques, the sample was designed to reflect the U.S. population in
terms of key demographics, including education, age, income, and gender.
Data in this chapter are also from the International Food Information Council
(IFIC) survey, Consumer Attitudes toward Functional Foods/Foods for Health. This
survey measures consumer awareness of, and interest in, functional foods/foods for
health as well as the application of genomics to nutrition and personal health, a science
otherwise known as nutrigenomics or personalized nutrition. This trending survey
was first commissioned in 1998, with subsequent surveys conducted in 2000 and 2002;
some survey questions gauge trends in consumer attitudes toward nutrition and health.
Questions regarding consumer perceptions of personalized nutrition were added in
2005. The quantitative Web-based survey was fielded in May 2005 with more than
1000 randomly selected U.S. adults, aged 18 years and older. The findings were
weighted by education, age, and ethnicity, reflecting the 2003 U.S. population census
estimate, to allow the findings to be representative of the American public as a whole.
The findings from both surveys rely primarily on univariate analyses and cross-
tabulations. The data were analyzed according to the demographic profile of the
population, including primary characteristics (e.g., age, income and gender), health-
based characteristics (e.g., health history, and current health status), and in some
cases, attitudinal characteristics (e.g., receptivity to medical advances).
15.2 RESULTS
15.2.1 GENOMICS: GENERAL ATTITUDES AND PERCEPTIONS
15.2.1.1 Awareness and Interest Regarding Genomics
Although three fourths (76%) of the American public say they have heard or read
something about genomics (defined as using individual genetic information to under-
stand and optimize health), a closer analysis indicates that this broad awareness is
quite limited in depth. First, only a very small number (6%) of Americans say they
have heard a lot about genomics, whereas half (49%) say they have heard only
a little bit. Further, when those who say they have heard something about genomics
were asked to describe what they have heard, about one fourth (27%) were unable
to recall any details, and another one third (36%) mention having heard only about
the basic connection between genes and health (Chart 15.1).
Still, there is a small group of Americans who understand the concept of genomics
and some of its practical implications. Specifically, of the approximately 75% of
Americans who have heard something about genomics, only about 15% say they
have heard that genes can be altered for health benefits and about 5% have heard
that genomics can aid in early detection of disease or that genomics can be used to
help customize health-related treatments.
Although knowledge is limited, more than half (52%) say they favor the idea
of using individual genetic information to understand and optimize health. Similarly,
nearly half (46%) of Americans say they are interested in using their personal genetic
information to understand and optimize their health. This includes using their personal
9281_C015.fm Page 207 Wednesday, July 18, 2007 3:11 PM
A lot
6%
A fair Nothing
amount 24%
21%
A little bit
49%
CHART 15.2 Moral reservations about genetic testing. n = 1015 (2006), 1018 (2005).
9281_C015.fm Page 209 Wednesday, July 18, 2007 3:11 PM
statements noted in Chart 15.2. Perhaps speaking to both moral and privacy concerns,
we see that almost half (46%) of all Americans agree with the statement, I am torn
on the use of genetics. The potential benefits are incredible, but the potential for
misuse is considerable.
Emotional uncertainty. The third challenge facing genomics is the emotional
burden that genetic test results can bring. For example, about one fourth (26%) of
the public agree that knowing their genetic profile is too great a responsibility
because it impacts [them], [their] spouse, and ultimately [their] children. Similarly,
one third (32%) agree that it would be too depressing to know [they were] going
to get a disease, particularly if there is nothing [they] can do about it. This finding
reinforces the consumer appeal of genomics when it addresses treatable diseases
and actionable preemptive choices.
More Americans have heard or read about using individual genetic information in
the context of nutrition and diet-related recommendations (42%) than in any other
context, including prescription drugs (36%), over-the-counter medications (29%),
9281_C015.fm Page 210 Wednesday, July 18, 2007 3:11 PM
and beauty and cosmetic products (19%). In fact, nearly half (47%) of all Americans
say they have heard or read something about personalized nutrition (defined as using
genetic information to make informed dietary choices).
When asked what they thought about various terms to describe the practice of
using genetic information to develop nutrition and diet-related recommendations,
Americans overwhelmingly preferred either personalized nutrition (70% like a
lot or a little) or individualized nutrition (68% like a lot or a little) over
terms such as nutrigenomics (19% like a lot or a little).
As with genomics in general, slightly more than half (51%) of the American
public hold a highly favorable view of personalized nutrition. Among the 48% of
Americans who hold an unfavorable opinion of personalized nutrition, most indicate
that their favorability is low due to a lack of knowledge about the science. Whereas
20% cite lack of knowledge directly, most do so indirectly, saying they are not sure
how they could benefit from personalized nutrition (17%) or dont see a need for
the science (14%). Others say they fail to see how genomics would be superior to
their doctors opinion or to good-old common sense. A small number (14%) say
they are not favorable at this point in time because the unknown consequences of
personalized nutrition may be harmful, whereas others have more immediate con-
cerns such as the perceived high cost of this technology (4%).
Interestingly, given Americans fears of discrimination when it comes to genom-
ics in general, only about 10% say their lack of favorability toward personalized
nutrition is attributable to concerns regarding misuse of genetic testing information.
When the topic turns away from general favorability toward using this technology
for a personal benefit, just less than half (42%) of the American public express a
strong level of interest in using genetic information to receive personalized, diet-related
recommendations. Only one fourth (27%) of the American public characterizes their
interest in personalized nutrition as low.
0 10 20 30 40 50 60
The power of the physician as guide and gatekeeper. Nearly 90% of Americans
say they would consult their doctor when deciding to pursue a genomics test to
facilitate personalized nutrition and diet-related recommendations, and almost a third
(31%) of all Americans would only pursue testing after receiving a doctors specific
recommendation.
The role of the doctor goes beyond deciding whether or not to have a test. Once
Americans have decided to take a genetic test for the purpose of receiving person-
alized nutrition recommendations, 67% cite the doctors office as the most desirable
location for administering such a test. The majority of Americans (70%) are also
most likely to indicate that they would want to receive their test results in a face-
to-face consultation similar to how many patients receive their medical test
information today.
The power of the individual and other sources in decision making. Although
the doctors opinion is undeniably important, more than half (57%) of Americans
say they will make their own decision after consulting their doctor, and an addi-
tional 10% say they will not consult their doctor at all. This means that a majority
of Americans have some measure of confidence in their own ability to decide whether
or not to pursue genetic testing.
Although secondary to physicians (70%), significant numbers of consumers rated
nurses and physician assistants (58%), and dietitians (57%) as highly credible
sources for information on using genetic information for personalized nutrition
information. Genetic counselors (48%) were also considered among the highly
credible sources. The inclusion of health associations as a credible source (52%)
demonstrates that consumers may not necessarily need a face-to-face, intimate
history for a high level of trust. Pharmacists (43%) were considered only slightly
less credible than genetic counselors. Moderately credible resources also included
university or other health newsletters (42%) and government agencies (37%). Other
sources, ranging from news media to product labels and insurers, received relatively
low ratings when it came to being a credible source on personalized nutrition (see
Chart 15.4 for the remaining figures).
Testing locations and results delivery. This research has shown the doctors office
as Americans most desirable testing location. However, a sizeable number of Americans
9281_C015.fm Page 212 Wednesday, July 18, 2007 3:11 PM
MDs
Nurses/PAs
Dietitians
Health associations
Genetic counselors
Pharmacists
University or other
health newsletters
Government agencies
Food product labels
Dietary supplement labels
Consumer groups, mass media,
Web, insurance cos.
0 10 20 30 40 50 60 70 80
say they would be interested in testing at home (40%), perhaps because of the privacy
it provides. In fact, at-home testing slightly edged out the offices of two other medical
professionals genetic counselors (39%) and dietitians/nutritionists (31%). Testing
locations that are largely unacceptable to consumers include health spas, fitness
centers, personal trainers office, and health food stores (each with 8%).
Furthermore, although face-to-face delivery of test results tops the list (70%),
not far behind are paper results (60%), which suggest that, given the right approach,
home testing could be widely accepted. Americans were less interested in their
willingness to receive their results through electronic media 35% were willing
to receive results via a secured Web site, 32% by e-mail, and 28% by disk. A phone
call with a consultant was the least preferred means of receiving test results (26%).
15.4 DISCUSSION
15.4.1 AWARENESS, INTEREST, AND BARRIERS REGARDING
GENOMICS
Research results show that consumers ideal scenario for genomics-related applica-
tions is one in which the genetic marker (or test) provides a high level of certainty
that one would actually experience a specific disease for which there are known
remedies. Given these high expectations, additional consumer education will likely
need to address the many factors that can impact disease risk, including genetics as
well as the environment (see Figure 15.1).
Barriers to adoption. Despite broad awareness, a positive perception, and general
interest in genomics and its applications, there are some formidable challenges facing
genomics. If left unchecked, the challenges have the potential of limiting widespread
usage of genomics, or even becoming the flashpoint for a negative shift in public
9281_C015.fm Page 213 Wednesday, July 18, 2007 3:11 PM
Reduce Risk of
Specific Diseases
Ensure
Condition Can Be
Treated
Identify Diseases
Certain to
Experience
FIGURE 15.1 Consumers are most interested in certain knowledge of treatable diseases.
opinion. The research showed there are three barriers of specific concern for genomic
testing. These concerns include: privacy, moral issues, and perceived emotional
drawbacks.
Almost two thirds of the American public were concerned about issues related
to privacy, which included insurance, government, and employer discrimination.
Given this finding, it is likely that a regulatory and statutory framework that takes
these concerns into account will be debated before widespread adoption of genomics
testing occurs. In the U.S., governing bodies are already beginning to look at genetic
testing and some of the challenges to come. In the scientific and academic commu-
nity, the Institute of Medicine held its first-ever meeting on nutrigenomics, sponsored
by the National Institutes of Healths Office of Dietary Supplements, the National
Cancer Institute, and the U.S. Department of Agricultures Agriculture Research
Service, which looked at the role of various stakeholders and their impact on this
emerging area of science.1 Additionally, in the U.S., the Department of Health and
Human Services has formed the Secretarys Advisory Committee on Genetics,
Health, and Society (SACGHS) to provide a forum for experts to discuss ethical,
social, legal, and medical issues related to innovations and applications in genetic
technology. The SACGHS also helps inform other government agencies about spe-
cific questions related to genetics and makes recommendations to the secretary on
such issues.2 For more than a decade, members of Congress have also been working
to pass the Genetic Information Nondiscrimination Act (GINA) to address the
growing concern surrounding the misuse of genetic information in insurance and
employment decisions.
Around the globe similar discussions have also begun. In Canada, Genome
Canada commissioned a project to compare different ethical approaches to public
involvement in governing genomics.3 The U.K. also has a commission in place that
9281_C015.fm Page 214 Wednesday, July 18, 2007 3:11 PM
provides the government with advice about genomics. This commission has a par-
ticular focus on ethical, legal, and social issues related to human genetics.4
Genomics is entering a crucial phase in which scientific advances are blossom-
ing, while simultaneously a sizable minority of the public is struggling to understand
how these advances relate to their deeply held convictions. Given that about one
third of the American public have not yet developed a firm moral stand on issues
related to genetic testing, these fence sitters may be highly influenced by messaging
regarding genomics either pro or con. If a substantial number of these individuals
are persuaded and join the concerned minority, the total group holding moral objections
would form a near majority. In contrast, messaging that promotes the responsible use
of genetic information as well as the implications for public (and personal) health may
swing the balance more heavily toward positive viewpoints (see Figure 15.2).
Emotional concerns held by Americans may, in part, signal recognition of the
significance or even value of genomics. However, there is also evidence that
the emotional burden for some may be too great, enough to preclude interest for
a sizable minority. Providers of genomic testing and products may be able to pursue
strategies to lessen this burden, especially through professionals who provide testing
coupled with consultation.
More research has been conducted to assess consumer acceptance and reactions
to genetic testing than in the area of personalized nutrition. One area, which this
research did not assess, was the behavioral consequences that could be associated
with receiving a genetic test. One study, involving healthy individuals, asked par-
ticipants to imagine that they were at increased risk for obesity based on the results
of a genetic test. This preliminary study found that a positive test (i.e., showing that
they were at increased risk of becoming obese) was a motivator for more healthful
st
tere
& In
ility
orab
Fav
Moral
Issues
Emotional
Consequences
Privacy Concerns
dietary behavior.5 This finding calls attention to the importance of providing moti-
vational or behavioral information along with a genetic test to help consumers
implement dietary or lifestyle changes.
It is not surprising, then, that for many, the doctors office would be a prelim-
inary step to pursuing any kind of genetic-based dietary approach. Thus, although
efforts to communicate the general benefits and practical knowledge of personal-
ized nutrition to the consumer are critical, so too is providing the latest develop-
ments and research in the field of genomics and its nutrition applications to the
primary care physicians whose advice and knowledge would be sought by most
individuals. This might, in part, help to alleviate the emotional burden that a lay
reading of the genetic test results might produce. Such an unfamiliar medical test
could potentially suggest a multitude of recommendations and changes. Creating
an interdisciplinary partnership among health professionals could help mitigate
the responsibility of one health professional being responsible for administering
a genetic test, analyzing the results, explaining the results to a client, making
recommendations on how to implement behavioral changes based on the results
of the test, and following up with the client to determine how his or her life has
been affected by the test results.
Considering the high levels of credibility and influence assigned to health-care
professionals beside physicians, face-to-face result consultations may well provide
an acceptable level of trust, particularly if performed by a dietitian/nutritionist or a
pharmacist. In studies completed with consumers and genetic testing, it appears
there are a multitude of factors that may influence an individuals decision to undergo
genetic testing. Research has shown a decrease in genetic testing for women at risk
of developing breast cancer after they spoke with a genetic counselor. However, in
situations where a genetic test was offered without genetic counseling, people were
more likely to undergo the genetic testing.7 Partnerships and information sharing
will be necessary to ensure that all those who work with genetic tests are providing
the most accurate information to consumers and are equipped to answer any ques-
tions or concerns related to the genetic test being offered.
As public knowledge of and familiarity with the field grows, so too might con-
sumers willingness to test and use the information in the privacy of ones own home.
One option could be to deliver paper results with an official seal of a trusted health
association. However, business case studies have already shown that even with the
best intentions of maximizing the benefits and minimizing the risks of a new science
and technology, not all innovations will prove socially acceptable. Ideally, science,
ethics, and the law working in concert could lay the foundation needed to ensure
optimal social outcomes and public acceptance of this new technology.8
about which genes are most important to our health and which may be impacted by
nutrition. It is important to continue to communicate what we know about the
promise of personalized nutrition, while at the same time communicating the limi-
tations of that knowledge. Reports of scientific advances in this field should be
accompanied by enough context to communicate the pieces of the puzzle that are
still missing. Although the science continues to emerge, effective communication
can help us move further down that road and avoid the obstacles that misinformation
can create.
There are both challenges and opportunities in communicating the potential
health benefits of foods and food components and how they ultimately may be
associated with the practice of personalized nutrition. Overall, awareness of
genomics is broad and interest is high, especially in regard to definitive tests for
treatable medical conditions. At the same time, few adults have sufficient knowl-
edge regarding genomics, leaving many unaware of the potential benefits to per-
sonal health or worse, vulnerable to misinformation. Moreover, the American
public has a number of latent concerns that could quiet enthusiasm or even turn
the tide against the entire field of genomics. According to the CGAT survey
conducted by Cogent Research as well as the IFIC survey, Consumer Attitudes
toward Functional Foods/Foods for Health, much of the American public is inter-
ested in using their personal genetic information to optimize their overall health
through personalized nutrition. Communications about personalized nutrition
should focus on balancing consumers expectations with the reality of the science
by not overpromising on potential outcomes. As applications of personalized
nutrition become more widespread, it will be important to communicate both
consumer benefits as well as address concerns. To build support, stakeholders,
such as government, industry, health professionals, and others, need to work
together to communicate effectively with each other and the public to increase
knowledge of the benefits associated with personalized nutrition.
Consumers are primed for personalized messages about foods that provide benefits
beyond basic nutrition and how to incorporate these foods into their diet. Because of
this research finding and the fact that most Americans feel hopeful and curious about
emerging science, a door is open for communicators to deliver more information that
increases the understanding of personalized nutrition and helps consumers enjoy
health-promoting foods as part of an overall healthful lifestyle. Keeping the consumer
in mind, and adherence to the following communication principles, will be vital to
consumer acceptance and success of personalized nutrition.
a checklist to help enhance the publics understanding of emerging science and the
role it plays in overall health.9 Communicators, ranging from health professionals,
educators, scientists, scientific journal editors, government officials, and journalists,
should consider these points when translating how the latest research about food and
nutrition, including personalized nutrition, could change what is on the publics plate:
One of the best ways to improve the publics understanding of personalized nutrition
is to put partnerships in place that provide clear and consistent communication
messages. Multiple communicators conveying the same messages increase the like-
lihood that those messages will resonate with consumers. It is particularly important
that these partnerships be created while the science is still developing and less
information has been imparted to consumers. These partnerships can help to ensure
that the first information consumers receive, related to the applications and benefits
of personalized nutrition, is the most accurate information.
KEY READINGS
Borra, S., Kelly, L., Tuttle, M., and Neville, K. Developing actionable dietary guidance
messages: dietary fat as a case study. J Am Diet Assoc. 2001; 101: 678684.
International Food Information Council. 2005 Functional Foods/Foods for Health Executive
Summary. July 2006. http://www.ific.org/research/funcfoodsres05.cfm. Accessed on
January 22, 2007.
International Food Information Council Foundation. Tools for Effective Communication:
Beginning a New Conversation with Consumers. http://www.ific.org/tools/intro.cfm.
Accessed on January 22, 2007.
Kaput, J. et al. The case for strategic international alliances to harness nutritional genomics
for public and personal health. Br J Nutr. 2005; 94: 623632.
Sanderson, S.C., Wardle, J., Jarvis, M., and Humphries, S. Public interest in genetic testing
for susceptibility to heart disease and cancer: a population-based survey in the U.K.
Prev Med. 2004; 39: 458464.
U.S. Department of Health and Human Services, National Institutes of Health, National
Cancer Institute, Making Health Communication Programs Work, Bethesda, MD,
2001.
9281_C015.fm Page 219 Wednesday, July 18, 2007 3:11 PM
REFERENCES
1. Institute of Medicine. Nutrigenomics and Beyond: Informing the Future. Nutrige-
nomics Workshop Presentations July 12, 2006. www.iom.edu/CMS/3788/31286/
33150/35223.aspx. Accessed on December 18, 2006.
2. Department of Health and Human Services. Secretarys Advisory Committee on
Genetics, Health, and Society. http://www4.od.nih.gov/oba/sacghs/sacghsdocuments.
html. Accessed on December 28, 2006.
3. Burgess, M. Starting on the right foot: public consultation to inform issue definition
in genome policy. Electronic Working Paper Series. W. Maurice Young Centre for
Applied Ethics, University of British Columbia. 2003.
4. Human Genetics Commission, Human Genetics Commission Fifth Report from April
2005 to March 2006. www.hgc.gov.uk/UploadDocs/DocPub/Document/Final%20PDF.
pdf. Accessed on December 19, 2006.
5. Dominick, L., Frosch, P.M., and Caryn, L. Behavioral consequences of testing for
obesity risk. Cancer Epidemiol Biomarkers Prev. 2005; 14(6): 14851489.
6. Debusk, R., Fogarty, C.P., Ordovas, J.M., and Kornman, K.S. Nutritional genomics
in practice: where do we begin? J Am Diet Assoc. 2005; 105: 589598.
7. Bowen, D.J., Battuello, K.M., and Raats, M. Marketing genetic tests. Health Educ
Behav. 2005; 32(5): 676685.
8. Kaput, J. and Rodriquez, R. Nutrients and norms: ethical issues in nutritional genomics
in Nutrition Genomics Discovering the Path to Personalized Nutrition. Wiley, 2006,
pp. 419434.
9. International Food Information Council Foundation, Institute of Food Technologists.
Guidelines for communicating the emerging science of dietary components for health.
2005. www.ific.org/nutrition/functional/guidelines/guidelinesfulldoc.cfm. Accessed
on December 19, 2006.
9281_C015.fm Page 220 Wednesday, July 18, 2007 3:11 PM
9281_C016.fm Page 221 Thursday, July 26, 2007 7:12 PM
16 Ethics of Personalized
Nutrition
Michiel Korthals
CONTENTS
16.1 INTRODUCTION
Personalized nutrition focuses on the value of health in choosing food, and prejudges
an individualizing policy instead of a public health nutrition policy; it is faced with
many uncertainties and is difficult to reconcile with research outcomes. Personalized
nutrition raises many issues of ethical concern. The three main issues are mostly
approached from a utilitarianist or rights-based perspective of individual autonomy
(informed consent or informed choice) that aims at protecting individuals against
harmful interventions. However, it is also fruitful to develop a more collective and
proactive ethical perspective, because of the collective consequences of individual
choices. The first ethical issue is that of the relationship between food and health
(drugs), which is at present subject to rather strict regulation. Owing to the rise of
nutrigenomics, new gray zones emerge, for instance, when food becomes a preven-
tive drug or when drugs are used by nonpatients, as in the case of enhancement
medicine. There are no compelling arguments against new developments, but we
are in need of debate about coping with the ethical impacts. The second issue is that
of the relations between personalized and public health nutrition, which requires
221
9281_C016.fm Page 222 Thursday, July 26, 2007 7:12 PM
oriented toward personalized nutrition. The first one emphasizes the protection of
consumer/citizens by viewing all ethical issues from the point of view of informed
consent or informed choice, i.e., from the point of view of individual freedom and
autonomy, which seeks to limit social powers that threaten to transgress the bound-
aries of privacy and of private freedom. Although issues of justice, i.e., the socially
fair distribution of resources plays a role here, the idea is primarily that social powers
should not be allowed to dominate individual consumers. Patients and consumers
should be enabled to protect themselves against powers that invade their personal
responsibility, by giving them rights [8]. In the alternative model, the emphasis is
on coordination and interaction between consumers, producers, and other stakeholders,
which means that individual consent and choice can play a role, but in the social
context of interaction and social learning, and of distributing responsibilities according
to issues and needs. Consultations and deliberations with stakeholders (as a matter
of fact, including consumers) to pinpoint agreement and disagreement play an
important role here. The aim is not to protect patients and consumers against
professional powers; patients and consumers are invited and encouraged to take part
in consultations and are given the opportunity to tell their narratives, interests, and
anxieties.
In both models, the concept of autonomy plays a pivotal role; however, in the
first as individual autonomy and, in the second, as social or collective autonomy.
The first concept of individual autonomy (and protection) is more connected to
utilitarianism, which emphasizes individual utility and happiness as the most important
criterion for the outcome of ethical decisions. The concept of personal autonomy lies
at the heart of the utilitarian doctrine of, for example, John Stuart Mill (18061873)[9]:
The only freedom which deserves the name is that of pursuing our own good in our
own way, so long as we do not attempt to deprive others of theirs, or impede their
efforts to obtain it. Each is the proper guardian of his own health, whether bodily, or
mental or spiritual [10].
As long as one can do what one thinks is in favor of ones own health and as long
as one does not harm others, then actions are ethically seen allowed. Harm toward
others is the category that marks the distinction between personal autonomy and
social life, and harm is the indicator that something is ethically wrong. This means
that individuals first and foremost should be protected from the harmful actions of
others or should be enabled to protect themselves from those harmful actions.
Of the two alternative models, the second one, of social autonomy, is directly
linked to the German philosopher Immanuel Kant (17241804) [11], the foremost
thinker of autonomy. Kant thinks more positively than Mills about the relation
between individual and society, in terms of enrichment and humanization and not
in terms of harm. Kant argues:
Laziness and cowardice are the reasons why such a large part of humanity, even long
after nature has liberated it from foreign control (naturaliter maiorennes), is still happy
to remain infantile during its entire life, making it so easy for others to act as its keeper.
It is so easy to be infantile. If I have a book that is wisdom for me, a therapist or
preacher who serves as my conscience, a doctor who prescribes my diet, then I do not
9281_C016.fm Page 225 Thursday, July 26, 2007 7:12 PM
need to worry about these myself. I do not need to think, as long as I am willing to
pay [11] (my italics).
food and health (see the case of food in curing diseases), in general, the distinction
between the two up to now has been rather strict. The most important distinctions
between food and health (drugs) are located in the dimensions of personal lives,
culture, politics, organizations, and market forces. First, food is for everyone, drugs
only for the ill; food should not have too severe negative effects, because of its
lifelong intake, and drugs sometimes have (acceptable) negative effects because of
their temporary intake; food choices involve multiple reasons, whereas drugs are
chosen for only one reason (to be cured); foods are freely available for all, and drugs
only on prescription; and finally, food is not tested for efficacy, but only for safety,
whereas drugs are tested for both efficacy and safety. When food is indeed becoming
a drug in a preventive way, it is possible for persons to get side effects caused by
their food choice; they are expected to choose their food only for health reasons;
they should consult their physicians before eating and have to learn new types of
behavior that redistribute responsibilities, duties, and rights in a fundamental way.
People will have to deal with the blurring of these important cultural, political,
personal, social, and scientific distinctions and their ethical implications [14,15]
(Table 16.1).
People have a complex set of considerations governing their food choice, which
are different from the considerations that determine their use of drugs. At least four
factors determine our food preferences, in general: genetic factors, social and cultural
factors, bodily history, and personal historical factors [17]. The genetic factors,
depending on genetic makeup, are generally not changeable; they determine taste
in general (like bitter and sweet) and, partially, taste development. The social and
cultural factors consist of determining organizational structures and values, such as
family relationships and health or nationalistic values. The bodily history influences
tongue and mouth, but also nose and ear, which are part and parcel of food choices.
When one is tasting food, one is also tasting ones own body. Finally, there are
TABLE 16.1
Some Differences between Food and Drugs
Food Drugs
personal historical factors, such as the family history and how it is internalized.
Moreover, when actually having a meal, additional factors do play a role as well,
such as the present conditions of the meal, the present condition of the body, and
the actual pleasure and displeasure in tasting. Gustatory taste and food choice are
strongly intentional and cognitive: it matters what one eats and to know what one
eats. They comprise the visual, the smell (olfactory), tongue senses (gustatory), and
the auditory (e.g., crunchy or not). The fact that taste is cognitive implies that taste
can be learned, and can develop in a certain direction. A yellowish substance with
a rotting smell turns suddenly into a delicious cheese upon tasting it, preferable with
nice and knowledgeable people.
More generally, in the domains of food and health culture, politics, science, and
society, the most important distinctions between food and drugs are that food is for
everyone, drugs only for the diseased; that food should not have negative effects,
because of its lifelong intake, and drugs sometimes have (acceptable) negative effects
because of their temporary intake; that food choices are motivated by multiple
reasons (see earlier text) and drugs by only one (to be cured or protected); that food
is freely available and drugs only on prescription available, and that food is not
tested for efficacy, but only for safety.
However, from an ethical point of view, objections against the identification of
food and health (drugs) can be heard; for example, food should not be seen as a
drug and society is not a hospital, meaning that health should not be the all-
determining value in food choices. For instance, Crawford [18] warns of the medi-
calization of daily life, which leads to a narrowing of perspective. As an observer
notices: For some decades the doctorpatient relationship has been the central
concern of medical ethics. This focus has marginalized public health issues by
concentrating on individual patients and individual practitioners, and thereby on one
aspect of medical structures in the richer parts of the world [19]. The British Food
Ethics Council views the exclusive orientation on health in food choices as a trans-
formation of society into a hospital [20], which is in the case of prevention of obesity
not desirable and not necessary as well, because physical activity is a good recom-
mendation as well [21]. Petrini [22], the founder of Slowfood, and a fierce defender
of the social and cultural aspects of enjoying food, quotes Madame Sevigny approv-
ingly, Health is enjoying the other enjoyments. When the other enjoyments are
taken away, we live longer, but we lose our health.
However, because of the development of the food sciences, traditional truths
regarding healthy food collapse: the health value of wine, chocolate, vegetable oils,
and milk are reevaluated. Food intake should correspond with changes in lifestyles:
What to eat in the new situation of sedentary work and other changing social
contexts?
The exact boundaries between food and health (drugs) are unclear, and there
have always some blurred boundaries (the ancient Greek thinker Hippocrates said:
Let food be your medicine![23]. Moreover, we are facing now two new gray areas:
food as drugs and drugs as enhancements, and we cannot eliminate them. This means
that the relationship between food and drugs needs to be reconsidered: the refusal
to reconsider the traditional distinction between food and drugs looks like sheer
dogmatism. Even with Kant [11] in hand, one could argue thus: There is a difference
9281_C016.fm Page 228 Thursday, July 26, 2007 7:12 PM
TABLE 16.2
New Gray Zones between Food and Drugs
Gray Zone: Food as
Drugs and Drugs for
Nonpatients (modafinil,
Ritalin, enhancement
Food Drugs drugs)
between obeying a physician and taking into account the advice of experts on food.
Moreover, the Eurobarometer [16] shows that the association of food with health is
in the EU only made by one person in five; the idea that society should transform
into a hospital is not very realistic. As long as health remains one of the consider-
ations in food choice and supply, and the multiple functions of food stay in place,
there is some reason to reconsider the intricate relationship between the two [14].
When personal nutrition on a modest scale is implemented, then the first area of
ethical concern can be overcome, is our final assessment. This allows for reflection
on the new gray zones between food and health when personalized nutrition on a
modest scale is implemented in certain fields. This reflection can begin by analyzing
the second and third main areas of ethical concern (Table 16.2).
nutrition and public health policy should have precedence over an individualist health
approach for reasons of efficacy [24] or for ethical reasons [25]. Moreover, they
argue that personal nutrition and health care have collective effects. When most rich
people in the Western world only want personal nutrition and care, the danger
emerges that the public nutrition and health system will seriously be damaged as a
result of a kind of personal/collective dilemma [18]. We will run the risk that the
food and medical problems of the poor in the West and the South will be neglected
because the richer people all will choose according to their own personal nutrition
and health projects. Because of these enormous collective effects, personalized
nutrition is not only a personal decision up to the individual, but affects the whole
population. Social responsibility seems to require that the whole population be
considered. For instance, it is a well-known fact that socioeconomic status (SES)
and health expectancy are strongly linked. The rich always have more opportunities
to look after their health than the poor and have therefore a longer life expectancy.
International public health (for example, World Health Organization [4]) aims at
improving health on a populational level. How far should personal responsibility for
health be taken into account in setting the agenda for national and global public
health nutrition policies, and what role has collective responsibility to play?
The answer of many food scientists and food industrialists to this question is
that personalized nutrition and its concomitants, such as a gene passport, comprising
the results of screening and testing, are means of individual empowerment and should
replace public health nutrition policies [26,27]. Moreover, insurance companies and
many governments are of the opinion that consumers should take more responsibility
for their health and not rely too much on governmental health regulations. They are
arguing: Youre responsible to do what is healthy and call this self-control and
autonomy (see the Blair government paper [28]).
However, some food companies are rather hesitant to enter the market of per-
sonalized nutrition. In an adjacent field, that of personalized medicine, which is
already more developed, it is very often the case that personalized medicines are
not commercially viable. Personalized medicine means that companies will have to
be content with only a (small) share of the market and cannot cover all the (potential)
patients. As a result, few new pharmagenomic drugs are filed for license in the U.S.
According to the FDA [29], the number of new drugs and biological applications
submitted to it has declined significantly. Moreover, the U.K.-based Public Health
Association [25] argues in its reaction to the Blair governments policies [28] that
public health should be the main target of governmental policies. This association
argues that in the long run the poorer parts of the population, and health itself, will
lose in a health and food system in which individual responsibility is the main
organizing principle [31]. In the same vein, Lang [24] also makes a strong case for
the primacy of public vs. individualized health nutrition politics. Targeting whole
populations provide governments with better chances of public health success,
whereas targeting at risk individuals could be socially divisive [24, p. 110]. But
in the end he concedes that individual tendencies [24, p. 120] do have to play a
role in a public health system.
Moreover, from an ethical point of view, one could argue with Kant [11] that
personalized nutrition in the sense of a gene passport or chart means disempowerment
9281_C016.fm Page 230 Thursday, July 26, 2007 7:12 PM
and not enrichment of social autonomy, because one makes oneself dependent on a
physician. Only as a socially autonomous member of a rational community can
personalized nutrition be given a try, and the issues boil down to the balanced
relationship between autonomy and solidarity, not an either/or relationship [14].
Concluding, there are some arguments in favor of personalized nutrition, as long
as it does not displace public health nutrition [30]. Also, from a utilitarian standpoint,
there is place for personalized nutrition as long as individual responsibility can
defray the costs.
confronts ethics with the necessity of transforming itself from a taken for granted
discipline to a searching and experimenting discipline. Ethics will need to incor-
porate these uncertainties and ambiguities into the use of nutrigenomics services
and products in the daily life of consumers, in adequate policy measures and in
ethical acceptable research agendas for research institutes.
An ethics for consumers can draw inspiration from Kants adage, dont obey
your doctor [11], which in a certain sense is right, but does not assist in establishing
the new necessary connections between food and health (drugs). The traditional
ethical concepts such as informed choice and individual responsibility are not
adequate for this new task [10]. When the collective implications of the individual
choice for personalized nutrition are so huge, more collective ways of opinion and
decision making should be tried in which all stakeholders have to readjust their
opinions and interests according to the ongoing discussion. This implies that the
ethics of protection of the individual against the apparently mighty powers of state
and professionals is not appropriate; more desirable is an ethics of participation in
continuous discussion on the intricacies of nutrigenomics.
Moreover, consumers need an ethics of dealing with uncertainties in the sense
of identifying and selecting between important, major, minor, and unimportant
choices. There are no general tools or guidelines to deal with that process of selection,
but there are general procedures such as consultations, deliberations, and exchange
of war stories and anecdotes. The main purpose of these procedures is to discern
commonalities and particularities in your own life and that of other affected indi-
viduals (Kants comparative universality! [12]). Some ethical support can be given
by Putnams distinction between commonsense doubt, meaning the selecting of more
and less certain cases, and philosophical doubt, i.e., the radical denial of all certain-
ties, which is most of the time not meaningful [34]:
We have to remind ourselves of the distinction between common sense doubt and
philosophical doubt. Finishing in believing in something is not really a human possi-
bility. Criticism cannot be a reason for universal skepticism. The fact that sometimes
we are wrong is not a reason to really doubt every particular conviction.
The reason that informed choice is not a good ethical concept in this context has to
do with the fact that it does not say anything about selecting the more certain and
less certain recommendations and the incorporation of well-established health con-
siderations into ones diet and personal health. The needed ethics of cooperation
between consumers and professionals and of sharing life experiences implies that
consumer groups should try to bridge the gap between production and consumption
of food and new technologies and to incorporate the new health considerations into
their food style together with the main stakeholders such as governments and food
industry [35].
With respect to the ethics for governments and their food policy, the uncertainties
in the gray zone between food and health are best tackled not by prohibiting the gray
zone but by regulating these on a social and technological level (coevolution) [14].
Governments should organize the research agenda of nutrigenomics in a democratic
way, not exclusively oriented toward personalized nutrition, but oriented toward the
9281_C016.fm Page 232 Thursday, July 26, 2007 7:12 PM
relative certainties of this new approach to identifying and selecting their chief
vulnerabilities and be able to incorporate these in their narrative life plans. This
implies not only room for informed choice, but also for consultations and deliber-
ations on these uncertainties. The ethics of these issues is still in its infancy, because
until now the ethics of food (and of health) has had a strong defensive, and not
proactive outlook. However, from a public health point of view, the potentialities of
nutrigenomics in improving the health of the population, in particular of the least
well off, should be developed as well. Governmental policies and research institutes
should allow their research agendas to be shaped not only by personalized nutrition
but by common nutritional issues as well.
KEY READINGS
Castle, D., Cheryl Cline, Abdallah S. Daar, Peter A. Singer, and Charoula Tsamis, 2006, Science,
Society and the Supermarket : The Opportunities and Challenges of Nutrigenomics, Wiley.
Food Ethics Council, 2005, Getting Personal: Shifting Responsibilities for Health, Brighton.
Korthals, M., 2004, Before Dinner: Philosophy and Ethics of Food, Springer, Berlin.
Korthals, M. (Ed.), Genomics and Obesity, Dordrecht, Springer, forthcoming.
Mller, M. and Kersten, S., 2003, Nutrigenomics: goals and strategies, Nature Reviews
Genetics, 4, 4, 315322.
REFERENCES
1. TIMES, Sweet Taste of Success, June 24, 1999.
2. Della Penna, D., Nutritional genomics: manipulating plant micronutrients to improve
human health, Science 285: 375379, 1999.
3. Porter, R., The Greatest Benefit of Mankind, Norton, New York, 1999.
4. WHO (2004): Global strategy on diet, physical activity and health. Fifty-seventh World
Health Assembly. WHA57.17. May 22, 2004; WHO (2005): The Bangkok Charter for
health Promotion in a Globalized World. WHO, August 2005. Available on www.who.int.
5. Friedman, J.F., Modern science versus the stigma of obesity. Nature Medicine, 10(6):
563569, 2004.
6. Roth, J., Qiang, X. et al., The obesity pandemic: where have we been and where are
we going? Obesity Research 12(Suppl.): 88S100S, 2004.
7. Kaput, J. and Rodriguez, R.L., Nutritional genomics: the next frontier in the postge-
nomic era. Physiological Genomics. 16, 166177. PMID: 14726599, 2004.
8. Chadwick, R., Nutrigenomics, individualism and public health. Proceedings of the
Nutrition Society, 63(1): 1616, 2004.
9. For a general discussion on utilitarianism and other ethical perspectives. Singer, P.,
Practical Ethics, Cambridge University Press, Cambridge, 1979; 2nd ed., 1993.
10. Mill, J.S., On Liberty, Oxford, Oxford University Press, 1859, 1978.
11. Kant, I., What is enlightenment?, Guyer, P. Ed. The Cambridge Companion to Kant.
Cambridge University Press, 1992. Translation from Was ist Aufklrung? 1785.
12. Kant, I., Anthropologie in pragmatischer Hinsicht (Anthropology from Pragmatic
Point of View) (1798/1790), idem, Kants Werke, Darmstadt, 1975.
13. ibidem, p. 567.
14. Korthals, M., Before Dinner, Dordrecht: Springer, 2004.
9281_C016.fm Page 234 Thursday, July 26, 2007 7:12 PM
15. Castle, D., Cline, C., Daar, A.S., Singer, P.A., and Tsamis C., Science, Society and the
Supermarket : The Opportunities and Challenges of Nutrigenomics, Wiley, 2006.
16. Eurobarometer, 2005, Risk issues, http://ec.europa.eu/public_opinion/archives/ebs/
ebs_238_en.pdf.
17. Korsmeyer, C., Making Sense of Taste, Cornell University Press,1999; Korsmeyer
doesnt take into account the personal history of tasting in listing factors that deter-
mine food preferences, p. 82, 98.
18. Crawford, R., Healthism and the medicalization of every day. International Journal
of Health Services, Vol. 10, No. 3, 1980.
19. ONeill, Onora, Public health or clinical ethics: thinking beyond borders, Ethics and
International Affairs, 16, 2002.
20. Food Ethics Council, Getting personal: shifting responsibilities for health, Brighton, 2005.
21. Hillsdon, M., Foster, C., Naidoo, B., and Crombie, H., The effectiveness of public
health interventions for increasing physical activity among adults: a review of reviews.
Evidence briefing of the NHS Health Development Agency. www.hda.nhs.uk/evidence.
2004.
22. Petrini, The Pleasures of Slow Food: Celebrating Authentic Traditions, Flavors and
Recipes, Chronicle Books, 2002.
23. Foucault, M., History of Sexuality, Vintage, Part 2, 1990.
24. Lang, T. and Heasman, M., 2005, Foodwars, London: Earthscan.
25. UKPHA, Public Health Association: Choosing Health or Losing Health? 2003.
26. German, J.B. and H.J. Watzke, Personalizing foods for health and delight. Compre-
hensive Reviews in Food Science and Food Safety 3: 145151, 2004.
27. Koelen, M.A. and Lindstrm, B., Making healthy choices the easy choices: the role
of empowerment. EJCN. 59, Suppl. 1, S10S16, 2005.
28. U.K. Department of Health, Choosing Health, London, 2004.
29. Darnton-Hill, I., Margetts, B., and Deckelbaum, R., Public health nutrition and genet-
ics: implications for nutrition policy and promotion, Proceedings of the Nutrition
Society, Vol. 63, No. 1, February 2004, pp. 173185(13).
30. Khoury, M.J. and Mensah, G.A., Genomics and the prevention and control of common
chronic diseases: emerging priorities for public health action. Prevention of Chronic
Diseases (serial online) April 2005. Available from http://www.cdc.gov/ pcd/issues/
2005/apr/05_0011.htm.
31. Cain, M. and Schmid G., From nutrigenomics science to personalized nutrition: The
market in 2010. The Institute for the Future, CAL.; Castle (2003). Clinical challenges
posed by new biotechnology: the case of nutrigenomics. Post-Graduate Medical
Journal, 79: 6566, 2003.
32. Sinha, G., the diet that fits analyzing metabolism for personalized nutrition,
Scientific American 292(3): 22 March 2005; DeCamp, M. and Sugarman, J. (2004).
Ethics and research assessing the relative roles of genes and the environment.
Accountability in Research: Policies and Quality Assurance 11: 161182.
33. Tate, T.K. and Goldstein, D.B., Will tomorrow s medicines work for everyone?
Nature Genetics 36(11): S34S42, 2004.
34. Putnam, H., Pragmatism: An Open Question, Oxford: Blackwell, pp. 5781, 1995.
35. Liakopoulos, M. and Schroeder, D., Trust and functional foods. New products, old
issues, Poiesis & Praxis: International Journal of Technology Assessment and Ethics
of Science, Vol. 2, No. 1, 4152, 2003.
36. Meek, J., Public misled by gene test hype. In The Guardian, March 12, 2002.
37. BBC-News. (July 17, 2005). DNA test for diabetes and obesity. Health. Retrieved
November 7, 2005.
9281_S003.fm Page 235 Wednesday, July 18, 2007 4:33 PM
Section III
The Future of Personalized
Nutrition
9281_S003.fm Page 236 Wednesday, July 18, 2007 4:33 PM
9281_C017.fm Page 237 Wednesday, July 18, 2007 3:18 PM
17 International Efforts
on Nutrigenomic Health
for Individuals in the
Global Community
Jim Kaput
CONTENTS
237
9281_C017.fm Page 238 Wednesday, July 18, 2007 3:18 PM
17.1 INTRODUCTION
Nutrigenomics is the study of how constituents of the diet interact with genes,
and their products, to alter phenotype and, conversely, how genes and their products
metabolize these constituents into nutrients, antinutrients, and bioactive com-
pounds. The data and results from nutrigenomics research are predicted to provide
individuals with knowledge for optimizing nutrient intakes with the goal of pre-
venting or delaying the onset of diseases. The research and application path to
personalized nutrition faces the challenges of the chemical complexity of food,
genetic heterogeneity of human populations, intricacies of biological processes,
and the need for new methods of analyzing high-dimensional data sets generated
by omics technologies. With the success of the Human Genome and HapMap
projects, many scientific disciplines are beginning to develop best practices and
share data sets for understanding biological processes. A number of large-scale
collaborative programs focusing on nutrientgene interactions have been initiated
within the past 3 years. Nutrigenomic researchers individually, in centers, and in
multi-institutional programs have recognized the need for developing best prac-
tices, fostering international collaborations, and sharing data sets. The scientific
and humanitarian need for forming a network of networks and the initial steps on
that pathway are reviewed in this chapter.
The excitement caused by the unparalleled international collaborations to ana-
lyze the human genome (1,2) and its variability (3) created lofty expectations in the
medical and pharmaceutical sectors, food industries, media, and informed public.
Information obtained with genetic and emerging omic tests are predicted to person-
alize medical treatments (4), drug selection (5), and nutritional advice (69), and
perhaps lead to the development of healthier processed foods targeted to individuals
(10,11). Although many believe these goals will be realized, expectations have given
way to the sobering recognition of the challenges of understanding genetic causes
of complex traits such as chronic diseases and the role of environment in regulating
expression of genetic makeup.
Results from genetic studies attempting to associate a gene or its variants to a
disease or specific metabolic phenotype illustrate the challenges: many published
genedisease associations could not be replicated in subsequent studies [e.g.,
(12,13)]. This led one accomplished epidemiologist to publish a report entitled Why
Most Published Research Findings Are False (14), identifying experimental designs
where sample sizes lacked appropriate statistical power, control groups were not
appropriately matched to cases, study participants had differing genetic admixtures
(populations stratification), and data that were overinterpreted (among others, see
References 15 to 18). Researchers working at the intersection of nutrition and
genetics added that dietgene interactions are major contributors to the control of
gene expression and could influence associations among genesphenotype and
dietary intake (7,1921). Reaching the goal of personalized nutrition will require an
understanding of the scientific challenges of identifying nutrientgene interactions
involved in health or disease development and realistic solutions to surmount those
challenges.
9281_C017.fm Page 239 Wednesday, July 18, 2007 3:18 PM
hydrolase gene (LCH) that arose as a mutation around 9000 years ago is responsible
for the altered expression pattern of the gene. Individuals not descended from the
founding European population retain the normal metabolism of being unable to
metabolize lactose as adults: less than 5% of the individuals in southeast Asia can
safely drink milk (7). Certain populations in Africa also exhibit lactase persistence
even though the African and European populations have been separated for at least
~35,000 years. DNA sequence analyses showed that a large percentage of Africans
in certain populations have a G-14010C polymorphism 5 to the LCH gene. This
polymorphism arose about ~7000 years ago (44). The maintenance of these LCH
promoter variants may confer selective advantages that include improved nutrition,
prevention of dehydration, and improved calcium absorption in environments that
are nutrient poor. The independent selection and maintenance of separate mutations
in African and European populations are a prime example of convergent natural
selection (44). Different alleles conferring the same phenotype (lactase persistence)
have practical consequences for both pharmacogenomics and nutritional genomics:
gene variantphenotype associations may be population specific because members
of different ancestral groups may have different polymorphisms affecting the same
gene.
The implications of this genetic similarity and diversity are also significant for
nutrigenomic researchers because the frequency of gene variants differs among
populations. Perhaps as importantly, gene variants and their products (either RNA
or DNA) interact with other genes and their products in pathways and networks.
Such genegene interactions [or epistasis (4547)] work at the protein and enzyme
[and probably iRNA (48)] levels to buffer biochemical reactions (49,50). The
concept of buffering is important because the presence or absence of a particular
gene variant (i.e., SNP) is not an all or none phenomenon. For example, the activity
of a gene A variant may be affected by a second gene B (through its protein product)
that is inherited separately (i.e., that is, on an unlinked chromosomal region). The
result of epistasis is that a single SNP is not deterministic but expresses itself within
the background of the individuals genome (that is, the total set of all variations in
their DNA). Hence, SNP analyses of individual genes must be accompanied by
analyses of genomic variations, an analytical approach that has yet to be developed.
A specific example is illustrative: a specific group of SNPs (called a haplotype,
and in this case, HapK) in leukotriene A4 hydrolase (LTA4H) was associated with
increased risk of myocardial infarction (MI). However, the risk of MI was significantly
greater in African Americans who carried the HapK haplotype derived from Europe
(51) compared to the presence of the HapK in a European genetic background. The
genetic explanation is that SNPs in LTA4H affect the activity of that enzyme product
and its interaction with other genes whose variants are found more frequently in
Africans. These results demonstrate the concept that a SNP or haplotype is context
specific (52); that is, a SNP or haplotype may contribute different amounts to disease
risk depending on ancestral background because of epistatic (genegene) interactions.
Because some of the genes that cause disease are influenced by diet, the same nutrients
may differentially affect individuals with different ancestral backgrounds. Researchers
have used self-described ethnicities as proxies for genetic architecture, but skin color
is a poor substitute for genetic analyses (53,54).
9281_C017.fm Page 242 Wednesday, July 18, 2007 3:18 PM
Analyzing complex data sets is not limited to biological data, and other disciplines
have developed algorithms that can discover nonlinear patterns in high-dimensional
data. Dimensionality reduction is a mathematical method of mapping multidimen-
sional data into a space of fewer dimensions (62,63), generating a deeper under-
standing of complex biological processes (e.g., Reference 64 and review in Reference
65). Among these methods are isomap (46,62,63), factor analysis (http://www2.
chass.ncsu.edu/garson/pa765/factor.htm), and classification tree analysis
(http://www.statsoft.com/textbook/stclatre.html), all of which can be used to analyze
complex patterns of gene variants and their influence on subphenotypes (insulin
levels or fasting glucose levels, etc.) in response to different dietary intakes. The
behavior of interacting genetic and environmental systems cannot yet be predicted
accurately and, hence, multiple approaches may be needed to extract knowledge
from experimental data. Nevertheless, data generated from different high throughput
technologies may be analyzed with more confidence when other paradigms (non-
linearity) are considered possible.
TABLE 17.1
Examples of Investigator and Center Networks
Topic or Field Title Goal Reference
TABLE 17.2
Examples of Multi-Institutional Centers for Studying
GeneNutrient Interactions
Partners/Host Institutiona Agency Focus
(continued)
9281_C017.fm Page 246 Wednesday, July 18, 2007 3:18 PM
LipGene Diet, genomics, and the metabolic syndrome: an integrated nutrition, agrofood,
social, and economic analysis
http://www.lipgene.org
Twenty-five partners European Union Role of dietary fats and genetic variation
Sixth Framework projects: Create agrofoods of LC n-3 PUFA
Food Quality and Safety
Ten European countries
Priority Alter milk fat from cows and fats in poultry
Consumer attitudes: MS and agrofood
technologies
Economic barriers to new agrofood
technologies
Diet vs. drug costs in management of the MS
Dissemination and education program
(continued)
9281_C017.fm Page 248 Wednesday, July 18, 2007 3:18 PM
Source: Adapted from Kaput, J. 2007. Nutrigenomics 2006 Update, Clinical Chemistry and Lab-
oratory Medicine, in press.
TABLE 17.3
Road Map for Nutritional Genomics Research
Topic Rationale Outcome
Data federation Scalable databases share genetic, Allow for greater statistical
phenotypic, dietary, nutritional status, and power across populations
other environmental and cultural information
Larger study Statistical power limitations Improve reliability of results
populations
Phenotypes More reliable analyses and consistency Improve reliability of results
Nutrient intakes Quantitative measurements Improve reliability of results
Genomic controls Ancestry background analyses Control for epistasis
Study Inclusion of diverse cultures and genotypes More complete understanding
participants of nutrientgene interactions
Assess other Affects expression of genetic info Reduce noise in data sets
environmental
variables
Partnerships Resource building for capturing nontraditional More complete understanding
among data and interactions among data sets of nutrientgene interactions
academia,
industry, society
Source: Adapted from Kaput, J., Ordovas, J.M., Ferguson, L. et al. 2005. Br J Nutr 94: 62332.
9281_C017.fm Page 250 Wednesday, July 18, 2007 3:18 PM
The only nutrigenomics organization whose mission is multinational and that is not
explicitly linked to a specific research program is The European Nutrigenomics
Organisation (NuGO). The mission of NuGO is to develop, integrate, and facilitate
genomic technologies, infrastructure, and research for nutritional science, to train a
new generation of nutrigenomics scientists, in order to improve the impact of nutri-
tion and genetics in health promotion and disease prevention. NuGO has recently
established the Nutrigenomics Society to provide its leadership and resources to all
in the international nutrigenomic community.
The first step of this effort was the creation of a central Web site (http://www.
nugo.org) that provides information to the researcher in pages called the Nutrigenomics
Information Portal (NIP) (71). Divided into Research, Technology, Outreach, and
Partnerships, the NIP provides a meeting and resource site for those interested in
nutrigenomics topics. A second section of the Web site, labeled Facts about Nutrige-
nomics (FAN), is designed for the educated public. FAN describes the field, key terms
and concepts and information about the collaborative effort. The final section of the
public pages are entitled Nutrigenomics Stakeholders (NS), which is a section focusing
on industry, health professional, regulators, politicians, patient organizations, and gov-
ernment departments. Many who use the Web recognize the need for reliable infor-
mation from experts, and the content of the Nutrigenomics Society intends to provide
balanced and accurate descriptions of nutrigenomics science and its applications. The
Web site is an ongoing effort that was initiated in mid-2006 (71). The ultimate goal
of the Web site registration of individuals and projects, resource development, and
best practices is to create data that can be shared across studies.
In addition to the education and outreach efforts of NuGO, the NCMHD Center of
Excellence in Nutritional Genomics at the University of CaliforniaDavis has a nutrige-
nomics LISTSERV to alert subscribers to various scientific and public news articles,
nutrigenomic activities, and issues facing the field. Started in July of 2002, the LIST-
SERV currently has over 1600 subscribers (January 2007) in over 50 countries.
Many scientists doing research at the turn of the millennium were taught reductionist
approaches to science. This philosophy and the associated methods have contributed
immensely to scientific progress and will always be needed to understand individual
pathways and reactions of complex networks. However, reductionism cannot fully
explain complex, interacting systems because expression of genetic information relies
on environmental factors that change during life. Developmental and aging processes
of biological systems also vary over time, possibly altering geneenvironment inter-
actions. Such complexity is rarely accounted for in reductionist experimental designs.
Nutrigenomics projects therefore will require best practices developed by experts
in different disciplines. NuGO began this process in early 2004 with ongoing meet-
ings and discussions among European scientists at different institutions. This effort
has generated three best practices papers in transcriptomics (72), metabolomics (68),
and proteomics (73). These recommendations focus on the technologies of each
9281_C017.fm Page 251 Wednesday, July 18, 2007 3:18 PM
discipline rather than experimental designs for human, laboratory animal, cell culture
experiments, or for pre- and postexperiment biocomputation methods. Because tech-
nologies and experience change, these best practices will require ongoing discus-
sions, which will, in the near future, include non-European scientists. Online forums
will be created to foster interactions across national and physical divides.
developed countries may soon enable inclusion of scientists and citizens of devel-
oping countries to participate in the research and its applications. Genetic testing
costs are continually falling and economies of scale may allow the analyses of
individuals in different populations at only incrementally higher costs. Collaborative
efforts will likely allow the transfer of technologies and information which may aid
these disadvantaged nations economically (9496).
Several different groups are exploring the moral issues of doing research in
resource-poor countries (97) and in developing ethical statements and procedures
for such research. The Nuffield Council on Bioethics (http://www.nuffieldbioeth-
ics.org/) has thoroughly reviewed this topic in 1999 (98), with follow-up discussions
published in 2005 (99). The Joint Centre for Bioethics of the University of Toronto
has provided guidelines for ethics and reasons for using technology and genomics
for science and economic development in disadvantaged countries (95,96,100).
The need for addressing the imbalances in nutrient availability, genetic diversity,
and economic development in developing countries has been more eloquently
expressed by nutritionists (76), geneticists (77), economists (100,101), and ethicists
(9599). Disadvantaged individuals and populations can, and should be, included
in new and ongoing studies through appropriate collaborations with scientists, eth-
icists, public health officials, and institutions in developing countries as can those
most vulnerable subpopulations in developed countries. Science and morality justify
these efforts.
These efforts are similar to the recent description of the new nutrition project
(102104), an elucidation of the precepts in the Giessen Declaration (105). That
declaration calls for the expansion of the definition, concepts, and dimensions of
nutritional research and its applications to include biological, social, and environ-
mental principles and methods. The new nutrition would address personal, popu-
lation, and planetary health through an integrative approach of studying and
understanding food systems. The authors of the Giessen Declaration believe that
nutrition sciences should be, and are rightfully, involved with addressing global food
and nutrition insecurity and inadequacy and the new epidemics of obesity, diabetes,
and other chronic diseases whose incidence is rising in developed and developing
countries. The sentiments of the Giessen Declaration and its accompanying expla-
nations are similar to those expressed by an informal international network of
nutrigenomic and related scientists (39).
issues are even more significant. Approximately 70% of urban dwellers have access
to safe drinking water with per-liter consumption of about 7 l. Only 15 to 20% of
country or rural inhabitants have access, with some consumption estimates of
between 3 and 12 l per day. The recommended amounts for basic needs of drinking,
culinary, personal hygiene, and other domestic uses is 20 to 50 l per day for rural
and urban residents, respectively (106). Accessibility is defined as the geographic
proximity of safe source of drinking water points within 15 minutes walk or a radius
of 1 km from users homes (106) [Emphasis added]. In Ethiopia, 21% of the rural
population, 84% of the urban dwellers, and 30% of the countrys inhabitants have
access to safe drinking water (called drinking water coverage). These numbers
translate to significant biological consequences:
The federal Ethiopian government promises full health care, but only ~52% of the
population has access (Ethiopian Ministry of Health, Annex 1). The number of
physicians per 1000 people is 0.029 in Ethiopia, and 2.3 in the U.S. The point of
this listing is that examining nutrientgene interactions, or simply nutrient require-
ments (76), will require significant planning and adjustments of protocols many
researchers take for granted there are few studies that monitor water intake or
energy expenditure of walking to water sources. Nutrigenomics research cannot
address all of these issues but can, and should, focus on extending its research and
humanitarian efforts to those in developing and transitional countries.
(71) is emerging that will encourage scientists from any related discipline to join
and participate. The organization of this democratic, ground-up approach will be
done initially through an international Web site hosted by NuGO (71). The interna-
tional team is committed to including people of all nations and ethnicities in the
research and benefits of that research, in large part to reduce the disparities in health
care and living standards across the world. Significant challenges for this research
effort must be overcome in developed economies the effort requires mainly
political will. For developing countries facing infectious diseases, lack of access to
safe water, food insecurity, poor sanitation and health care, the issues are significantly
greater. Genetic variation is likely to produce differences in requirements of micro-
nutrients needed to maintain health and allow for full attainment of physical and
mental health. Hence, the perspective for nutrigenomics is not only improved
nutrition for individuals in developed countries, but also the potential for evidence-
based improvements of public health for citizens of the world.
ACKNOWLEDGMENTS
The preparation of this manuscript was supported by the National Center for Minority
Health and Health Disparities Center of Excellence in Nutritional Genomics, Grant MD
00222 and from the European Union, EU FP6 NoE Grant, Contract No. CT2004-505944.
KEY READINGS
1. NCMHD Center of Excellence in Nutritional Genomics Web site: http://nutrigenomics.
ucdavis.edu.
2. NuGO Web site: http://www.nugo.org.
3. The Case for Strategic International Alliances to HarnessNutritional Genomics for
Public and Personal Health (39).
4. A variant of the gene encoding leukotriene A4 hydrolase confers ethnicity-specific
risk of myocardial infarction (51).
5. Developmental origins of the metabolic syndrome: prediction, plasticity, and
programming (59).
REFERENCES
1. Lander, ES, Linton, LM, Birren, B et al. 2001. Initial sequencing and analysis of the
human genome. Nature 409: 860921.
2. Venter, JC, Adams, MD, Myers, EW et al. 2001. The sequence of the human genome.
Science 291: 130451.
3. The International HapMap, C. 2005. A haplotype map of the human genome. Nature
437: 12991320.
4. Kaput, J, Noble, J, Hatipoglu, B, Kohrs, K, Dawson, K, and Bartholomew, A. 2007.
Application of Nutrigenomic Concepts to Type 2 Diabetes Mellitus. Nutrition, Metab-
olism, and Cardiovascular Disease, 17: 89103.
5. Kaput, J, Perlina, A, Hatipoglu, B, Bartholomew, A, and Nikolsky, Y. 2007. Nutrigenom-
ics: Concepts and Applications to Clinical Medicine. Pharmacogenomics, 8: 369390.
9281_C017.fm Page 256 Wednesday, July 18, 2007 3:18 PM
30. Reynolds, TL, Nemeth, MA, Glenn, KC et al. 2005. Natural variability of metabolites
in maize grain: differences due to genetic background. J Agric Food Chem 53: 100617.
31. Haytowitz, D, Pehrsson, PR, and Holden, JM. 2002. The Identification of Key Foods
for Food Composition Research. J Good Composition Anal 15: 183194.
32. Kim, KH and Sobal, J. 2004. Religion, social support, fat intake and physical activity.
Public Health Nutr 7: 77381.
33. Irwin, M. 2002. Effects of sleep and sleep loss on immunity and cytokines. Brain
Behav Immun 16: 50312.
34. Redwine, L, Hauger, RL, Gillin, JC et al. 2000. Effects of sleep and sleep deprivation
on interleukin-6, growth hormone, cortisol, and melatonin levels in humans. J Clin
Endocrinol Metab 85: 3597603.
35. Gleeson, M, Nieman, DC, and Pedersen, BK. 2004. Exercise, nutrition and immune
function. J Sports Sci 22: 11525.
36. Nieman, DC, Davis, JM, Henson, DA et al. 2003. Carbohydrate ingestion influences
skeletal muscle cytokine mRNA and plasma cytokine levels after a 3-h run. J Appl
Physiol 94: 191725.
37. Nieman, DC, Dumke, CI, Henson, DA et al. 2003. Immune and oxidative changes
during and following the Western States Endurance Run. Int J Sports Med 24: 5417.
38. Nieman, DC, Davis, JM, Brown, VA et al. 2004. Influence of carbohydrate ingestion
on immune changes after 2 h of intensive resistance training. J Appl Physiol 96:
12928.
39. Kaput, J, Ordovas, JM, Ferguson, L et al. 2005. The case for strategic international
alliances to harness nutritional genomics for public and personal health. Br J Nutr
94: 62332.
40. Patterson, N, Hattangadi, N, Lane, B et al. 2004. Methods for high-density admixture
mapping of disease genes. Am J Hum Genet 74: 9791000.
41. Tishkoff, SA and Verrelli, BC. 2003. Patterns of human genetic diversity: implications
for human evolutionary history and disease. Annu Rev Genomics Hum Genet 4:
293340.
42. Jorde, LB and Wooding, SP. 2004. Genetic variation, classification and race. Nat
Genet 36(Suppl. 1): S2833.
43. Enattah, NS, Sahi, T, Savilahti, E et al. 2002. Identification of a variant associated
with adult-type hypolactasia. Nat Genet 30: 2337.
44. Tishkoff, SA, Reed, FA, Ranciaro, A et al. 2007. Convergent adaptation of human
lactase persistence in Africa and Europe. Nat Genet 39: 3140.
45. Chiu, S, Diament, A.L., Fisler, J.S., and Warden, C.H., Gene-Gene Epistasis and Gene
Environment Interactions Influence Diabetes and Obesity, in Nutritional Genomics.
Discovering the Path to Personalized Nutrition, J Kaput, Rodriguez, RL, Eds. 2006,
John Wiley and Sons: Hoboken, NJ. pp. 135152.
46. Moore, JH. 2004. Computational analysis of gene-gene interactions using multifactor
dimensionality reduction. Expert Rev Mol Diagn 4: 795803.
47. Carlborg, O and Haley, CS. 2004. Epistasis: too often neglected in complex trait
studies? Nat Rev Genet 5: 61825.
48. Nakahara, K and Carthew, RW. 2004. Expanding roles for miRNAs and siRNAs in
cell regulation. Curr Opin Cell Biol 16: 12733.
49. Hartman, JL, Garvik, B, and Hartwell, L. 2001. Principles for the buffering of genetic
variation. Science 291: 10014.
50. Hartman, JL, IV, Genetic and Molecular Buffering of Phenotypes, in Nutritional
Genomics. Discovering the Path to Personalized Nutrition, J Kaput, Rodriguez, RL,
Eds. 2006, John Wiley and Sons: Hoboken, NJ. pp. 105134.
9281_C017.fm Page 258 Wednesday, July 18, 2007 3:18 PM
51. Helgadottir, A, Manolescu, A, Helgason, A et al. 2006. A variant of the gene encoding
leukotriene A4 hydrolase confers ethnicity-specific risk of myocardial infarction. Nat
Genet 38: 6874.
52. Klos, KLE, Kardia, SLR, Hixson, JE et al., Linkage Analysis of Plasma ApoE in Three
Ethnic Groups: Multiple Genes with Context-Dependent Effects. 2005. p. 157167.
53. Kittles, RA and Weiss, KM. 2003. Race, ancestry, and genes: implications for defining
disease risk. Annu Rev Genomics Hum Genet 4: 3367.
54. Shriver, MD, Mei, R, Parra, EJ et al. 2005. Large-scale SNP analysis reveals clustered
and continuous patterns of human genetic variation. Hum Genomics 2: 8189.
55. Kaput, J. An Introduction and Overview of Nutritional Genomics: Application to
Type 2 Diabetes and International Nutrigenomics, in Nutritional Genomics. Discov-
ering the Path to Personalized Nutrition, J Kaput, Rodriguez, RL, Eds. 2006, John
Wiley and Sons: Hoboken, NJ. pp. 136.
56. Steinmetz, A. 2003. Treatment of diabetic dyslipoproteinemia. Exp Clin Endocrinol
Diabetes 111: 23945.
57. Cordain, L, Eades, MR, and Eades, MD. 2003. Hyperinsulinemic diseases of civilization:
more than just Syndrome X. Comp Biochem Physiol A Mol Integr Physiol 136: 95112.
58. Curtis, J and Wilson, C. 2005. Preventing type 2 diabetes mellitus. J Am Board Fam
Pract 18: 3743.
59. McMillen, IC and Robinson, JS. 2005. Developmental origins of the metabolic
syndrome: prediction, plasticity, and programming. Physiol Rev 85: 571633.
60. Rosenzweig, JL, Weinger, K, Poirier-Solomon, L et al. 2002. Use of a disease severity
index for evaluation of healthcare costs and management of comorbidities of patients
with diabetes mellitus. Am J Manage Care 8: 9508.
61. Florez, JC, Hirschhorn, J, and Altshuler, D. 2003. The inherited basis of diabetes
mellitus: implications for the genetic analysis of complex traits. Annu Rev Genomics
Hum Genet 4: 25791.
62. Roweis, ST and Saul, LK. 2000. Nonlinear dimensionality reduction by locally linear
embedding. Science 290: 23236.
63. Tenenbaum, JB, de Silva, V, and Langford, JC. 2000. A global geometric framework
for nonlinear dimensionality reduction. Science 290: 231923.
64. Dawson, K, Rodriguez, RL, and Malyj, W. 2005. Sample phenotype clusters in high-
density oligonucleotide microarray data sets are revealed using Isomap, a nonlinear
algorithm. BMC Bioinformatics 6: 195.
65. Kaput, J and Dawson, K. 2007. Complexity of type 2 diabetes mellitus data sets
emerging from nutrigenomic research: a case for dimensionality reduction? Mutation
Research. In press.
66. Hood, L, Heath, JR, Phelps, ME et al. 2004. Systems biology and new technologies
enable predictive and preventative medicine. Science 306: 6403.
67. Bijlsma, S, Bobeldijk, I, Verheij, ER et al. 2006. Large-scale human metabolomics
studies: a strategy for data (pre-) processing and validation. Anal Chem 78: 56774.
68. Gibney, MJ, Walsh, M, Brennan, L et al. 2005. Metabolomics in human nutrition:
opportunities and challenges. Am J Clin Nutr 82: 497503.
69. van Ommen, B. 2004. Nutrigenomics: exploiting systems biology in the nutrition and
health arenas. Nutrition 20: 48.
70. Kaput, J. 2007. Nutrigenomics 2006 Update. Clinical Chemistry and Laboratory
Medicine, 45: 279287.
71. Kaput, J, Astley, S, Renkema, M, Ordovas, J, and van Ommen, B. 2006. Harnessing
nutrigenomics: development of web-based communication, databases, resources, and
tools. Genes Nutr. 1: 512.
9281_C017.fm Page 259 Wednesday, July 18, 2007 3:18 PM
72. Garosi, P, De Filippo, C, van Erk, M et al. 2005. Defining best practice for microarray
analyses in nutrigenomic studies. Br J Nutr 93: 42532.
73. Fuchs, D, Winkelmann, I, Johnson, IT et al. 2005. Proteomics in nutrition research:
principles, technologies and applications. Br J Nutr 94: 30214.
74. Qi, L, Tai, ES, Tan, CE et al. 2005. Intragenic linkage disequilibrium structure of the
human perilipin gene (PLIN) and haplotype association with increased obesity risk
in a multiethnic Asian population. J Mol Med.
75. Shin, HD, Kim, KS, Cha, MH et al. 2005. The effects of UCP-1 polymorphisms on
obesity phenotypes among Korean female subjects. Biochem Biophys Res Commun
335: 62430.
76. Uauy, R. 2005. Defining and addressing the nutritional needs of populations. Public
Health Nutr 8: 77380.
77. Consortium, IH. 2004. Integrating ethics and science in the International HapMap
Project. Nat Rev Genet 5: 46775.
78. Jiang, YH, Bressler, J, and Beaudet, AL. 2004. Epigenetics and human disease. Annu
Rev Genomics Hum Genet 5: 479510.
79. Lucas, A. 1998. Programming by early nutrition: an experimental approach. J Nutr
128: 401S406S.
80. Gallou-Kabani, C and Junien, C. 2005. Nutritional epigenomics of metabolic syn-
drome: new perspective against the epidemic. Diabetes 54: 1899906.
81. Victora, CG, Morris, SS, Barros, FC et al. 1998. The NCHS reference and the growth
of breast- and bottle-fed infants. J Nutr 128: 11348.
82. Hughes, RG and Lawrence, MA. 2005. Globalization, food and health in Pacific
Island countries. Asia Pac J Clin Nutr 14: 298306.
83. Wilcken, B, Bamforth, F, Li, Z et al. 2003. Geographical and ethnic variation of the
677C>T allele of 5,10 methylenetetrahydrofolate reductase (MTHFR): findings from
over 7000 newborns from 16 areas world wide. J Med Genet 40: 61925.
84. Chen, WJ, Chen, CC, Yu, JM et al. 1998. Self-reported flushing and genotypes of
ALDH2, ADH2, and ADH3 among Taiwanese Han. Alcohol Clin Exp Res 22:
104852.
85. Nakajima, T, Wooding, S, Sakagami, T et al. 2004. Natural selection and population
history in the human angiotensinogen gene (AGT): 736 complete AGT sequences in
chromosomes from around the world. Am J Hum Genet 74: 898916.
86. Darnton-Hill, I, Margetts, B, and Deckelbaum, R. 2004. Public health nutrition and
genetics: implications for nutrition policy and promotion. Proc Nutr Soc 63: 17385.
87. Neel, JV. 1962. Diabetes mellitus: a thrifty genotype rendered detrimental by
progress? Am J Hum Genet 14: 35362.
88. Lieberman, LS. 2003. Dietary, Evolutionary, and Modernizing Influences on the
Prevalence of Type 2 Diabetes. Annu Rev Nutr:
89. Epping-Jordan, JE, Galea, G, Tukuitonga, C et al. 2005. Preventing chronic diseases:
taking stepwise action. Lancet 366: 166771.
90. James, PT, Leach, R, Kalamara, E et al. 2001. The worldwide obesity epidemic. Obes
Res 9(Suppl. 4): 228S233S.
91. Iannotti, LL, Tielsch, JM, Black, MM et al. 2006. Iron supplementation in early
childhood: health benefits and risks. Am J Clin Nutr 84: 126176.
92. McCann, JC and Ames, BN, Is docosahexaenoic acid, an n-3 long-chain polyunsaturated
fatty acid, required for development of normal brain function? An overview of evidence
from cognitive and behavioral tests in humans and animals. 2005. pp. 281295.
93. Research, GFfH, The 10/90 Report on Health Research 2001 2002. 2002, Global
Forum for Health Research, World Health Organization: Geneva, Switzerland.
9281_C017.fm Page 260 Wednesday, July 18, 2007 3:18 PM
94. Acharya, T, Daar, AS, Thorsteinsdottir, H et al. 2004. Strengthening the role of
genomics in global health. PLoS Med 1: e40.
95. Daar, AS, Thorsteinsdottir, H, Martin, DK et al. 2002. Top ten biotechnologies for
improving health in developing countries. Nat Genet 32: 22932.
96. Singer, PA and Daar, AS. 2001. Harnessing genomics and biotechnology to improve
global health equity. Science 294: 879.
97. Marshall, PA. 2005. Human rights, cultural pluralism, and international health
research. Theor Med Bioeth 26: 52957.
98. Bioethics, NCo. 2002. The ethics of research related to healthcare in developing
countries: 190.
99. Bioethics, NCo. 2005. The ethics of research related to healthcare in developing
countries: a follow-up Discussion Paper:
100. Sachs, JD and Reid, WV. 2006. Environment. Investments toward sustainable devel-
opment. Science 312: 1002.
101. Sachs, JD, The End of Poverty. Economic Possibilities for Our Time. 2005, New York:
The Penguin Press.
102. Leitzmann, C and Cannon, G. 2005. Dimensions, domains and principles of the new
nutrition science. Public Health Nutr 8: 78794.
103. Beauman, C, Cannon, G, Elmadfa, I et al. 2005. The principles, definition and
dimensions of the new nutrition science. Public Health Nutr 8: 6958.
104. Cannon, G and Leitzmann, C. 2005. The new nutrition science project. Public Health
Nutr 8: 67394.
105. 2005. The Giessen Declaration. Public Health Nutr 8: 7836.
106. Kunie, A and Ahmed, A. 2005. An overview of environmental health status in Ethiopia
with particular emphasis to its organization, drinking water and sanitation: a literature
survey. Ethiop J Health Dev 19: 89103.
107. Sorsa, S, Kidanemariam, T, and Erosie, L. 2002. Health Problems of street children
and women in Awassa, Southern Ethiopia. Ethiop J Health Dev 16: 129137.
108. Churchill, GA, Airey, DC, Allayee, H et al. 2004. The Collaborative Cross, a com-
munity resource for the genetic analysis of complex traits. Nat Genet 36: 11337.
109. Khoury, MJ. 2004. The case for a global human genome epidemiology initiative. Nat
Genet 36: 10271028.
110. Ioannidis, JPA, Gwinn, M, Little, J et al. 2006. A road map for efficient and reliable
human genome epidemiology. Nat Genet 38: 35.
111. Cavanaugh, JA. 2003. IBD International Genetics Consortium: international cooper-
ation making sense of complex disease. Inflamm Bowel Dis 9: 1903.
9281_C018.fm Page 261 Wednesday, July 18, 2007 3:20 PM
CONTENTS
18.1 INTRODUCTION
Although not typically appreciated, processed foods are perhaps the most critical
elements to the success of modern civilization. In fact, it is a tribute to the success of
the processed food industry that urbanized societies are largely blissfully unaware of
the multiple values inherent in their foods that make life as we know it possible. The
core values of processing of foods are primarily to convert unstable, unsafe, and
sparingly nutritious agricultural commodities into safe, stable, and completely nourish-
ing foods. Industrialized societies continue to add and combine more values within
the foods that they consume. This chapter will first examine the core values of indus-
trialized foods and then suggest where the most obvious opportunities lie to increase
or add value in the future. Finally, the chapter will examine the advances in science
and technologies that are emerging, or would be necessary, to enable such a future.
261
9281_C018.fm Page 262 Wednesday, July 18, 2007 3:20 PM
Consumer
Demands
Food
Scientic Industrial
Knowledge Technology
FIGURE 18.1 Key inputs to food design, formulation, and process innovations.
9281_C018.fm Page 264 Wednesday, July 18, 2007 3:20 PM
Product-centered/Commodity-driven
Product matches expectations
Sensory Nutrition
Functionality Optimization Shelf-Life
STOP
Food Safety
Packaging Home
Raw Ingredients Body
Processing PRODUCT Storage Preparation
Material Preparation Eects
Distribution Eating
FIGURE 18.2a Current approach to food processing, designed to assemble food products
according to the consumers anticipated view of particular products or product categories.
all three of these areas, with progressive enhancements in the values provided by
foods. Although many trends will be discernible, one unifying theme is the increasing
personalization of foods, and this singular trend toward more personalized food
product propositions will transform all of the values of processed foods.
The basic driving forces of the current industrial food sector are illustrated in
Figure 18.2a. Food production today is a product-centric, commodity-driven indus-
try. With food safety as the critical, nonnegotiable core, the food product itself is
the defining element. Once established in the consumers mind as a viable product,
whether a fast food hamburger or a sweet carbonated beverage, the consumer is
buying a familiar product with known organoleptic properties. The profit drivers all
lie within the ability of competitors to drive the costs of delivering that same product
lower. Nutrition is only considered as an ingredient additive at the end (vitamin-
enriched), without compromising the basic product perception. This is a model that,
while providing food products at spectacular volumes and low prices, is providing
very little net value either to the consumer or to the production chain itself. Agri-
culture is essentially eating itself alive trying to provide cheaper and cheaper food
products, and the consumers are literally eating themselves to death because of the
poor nutritional contents. It is now possible to foresee an entirely different model
(Figure 18.2b) in which the consumer becomes the defining element. In this model
Consumer-centered/Benet-driven
Food delivers benets
FIGURE 18.2b Proposed approach to foods for the future, designed around the consumer
and delivering foods that address the specific needs, desires, and aspirations of each consumer
for their individual health and well-being.
9281_C018.fm Page 265 Wednesday, July 18, 2007 3:20 PM
the goal is to provide values to the consumer. With the consumer as the goal, many
values such as health, performance, and even enjoyment and delight become
attributes that enhance the overall value proposition to the consumer. In this model,
any stage in the agricultural process that is capable of adding, or enhancing, benefits
to the consumer creates value at that level, from seed producer through farmer
through processor to delivery system. It is critical to such a model that the variation
among consumers be recognized, and thus foods must become more personalized.
Consumers themselves will be the major drivers of the trend to personalization.
The average consumer is increasingly gaining awareness of his or her wants and
needs, and growing scientific knowledge and technological advances will race to
meet them. Examples of this ongoing progressive trend are numerous, yet the
diversity of foods and consumers makes it difficult to predict the future. Nonetheless,
it is possible to categorize innovations within each of the previously stated areas
(desired consumer values, molecular scientific knowledge, and efficient industrial
technologies) to gain a sense of predictability. From the consumer perspective, the
major trend is toward individualization of health. The last decade has seen an
explosion in the personalization of consumer products of virtually all kinds from
computers to entertainment, from transportation to cosmetics (German et al., 2005b).
This trend has been evident in foods, extending to all of the values of foods, but
will increasingly include health as a personal value proposition. Consumers know
more about their own health issues and the potential of foods to alter them. Although
it has been a core value of processed foods to ensure safety, the future safety of
foods will take on a more personal dimension. Foods will include means of making
consumers themselves safer. Initially this will mean more specific control of food
processing and specific labeling (no nuts, gluten-free, etc.). Eventually foods will
incorporate various means of ensuring that however consumers choose to assemble
their diet, risks to safety will be minimized.
It is certainly not revolutionary for a wine maker who has used yeast to ferment
grape juice into wine to imagine a life-form participating in the processing of an
agricultural commodity. Nonetheless, the improvements in molecular dexterity, cost,
and control that biological strategies contribute to food processing will lead to a
dramatic increase in biologically based innovations in foods. Many of the compo-
nents of human milk are active catalysts whose benefits to the infant are provided
by the catalytic activity itself (e.g., bile-salt-stimulated lipase makes lipids more
available for absorption). At present such activities are destroyed during processing,
whereas in future they will be retained. Other molecules present in milk are highly
dependent on the subtleties of molecular structure lost during processing, for exam-
ple, the milk fat globule in which surface carbohydrates capable of binding microbial
toxins are lost because of homogenization. Again, processes designed to maintain
these structures will be valuable but likely only to a subset of consumers; hence,
the need to personalize the value chain from processing to product.
more diverse agriculture. Human agriculture is based on a very limited subset of all
living organisms. The reason for this is that most organisms evolved under the constant
Darwinian selective pressure to avoid being eaten. As a result of this pressure, most
plants elaborated secondary metabolism leading to the production of compounds to
discourage animal predation. Antinutritive, and overtly toxic alkaloids, flavonoids,
protease inhibitors, phytates, etc., render most existing wild plants inedible. However,
as we gain mastery of the genomics of plants and the history of their evolution becomes
clear, it will be possible to selectively eliminate the antinutritional and toxic elements.
With such tools, plant geneticists will be able to dramatically expand the available
genetic stocks for agriculture and increase the diversity of inputs to the food supply.
What the organic food movement of today is struggling to provide as value proposi-
tions, with increased sustainability, environmental protection, and species diversity
will be dramatically enhanced by the ability to understand the genetics of all of the
agricultural players from plants to animals, and even microorganisms.
In addition to making plant diversity an asset to food production, genomics will
increase our biotechnology dexterity with a wide array of microorganisms. Bacteria,
yeasts, and molds represent a very successful traditional means of food processing
providing increased safety, stability, and nutritional quality while modifying many
of the taste, flavor, and textural properties of biomaterials from cultured vegetables,
meats, cheeses, and cereals. The combination of knowledge and biological tools that
microbial genomics is providing will revolutionize the bioprocessing of foods.
Not only bacteria, yeasts, and molds but viruses too will be recruited to the task of
improving food values.
Although agricultural species diversity will increase dramatically, one of the
interesting outcomes of this knowledge explosion involves understanding the diver-
sity within species. Research is revealing all of the ways that humans differ from
each other, one of them certainly being the way different humans respond to diet.
The fields of nutrigenomics and nutrigenetics are rapidly compiling examples of
genetic variation in the normal population that predicts variation in health outcome.
From dietary fat and lipoprotein atherogenicity (Krauss, 2001; Ordovas, 2003) to
dietary carbohydrate and weight loss (Martinez et al., 2003), to dietary vitamins and
risk of birth defects (Whetstine et al., 2002), genetic variation is being demonstrated
to be a key component of the variation in human health that is ascribable to diet.
The most widely used index of metabolic health is the level of circulating lipids in
blood, primarily cholesterol and triglycerides. Just by looking at the mechanisms
by which lipids are cleared from blood, we see a vivid story of the genetic variation
within the human population. Clearance of triglyceride-rich lipoproteins is affected
by polymorphisms at several gene loci, including apoB (Rantala et al., 2000), apoAV
(Lai et al., 2003), apo E (Ostos et al., 1998), and lipoprotein lipase (Mero et al.,
1999). The consequences of these genotypic differences are that individuals can be
assigned to varying responses to, for example, high- and low-fat diets. The size and
atherogenicity of cholesterol-rich, low-density lipoproteins, and their response to
diet are mitigated by heritable genes that can be demonstrated to persist in the
offspring of phenotypic carriers (Krauss, 2001). Again, as a result of defining these
genotypes, individuals who vary in response to diet will eventually be
assigned/guided to lower or higher saturated fat-containing diets.
9281_C018.fm Page 269 Wednesday, July 18, 2007 3:20 PM
The success of these emerging studies implies that parts of our health will be
predictable from simple genotypic analyses. However, this is not the only basis on
which individuals in the population differ with respect to responses to diet. We are
all exposed to different environments, (exercise, diets, etc.), and these are now
recognized as increasingly important to the net effects of dietary choices on our
health (Bateson et al., 2004). If multiple variables relate to the response of health
to diet beyond genotype of individuals, then health itself will have to be explicitly
measured using technologies akin to disease diagnostics. However, these assessors
will need to be more accurate and capture the fluctuations in metabolism and
physiology that correspond to variations in aspects of health, not simply the diag-
nostics of disease (German et al., 2005a).
Some individuals have high levels of circulating cholesterol because their hepatic
and peripheral biosynthesis rates are high (Schmitz and Langmann, 2006). Some
have high cholesterol because they absorb inordinately high levels from the intestine,
and still others have high cholesterol because they secrete less sterol both as cho-
lesterol and bile acids from the liver as bile (Plosch et al., 2005). By measuring
sterol biosynthesis intermediates quantitatively in plasma, relative sterol synthesis
rates can be estimated, thus identifying those who synthesize high levels of choles-
terol; by measuring the immediate products of bile acid synthesis in plasma, those
who produce and excrete less bile can be identified, and by measuring phytosterols
in plasma, those individuals who hyperabsorb sterols can be identified (reviewed in
German et al., 2005a). Now, with each of the different causes of high cholesterol
identified within individuals, appropriate strategies for effective intervention can also
be designed. Cholesterol synthesis drugs are highly effective in those who oversyn-
thesize cholesterol, but are relatively ineffective in those who hyperabsorb. Similarly,
sterol absorption inhibitors are highly effective in those who hyperabsorb sterols.
The new sciences of omics genomics, transcriptomics, proteomics and
metabolomics have emerged as a result of the developments of complete databases
of genomes and their products, and the technologies to simultaneously analyze all,
or significant subsets of all members of a biological class of molecules (genes,
mRNAs, proteins, and metabolites) (Raamsdonk et al., 2001). These tools have
dramatic implications for the routine assessment of health because of their potential
to literally measure a complete picture of functional biology in a tissue or fluid
compartment at once (Nicholson and Wilson, 2003). Omic assessment technologies,
and in particular metabolomics, thus offer a unique opportunity for clinical chemistry
not only to monitor the progress of disease states but also to characterize and define
how to maintain health. With such information on health status, individuals will be
able to gain knowledge of their unique metabolic needs, and most importantly, will
be able to act on these needs with diet as the major intervention. Assessment conducted
routinely (e.g., monthly, yearly) will also provide feedback regarding the success of
all food, drug, and lifestyle changes within a time frame that reinforces them or
redirects them as appropriate. Again, this type of assessment will bring a much more
empowered consumer population to the marketplace for agriculture and its products
foods. Even more importantly, with assessed health status of individuals in place as
a consumer asset, it will be possible for diets to be much more effective in preventing
diseases long before they occur, at which points foods become a competitive alter-
native to drugs in maintaining health and preventing disease.
There is an important economic argument for disease prevention that undermines
most attempts to bring preventive medicine into practice. How much will a consumer
pay for a drug/therapeutic/bioactive substance to prevent a disease that they will
never have? The answer is discouragingly simple, very little. Drugs are perceived
to be a valuable product category precisely because they are designed to cure existing
disease. In prevention, you are currently healthy and by following a particular
regimen of food consumption, behavior, or lifestyle, you are unlikely to develop a
future disease. Hence, the value of any single therapeutic preventive is very low. In
order to be successful, preventive strategies have to be very cost-effective. In fact,
it is precisely because foods are already routinely consumed, carry an existing value
9281_C018.fm Page 271 Wednesday, July 18, 2007 3:20 PM
proposition, and can easily deliver multiple biological activities that they are so
appropriate as the engines of prevention.
formulate, stabilize, and texturize food products, many of these desired goals could
be achieved without the macroscopic, high-energy unit operations currently used
today. In fact, to the extent possible, food processing will take advantage of the
properties of the molecules within biomaterials to spontaneously self-assemble into
desired forms (Leser et al., 2006). Self-assembly of biomolecules is not a new
concept invented for the food sciences life itself is based upon it. The most
remarkable property of life is that every organism, from the smallest to the largest,
is ostensibly self-assembled from simple biopolymers. As the sciences of genomics
begin to understand the more structure-specific properties of molecules as gene
products, the self-assembly properties of biomolecules even complex colloidal
properties will similarly be understood with sufficient accuracy to control them (de
Campo et al., 2004). There are already many examples of adapting the natural
properties of biopolymers for the net benefit of food products. Cheese is an obvious
illustration of this principle: the properties of casein micelles are captured for
aggregation into three-dimensional gel networks.
18.6 CONCLUSIONS
The growing emphasis in the food industry in the 21st century is, and will continue
to be, to deliver safe, convenient, affordable, and delicious food products that provide
the means to assemble diets that maintain and improve the health of consumers. The
power of nutrition research and food processing to solve health problems is illustrated
by the success of identifying the essential nutrients, recognizing their absolute
requirements, and retaining, enriching, and fortifying the food supply to ensure that
9281_C018.fm Page 275 Wednesday, July 18, 2007 3:20 PM
KEY READINGS
Collins FS, Green ED, Guttmacher AE, Guyer MS; U.S. National Human Genome Research Institute.
2003 A vision for the future of genomics research. Nature. 24; 422(6934): 83547.
Morris CE, Sands DC. The breeders dilemma yield or nutrition? Nat Biotechnol. Sep-
tember 2006; 24(9): 107880.
Sturino JM, Klaenhammer TR. Engineered bacteriophage-defence systems in bioprocessing.
Nat Rev Microbiol. May 2006; 4(5): 395404.
9281_C018.fm Page 276 Wednesday, July 18, 2007 3:20 PM
REFERENCES
Alberti G., 2001, Noncommunicable diseases: tomorrows pandemics, Bull. WHO 79, 907.
Bateson P., D. Barker, T. Clutton-Brock, D. Deb, B. DUdine, R. A, Foley, P. Gluckman, K.
Godfrey, T. Kirkwood. M. M. Lahr, J. McNamara, N. B. Metcalfe, P. Monaghan, H.
G. Spencer and S. E. Sultan, 2004, Developmental plasticity and human health, Nature
430(6998), 419421.
Bruin S. and ThRG Jongen, 2003, Food process engineering: the last 25 years and the
challenges ahead, Crit. Rev. Food Sci. Food Safety 2, 4281.
Catchpole G. S., M. Beckmann, D. P. Enot, M. Mondhe, B. Zywicki, J. Taylor, N. Hardy, A.
Smith, R. D. King, D. B. Kell D.B., O. Fiehn and J. Draper, 2005, Hierarchical
metabolomics demonstrates substantial compositional similarity between genetically
modified and conventional potato crops, Proc. Natl. Acad. Sci. USA 102, 1445814462.
Clydesdale F. M., 1997, A proposal for the establishment of scientific criteria for health claims
for functional foods, Nutr. Rev. 55, 413422.
Clydesdale F., 2004, IFT Expert Panel Report on Functional Foods: Opportunities and Chal-
lenges, Food Technol. 58, 3540.
Collins F.S., E.D. Green, A.E. Guttmacher and M.S. Guyer; US National Human Genome Research
Institute. 2003 A vision for the future of genomics research. Nature. 24; 422(6934): 83547.
de Campo L., A.Yaghmur, L. Sagalowicz, M.E. Leser, H. Watzke and O. Glatter, Reversible
phase transitions in emulsified nanostructured lipid systems. Langmuir. June 22, 2004;
20(13): 525461.
Delmer D. P., 2005, Agriculture in the developing world: Connecting innovations in plant
research to downstream applications, Proc. Natl. Acad. Sci. USA 102, 1573915746.
European Plant Science Organization, 2005, European plant science: a field of opportunities,
J. Exp. Bot. 56, 1699709.
German J. B, F. L. Schanbacher, B. Lnnerdal, J. Medrano, M. a. McGuire, J. L. McManaman,
D. M. Rocke, T. P. Smith, M. C. Neville, P. Donnelly, M. C. Lange and R. E. Ward,
2006, International milk genomics consortium, Trends Food Sci.Technol. 17, 656661.
German J. B., B. D. Hammock and S. M. Watkins, 2005a, Metabolomics: building on a
century of biochemistry to guide human health, Metabolomics 1, 38.
German J. B., C. Yeretzian and H. J. Watzke, 2005b, Personalizing foods for health and
preference, Food Technol. 58, 2631.
Gould G. W., 2000, Preservation: past, present and future, Br. Med. Bull. 56, 8496.
Grigorov M. G., 2005, Global properties of biological networks, Drug Discov. Today 10,
365372.
Krauss R. M., 2001, Dietary and genetic effects on low-density lipoprotein heterogeneity,
Annu. Rev. Nutr. 21, 283295.
Lai C. Q., E. S. Tai, C. E. Tan, J. Cutter, S. K. Chew, Y. P. Zhu, X. Adiconis and J. M. Ordovas,
2003, The apolipoprotein A5 locus is a strong determinant of plasma triglyceride
concentrations across ethnic groups in Singapore, J. Lipid Res. 44, 23652373.
Lange M. C., D. G. Lemay and J. B. German, 2007, A multi-ontology framework to guide
agriculture and food towards diet and health, J. Sci. Food Agric. (in press).
Lemay D. G., A. M. Zivkovic and J. B. German, 2007, Building the bridges to bioinformatics
in nutrition research, Am. J. Clin. Nutr. 13 Apr 2007, online, 10.1002/jsfa. 2832.
Leser M. E., L. Sagalowicz, M. Michel, and H. J. Watzke, Self-assembly of polar food lipids.
Adv. Colloid Interface Sci. 2006 Nov 16; 123126: 12536.
Martinez J. A., M. S. Corbalan, A. Sanchez-Villegas, L. Forga, A. Marti and M. A. Martinez-
Gonzalez, 2003, Obesity risk is associated with carbohydrate intake in women
carrying the Gln27Glu beta2-adrenoceptor polymorphism, J. Nutr. 133, 25492554.
9281_C018.fm Page 277 Wednesday, July 18, 2007 3:20 PM
Index
A B
Adipocyte fatty-acid-binding protein (-FABP), 20 Beta-carotene, 9697
Adiponectin, 92 Biofluids, 1620
Adipose tissue Biomarker(s), 269
nutrients and, 9394 blood glucose, 67
plasticity, 9293 candidates, 4
remodeling, 97 inflammation, 67
vascularity, 90 insulin resistance, 67
Advanced Foods and Materials Network, 245 profiling, 10
Alcohol, 118, 137 protein, 19
bowel cancer risk and, 80 proteome analysis and discovery of, 1820
dependence, 118, 122, 124125, 135, 222 rheumatoid arthritis, 19
diabetes and, 64, 66 universal, 30
metabolizing genes, 252 Bladder cancer, 96
predictors of consumption, 124 Bowel cancer, 78. See also Colorectal cancer
Alcoholism, 118, 122, 124125, 135, 222 Breast cancer, 96, 157
Alzheimers disease, 19, 49, 51 angiogenesis and, 95
Angiogenesis, 89, 97 antioxidants and, 80
adipose tissue plasticity, 9293 carotenoids and, 82
cancer and, 90, 95 folate intake and, 80
gene expression and, 94 genes and, 53, 78, 79, 137
inhibition, 92, 94, 96 hereditary, 137
main principles, 9095 risk, 230
nitric oxide and, 92 genetic testing for, 216
personalized nutrition and, 9597 wine and, 230
polymorphism and, 97
polyphenols and, 9596
regulation, 92, 94, 97 C
Antioxidant(s), 49, 52
breast cancer and, 80 Calcium, 19, 65, 104, 241
dietary and, 55 Cancer, 4, 8, 49, 53
mitochondrial defense, 80 angiogenesis in, 90, 95
phytochemicals as, 117 biology, 7677
polyphenol, 55 bladder, 96
prostate cancer and, 81 breast, 53, 78, 80, 95, 96, 137, 157, 216, 230,
Antioxidant response element, 52 231 (See also Breast cancer)
ARS Childrens Nutrition Research Center, 245 colon, 51, 75, 95
Arthritis, 27, 110 colorectal, 52, 78, 79, 82, 95, 136, 137
rheumatoid, 19, 50, 51, 95 diagnostics, 1819
Aspirin, colorectal cancer and, 82 diet and, 76, 8082, 170, 222
Assessment technology, 269271 gastric, 52, 78, 83
Atherosclerosis, 20 genes and, 53, 75, 76, 78, 79, 137
angiogenesis and, 95 immune response and, 54
development, 267 incidence, 137
inflammation and, 51 initiation, 51
measure, 37 liver, 140, 141
Athletes, 160163 lung, 53, 78, 95, 96
279
9281_C019.fm Page 280 Wednesday, July 18, 2007 4:13 PM
Index 281
Index 283
Index 285
Index 287