Methods in Enzymology Volume 58 Cell Culture
Methods in Enzymology Volume 58 Cell Culture
Methods in Enzymology Volume 58 Cell Culture
Many of the problems that have caught the interest and imagination of
biochemists are studied best with cultured cells. We offer in this volume,
in a format familiar to investigators in biochemistry, the general techniques necessary for working with cells in culture and illustrate such
general methods with specific examples from the large variety of cells that
have been cultivated.
The tools and methods for cell culture are presented in Part I. Part II
provides a group of specialized techniques that are useful for many of the
applications that biochemists and other investigators with their widely
different approaches may require. Part III is concerned with specific
methods for specific cell types that have been chosen to represent the
wide range of cells that may now be prepared.
There is some duplication in the presentations. For example, portions
of certain methods are repeated in one or another form both in Part I and
Part III. We believe that this repetition is necessary to convey faithfully to
the reader a complete method of proven effectiveness. Additionally, we
hope that a heuristic effect will be achieved that will enable investigators
unfamiliar with cell culture to assess what is available and to predict what
might be most suitable for their own purposes.
WILLIAM B. JAKOBY
IRA H. PASTAN
xiii
Contributors to V o l u m e L V I I I
Article numbers are in parentheses following the names of contributors.
Affiliations listed are current.
Department of
Pediatrics, Division of Medical Genetics,
Washington University School of Medicine, St. Louis, Missouri 63110
P. M. GULLINO (14), Laboratory of
Pathophysiology, National Cancer Institute, National Institutes of Health,
Bethesda, Maryland 20014
RICHARD G. HAM (5), Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder,
Colorado 80309
EDWARD HAWROT (53), Department of
Neurobiology, Harvard Medical School,
Boston, Massachusetts 02115
IZUMI HAYASHI (6), Department of Biology,
University of California, San Diego, La
Jolla, California 92093
W. FRED HINt (39), Department of Entomology, Ohio State University, Columbus, Ohio 43210
BHARATI HUKKU (13), The Child Research
Center of Michigan, Children's Hospital
of Michigan, Detroit, Michigan 48201
ERIC HUNTER (32), Department of Microbiology, The Medical Center, University of Alabama in Birmingham, Birmingham, Alabama 35294
SHARON HUTCHINGS (6), Department of Biology, University of California, San Diego, La Jolla, California 92093
ROGER H. KENNETT (28), Department of
Human Genetics, The Human Genetics
Cell Center, University of Pennsylvania
School of Medicine, Philadelphia,
Pennsylvania 19104
GEORGE KHOURY (34), Laboratory of DNA
Tumor Viruses, National Cancer Institute,
National Institutes of Health, Bethesda,
Maryland 20014
MICHAEL KLAGSSRUN (50), Departments of
Surgical Research and Biological Chemistry, Children's Hospital Medical Center,
Harvard Medical School, Boston, Massachusetts 02115
R. A. KNAZEK (14), Laboratory of
Pathophysiology, National Cancer Institute, National Institutes of Health,
Bethesda, Maryland 20014
K. S. KOCH (47), The Salk Institute, Post
Office Box 1809, San Diego, California
92112
JEFFREY GRUBB (38),
xi
xii
[1]
ASPECTS
OF
TISSUE
CULTURE
LABORATORY
[1 ] P h y s i c a l A s p e c t s o f a T i s s u e C u l t u r e L a b o r a t o r y
BASIC METHODS
[1]
[1]
cells but also with such things as pipettes which are used to transfer
culture media. This is of particular importance in connection with toxic
substances introduced into glassware by normal processing. While
everyone is aware of the problems associated with microbial contamination, little thought may be given to toxic organic products introduced
during the manufacture of certain items, inorganic residues from detergent washes, or contamination by metal ions sloughed from pipes. Therefore, proper procedures for cleaning and sterilization of glassware should
be carefully followed; several such procedures are available in the literature. 3''~-7 Also it is recommended that glassware used in cell culture procedures be employed exclusively for this purpose and not mixed with
glassware used for other purposes. This includes not only the culture
vessels themselves but flasks, pipettes, and other miscellaneous items.
This precaution insures that all material used in culturing techniques has
been subjected to the same vigorous cleaning and that diffficult-to-remove
reagents do not contaminate the culture systems.
A complete separation of the cleaning facilities from the preparation
and the aseptic areas is the ideal situation; however, because of space
limitations, the preparation area can be combined with the cleaning area if
the glassware is not routinely contaminated with viruses or bacteria. If at
all possible, the aseptic areas should be maintained in a location isolated
from the cleaning area. The general size of the cleanup area is largely
dependent upon the quantity of material to be handled, but a laboratory of
100 to 150 ft2 will accommodate the maximum amount of equipment that
would be needed. In laboratories where acid cleaning is to be employed,5.7
a fume hood with sufficient ventilation and safety features should be incorporated into the overall design.
In general the layout of the laboratory will be determined by the location of the sink. The sources of hot and cold tap water will dictate the
placement of the washing equipment, i.e., decontamination and soaking
buckets, water purification system, and pipette washer. If the volume of
glassware is sufficiently large, a built-in glass washer would be advantageous. Several commercially available models are acceptable; however, an
adequate supply of purified water is necessary for the final rinses. Water
of suitable purity for these final rinses can be obtained by a single glass
distillation, demineralization, or reverse osmosis. The choice of purification method depends on such factors as the condition of the untreated
source water, the quantity of water needed, and the amount of space
available for the necessary equipment. It may be practical to employ a
single water purification system for all laboratory needs if the system is
r F. M. Price and K. K. Sanford, Tissue Cult. Assoc. Man. 2, 379 (1976).
BASIC METHODS
[1]
[1]
Since autoclaving is the most commonly used method, a word of caution is necessary about the quality of steam used to supply the autoclave.
When the steam is heavily contaminated with impurities, they settle on
the surfaces during autoclaving and the advantages of careful washing and
rinsing procedures are lost. This may be a major problem where larger,
shared facilities are supplied by house steam. Whatever the situation,
however, it is recommended that any autoclave using house steam be
equipped with a filtering device to remove contaminating material. A
better solution to the problem is to use an autoclave that has provisions
for its own steam generation. The water for such a unit can be obtained
from a purified source thereby eliminating the contaminants at the source.
In addition to the major items of equipment mentioned in this section,
several minor ones have proven useful and should be considered when
outfitting the cleaning and sterilizing facilities. These include carts to
facilitate the transfer of articles between the different areas of the laboratory. These are almost a necessity when the different functional units are
very widely separated. Also, provisions should be made for disposal containers in the cleanup area. Ideally, these should be closed containers
which would serve as receptacles for used disposable labware, wrappings
from sterilized items, and the like. Other items are pipette jars for soaking
pipettes before washing, a liquid detergent dispenser, and an ultrasonic
cleaning bath for hard-to-clean glassware.
BASIC METHODS
[1]
[l]
have been employed for cell culture purposes including porcelain, asbestos, sintered glass, and membrane filters. Because of their uniform
characteristics, ready availability, and superior filtering qualities, membrane filters with a 0.22 /zm pore size are most widely used for filter
sterilization in cell culture laboratories. These filters can be obtained in
sizes ranging from 13 to almost 300 mm in diameter with the subsequent
capability of sterilizing from a few milliliters to several liters of material.
The filter holders are constructed of noncorrosive material and, like the
filters, come in various sizes and configurations, some of which are prepackaged in a sterile disposable form. It is necessary to have a variety of
sizes on hand. The most useful are syringe adapters which take the 13 and
25 mm diameter filters for small volumes; the pressure filter holder for the
47 mm filter which is used for volumes up to 1 liter; and at least one of the
larger sizes for sterilizing liter quantities of media. All membrane filtration
should be accomplished under positive pressure, thereby reducing foaming, by use of either 95% air-5% CO2 or nitrogen. Larger quantities of
media may be placed in specially designed stainless-steel pressure tanks
and passed through the filter into a receiving vessel fitted with a filling bell
for dispensing into bottles for storage. The entire filtering assembly and
receiving vessel can be sterilized as a unit by autoclaving.
Both before and after cell culture media and related reagents are formulated and sterilized, specific types of storage facilities are required. At
room temperature, closed, dust-free space is necessary for the storage of
stable chemicals, pipettes and other glassware, and filtering apparatus.
Again, the size of this space depends upon the size of the operation and
the amount of formulation that is anticipated. In addition, a refrigerator,
- 2 0 freezer, and - 7 0 ultrafreezer space are required. The refrigerator is
used to store a medium once it has been sterilized. Filtered media are
usually dispensed into 100 or 500 ml bottles and are stable at refrigerator
temperatures for several weeks. The - 2 0 freezer is needed for the shortterm storage of labile biochemicals, stock solutions of amino acids and
vitamins, and serum. For longer term storage of these materials, a - 7 0
ultrafreezer is necessary.
In addition to the equipment that has been mentioned as being required
for a media preparation and storage area, several more items are recommended to complete the facility. These include an analytical balance, a
pH meter, an osmometer, a hot plate, a magnetic stirrer, and a full range
of glassware, including a good supply of bottles for media storage. It is
possible to use much of this equipment in other laboratory areas; but
certain instruments, e.g., the osmometer, should be maintained in the
media preparation area.
10
BASIC METHODS
[l]
[l]
11
closed, dust-free cabinets. Sterile, reusable glassware should be replenished frequently and a schedule of resterilization of unused material
should be maintained. Those items that are to be used in a specific cell
culture procedure should be placed in the hood prior to work. This step is
particularly important when working in a laminar flow system. If the
materials are placed in the unit 10 to 15 min before work commences then
the laminar flow system has ample time to remove particulates from the
work area.
V. Equipment for Routine Cell Maintenance
Basic tissue culture operations require an incubator, microscope, cell
repository materials, and culture vessels in which to grow the cells.
A. Incubators
Incubators may range from temperature-regulated boxes to elaborate
units which control temperature, humidity, and carbon dioxide levels in
the atmosphere. The units should be maintained on emergency power
circuits in case of an electrical power failure and should be placed close to
the aseptic work area. If the buffering system in the tissue culture medium
requires equilibration with CO2, e.g., Earle's salts, one will want to use a
CO2 incubator. When selecting a CO2 incubator for purchase, consideration should be given to the type of air flow patterns that exist in the unit.
An even temperature distribution within the incubator is essential, and
experience indicates that horizontal air flow units provide a more uniform
distribution of both temperature and CO2. Some incubators are waterjacketed but this is not a necessity. When tissue culture medium containing Hanks' salt is used or when closed culture systems are employed, a
CO2 incubator is not necessary and one may simply use a unit that
provides a constant and even temperature distribution throughout the
chamber.
B. Microscopes
Microscopes are essential since it is important to examine cultures
daily to observe their morphology and rate of growth. Many investigators
grow cells, but do not take the time to become familiar with the
morphological features of their cultures. In the last few years many laboratories have encountered problems of contamination of their cell cultures
with other cell types (see this volume [2]). Frequently, cultures have
become contaminated with HeLa cells; these cells replicate rapidly and
12
BASIC METHODS
[1]
[I]
13
they are resistant to heat and can be washed and sterilized repeatedly
without adverse effect on utility. The selection of vessel is usually based
partly on personal preference and on the availability of a properly
equipped and functioning glassware washing facility.
When borosilicate glassware is reused, care must be taken during the
washing procedure to remove all serum and cellular residues from the
glassware. In addition, washed glassware must be adequately rinsed to
insure complete removal of all traces of solvents and detergents. Because
of the problems encountered in washing and rinsing tissue culture
glassware, many laboratories routinely use commercially available,
sterile, disposable tissue culture plasticware for their monolayer culture
work. Sterile, polystyrene petri dishes are available in a variety of sizes
(35, 60, 100, and 150 mm diameter). It is important to order tissueculture-grade plastic petri dishes because cultured cells generally do not
attach to bacteriological-grade petri dishes. Various sizes of plastic tissue
culture flasks are also available (T-25, T-75, T-150 flasks). These vessels
provide 25, 75, or 150 cm 2 of surface area, respectively, for growing cells.
In addition, plastic tubes or multiwell plates can also be used as substrates
for growing monolayer cultures.
For many biochemical analyses a large number of cells are required.
The method selected for growing mass cultures is clearly related to the
particular cell strain or cell line to be used. If diploid, anchoragedependent cells are employed, then monolayer cell culture techniques are
required. In contrast, if heteroploid or other nonanchorage-dependent cell
types are selected then suspension culture techniques most probably can
be utilized. Culture systems that permit growth of large numbers of cells
with either monolayer or suspension culture techniques are commercially
available.
For large-scale growth of anchorage-dependent cells in monolayer culture, many laboratories use roller bottles. These culture vessels are available in a variety of sizes in both borosilicate glass or polystyrene plastic,
and they permit harvests of up to 2 107 viable cells from one 730-cm z
vessel. In 1970 Kruse 11 introduced a roller bottle perfusion apparatus that
permits harvest of 1.9 108 normal diploid cells or 2 109 hetcroploid
cells from a single 730 cm 2 borosilicate glass roller bottle. This high cell
density is achieved by the perfusion of fresh medium into the culture
vessels at programmed intervals. The system permits control of pH and
influent nutrient levels in a more precise manner than is possible in
monolayer cultures fed at 2 to 3 day intervals.
Polystyrene roller bottles containing a spiral of Melinex are also commercially available. The Melinex polyester sheet provides 8000 cm 2 of
11 p. F. Kruse, Jr., L. N. Keen, and W. L. Whittle, In Vitro 6, 75 (1970).
14
BASIC METHODS
[1]
culture surface, and from a single roller bottle 2 10a heteroploid cells
can be routinely harvested. 12
Suspension culture techniques are available for the large-scale production of nonanchorage-dependent cells. Cells can be maintained in suspension by use of a rotary shaker or by stirring. The shaker technique was
introduced by Earle et al. 13 and is particularly suited to studies requiring
replicate cultures. The cells are suspended in medium containing methylcellulose (15 cps) in Erlenmeyer or round-bottom flasks and placed in a
rotary shaker. Currently the most popular method for suspension cultures
uses a magnetic stirrer to keep the cells constantly agitated in tissue
culture medium supplemented with methylcellulose.14 A wide range of
sizes of spinner culture vessels are available. The stirring rate varies
depending on the cell type used, but it must be slow enough to avoid
foaming.
A recent modification of the spinner system is the spin filter culture
device. 15 This unit was developed to grow cells to high population densities and to provide for removal of drugs from the tissue culture medium at
accelerated rates. Medium is removed through a rotating filter, the design
of which prevents the accumulation of cells on or within the filter. The
volume of medium used in this unit can vary from 200--1000 ml, and cells
routinely can be grown to densities of 10s cells/ml.
Two other culture systems have recently been developed for specific
purposes, but they warrant mention because of their applicability to a
variety of research systems. The dual-rotary circumfusion system
developed by Rose permits maintenance of cells for periods of several
months in a differentiated, functional state in vitro.16 Such cells can be
readily examined throughout their in vitro culture period by highresolution microscopy, and replicate cultures are possible. The cells are
grown in Rose chambers under a cellophane membrane. In many instances the monolayer cultures are initiated from explants. The cells in
this culture system undergo only limited replication but remain in a viable
and differentiated state for several months in vitro. 18
The artificial capillary system introduced by Knazek and Gullino lr
permits cells to grow to tissue-like densities that often approximate the in
lz W. House, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, Jr., eds.), p. 338. Academic Press, New York, 1973.
~s W. R. Earle, E. L. Schilling, J. C. Bryant, and V. J. Evans, J. Natl. Cancer Inst. 14, 1159
(1954).
~ R. S. Kuchler and D. J. Merchant, Proc. Soc. Exp. Biol. Med. 92, 803 (1956).
~5 p. Hemmelfarb, P. S. Thayer, and H. E. Martin, Science 164, 555 (1969).
~8G. G. Rose, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, Jr., eds.), p. 283. Academic Press, New York, 1973.
~r R. A. Knazek and P. M. Gullino, in "Tissue Culture: Methods and Applications" (P. F.
Kruse, Jr. and M. K. Patterson, Jr., eds.), p. 321. Academic Press, New York, 1973. See
this volume [14].
[1]
15
polycarbonate membranes through which tissue culture media is constantly perfused. Diffusable cell products can be retrieved from the perfusate, and in several instances hormone-producing cells synthesize and
secrete at much greater rates in the artificial capillary system than comparable numbers of cells growing in monolayer culture.
VI. Conclusion
The material in this article, including the partial list of commercial
sources provided in the table, outlines in general terms the equipment and
A PARTIAL LIST OF SUPPLIERS OF CELL CULTURE MATERIALS
K. C. Biologicals, Inc.
P. O. Box 5441
Lenexa, KS 66215
Microbiological Associates, Inc.
4733 Bethesda Ave.
Bethesda, MD 20014
Miles Laboratories
Elkhart, IN 46514
(contmued)
16
BASIC METHODS
A PARTIAL LIST OF SUPPLIERS OF CELL CULTURE MATERIALS (continued)
Plasticware
Cooke Laboratory Products
900 Slaters Lane
Alexandria, VA 22314
Coming Glass Works
Science Products Div.
Coming, NY 14830
Costar
Div. Data Packaging Corp.
205 Broadway
Cambridge, MA 02139
Incubators
Forma Scientific
P. O. Box 649
Marietta, OH 45750
Hotpack Corp.
5086A Cottman Ave.
Philadelphia, PA 19135
Lab-Line Instruments, Inc.
Bloomingdale Ave.
Melrose Park, IL 60160
National Appliance Co.
Heinicke Instruments Co.
P. O. Box 23008
Portland, OR 97223
Percival Refrigeration & MFG. Co. Inc.
P. O. Box 249
Boone IA 50036
Wedco, Inc.
P. O. Box 223
Silver Spring, MD 20907
Falcon
Div. Becton-Dickinson
P. O. Box 243
Cockeysville, MD 20130
Filtration Systems
Lab-Tek Products
Div. Miles Laboratories
30 W. 475 N. Aurora Rd.
Naperville, IL 60540
Bioquest
Div. of Becton-Dickinson & Co.
P. O. Box 243
Cockeysville, MD 21030
Linbro Scientific
681 Dixwell Ave.
New Haven, CT. 06511
[1]
[1]
Water Systems
Barnstead Sybron Corp.
225 Rivermoor St.
Boston, MA 02132
Bellco Glass Co.
340 Edwardo Rd.
Vineland, NJ 08360
Coming Glass Works
Science Products Div.
Coming, NY 14830
Culligan USA
1 Culligan Parkway
Northbrook, IL 60062
Hydro Service & Supplies, Inc.
P. O. Box 2891
Durham, NC 27705
Ultrasciences, Inc.
2504 Gross Point Rd.
Evanston, IL 60201
Cryogenic Supplies
Forma Scientific
Box 649-T
Marietta, OH 45750
Kelvinator Commercial Products, Inc.
621 Quay St.
Manitowoc, WI 54220
Microscopes
Revco, Inc.
Scientific and Industrial Div.
1188 Memorial Dr.
West Columbia, SC 29169
Union Carbide
Linde Division
270 Park Avenue
New York, NY 10017
17
18
BASIC METHODS
[9.]
space required to initiate a cell culture program in a biochemistry laboratory. Few specific recommendations were given because the absolute
requirements for any individual program will depend upon the existing
facilities, the projected scope of the program, as well as the type of cells
to be cultured and their intended use. Although the specifics will change
according to the individual circumstances, the basic principles and problems considered here will remain unchanged regardless of the situation.
Whether coverslip cultures or mass cultures are employed, or whether the
entire operation is contained in one room or a suite of laboratories, cleanliness of glassware, purity of water and other reagents, and maintenance
of sterility will ultimately determine the reliability of any cell culture
system. The information presented here is a guide to what is necessary to
achieve these basic goals.
[2] D e t e c t i o n o f C o n t a m i n a t i o n
By GERARD J. MCGARRITY
The presence of adventitious agents in cell cultures is incompatible
with the concept of standardized, defined systems. Presence of extraneous agents may produce either gross turbidity and rapid destruction of the
culture, or no turbidity and little to moderate cytopathic effects on the
culture. The latter type can remain undetected for prolonged periods, and
have profound effects on the cell culture and experimental results.
Contamination may originate in the tissue specimen used to initiate the
cell culture, in media, especially in bovine serum, or in the general environment. The contamination may be bacterial, mycoplasmal, fungal, viral, or cellular. Detection methods described here will effectively monitor
microbiological contamination in cell cultures and media. These should be
viewed as part of an overall quality-control program. Methods to prevent
and control contamination are described elsewhere. 1-4
The limitations of detection methods must be appreciated. The major
limitations for detection of bacterial, mycoplasmal, and fungal organisms
are sample size, level of contamination, type of growth media utilized,
and presence of antibiotics in the sample. Contamination in bovine sera
i G. J. McGarrity and L. L. Coriell, In Vitro 6, 257 (1971).
2 G. J. McGarrity, Tissue Cult. Assoc. Manual 1, 167 (1975).
3 G. J. McGarrity, Tissue Cult. Assoc. Manual 1, 181 (1975).
4 G. J. McGarrity, V. Vanaman, and J. Sarama, in "Mycoplasma Infection of Cell Cultures" (G. J. McGarrity, D. G. Murphy, and W. W. Nichols, eds.), p. 213. Plenum, New
York, 1978.
[2]
DETECTION OF CONTAMINATION
19
20
BASIC METHODS
[2]
[2]
DETECTION OF CONTAMINATION
21
Repeat this procedure once a week for four passages and observe for
growth, morphology, full sheeting of cells after 1 week incubation, and for
evidence of toxicity, granularity, and vacuolization as compared to
growth of the same cells in a standard nontoxic serum.
By the end of 1 month data are available on sterility tests and growth
promotion and on whether the serum lot can be accepted or rejected.
Note: The above time and cell concentration references may have to be
modified for different cell cultures.
Cell cultures free of antibiotics can be tested for bacterial and fungal
contamination by procedures described in paragraph 1 above except that
broths are incubated only at 37 and observed for 10 days.
Mycoplasmal Testing
Mycoplasmas are particularly well suited as infectants of cell cultures.
They grow to high concentrations in cell cultures, 106 to I08 colony fprming units (CFU) per milliliterof supernatant medium being representative;
they are resistant to many antibiotics; they usually produce no gross
turbidity in the cultures they infect; they are relatively difficultto detect;
and they have profound effects on the cultures they infect. Reviews of the
effects mycoplasmas have on cell cultures and other effects are
available.6.7
E. Stanbridge, Bacteriol. Rev. 35, 206 (1971).
r G. J. McGarrity, D. G. Murphy, and W. W. Nichols, eds., "Mycoplasma Infection of Cell
Cultures." Plenum, New York, 1978.
22
BASIC METHODS
[2]
[2]
DETECTION OF CONTAMINATION
23
ited basis, the volume of medium prepared at one time can be reduced
accordingly.
For agar medium, 9 g of Noble agar (Difco Labs, Detroit, Michigan)
are added per liter of base medium, autoclaved, cooled, and dispensed in
75-ml aliquots in containers that will conveniently hold 100 ml. These
bottles are stored in the refrigerator until use. For preparation of agar
plates, heat the base medium to 96 to dissolve the agar. Place the bottles
of base medium, horse serum, yeast extract, and DNA into a water bath
at 50. Allow all components to equilibrate at 50. Add 5 ml yeast extract,
20 ml horse serum, and 1 ml of a 0.2% DNA solution to 75-ml aliquots.
Dispense into 60 x 15 mm Petri dishes (Falcon Plastics, Oxnard, California), approximately 10 ml per plate. Final mixing of media components
above 50 can result in cloudy plates that are more difficult to read. Mixing
and dispensing should be done quickly since agar will solidify at 45. Agar
plates are refrigerated and used within 1-2 weeks. Wrapping stacks of
plates in aluminum foil or plastic will reduce dehydration. The gel strength
of different lots of Noble agar may vary. The lowest concentration that
yields a gel should be used. All media should be autoclaved as soon after
mixing as pos'sible. Final pH should be 7.2 ___ 0.2.
2. Testing Serum
Testing serum for mycoplasma is performed by inoculation of 25 ml of
tl~e serum under test into each of two bottles containing 100 ml of mycoplasma broth and incubation at 37 under aerobic and anaerobic conditions.
Anareobic atmospheres can be generated easily in a Gas Pak system
(Bioquest, Inc.) Aliquots of 0.1 ml are subcultured from the broths to agar
plates after 4 and 7 days incubation. Agar plates are examined microscopically (x 100) for colonies for at least 3 weeks. Aerobic plates should be
taped to reduce drying.
3. Testing Cell Cultures
a. Monolayer Cultures. Monolayer cultures should be at least 60%
confluent for testing and be in antibiotic-free medium. Since proteolytic
enzymes may have an adverse effect on mycoplasma, cells should be
scraped from the container surface.
Remove and discard all but 3 ml of the antibiotic-free medium. Scrape
some cells from the monolayer surface with a rubber policeman or glass
rod. Place the suspension of cells and spent medium in a Wasserman or
tube of similar size. Cap. To test the suspension cultures, gently shake
flask and remove 3 ml for assay.
24
BASIC METHODS
[2]
[2]
DETECTION OF CONTAMINATION
25
working solutions are available from Research Resources Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20014. The stain is allowed to incubate for 30 min at room temperature and is rinsed with PBS. Buffered glycerol (Bioquest, Inc.) is used as
a mounting medium.
d. DNA Stain. This procedure has been reported by Chen. TM We have
incorporated the modifications of DelGiudice and Hopps into our procedures.ll
Prepare stock solution of stain to a concentration of 50 ~g/ml in distilled water. This is stored at 4 in light-protected bottles. Thimersol
(1:10,000) or similar disinfectant is added to retard microbial growth.
Working solutions are prepared fresh for each application by dilution in
potassium phospate-citric acid, pH 5.5, to make a final stain concentration of 0.05 ug/ml.
Keep the coverslip submerged in 1 ml of medium and add 1 ml of
fixative, acetic acid methanol (1:3). Aspirate after 2 min. Air dry and stain
for 10 min; wash twice with distilled water, and mount in buffered glycerol
(Bioquest, Inc.).
e. Microscopy. Preparations can be viewed with either transmitted or
incident illumination. Incident illumination has been used in this laboratory (Leitz Orthoplan plus Ploem Illumination). In immunofluorescent
preparations, individual M. hyorhinis organisms will fluoresce. This is
particulary noticeable on plasma membranes of the cultured cells. With
the DNA stain, appearance of mycoplasma and other prokaryotic organisms can be clearly observed as extranuclear fluorescent particles.
Controlled studies have indicated that the DNA stain is effective in
detecting mycoplasmas11 although standardization, proper controls, and
confirmation testing by other techniques are necessary.
f. Biochemical Tests. A variety of biochemical tests are available.
These are based on properties of mycoplasmas not shared by mammalian
cells in culture. Depending on the capabilities and facilities, these procedures might be more suitable than the techniques described above. The
limitations of each test method must be acknowledged. Appropriate controls must be included in each test.
'g. Uridine Phosphorylase Assay. This procedure was developed by
Levine and is based on the presence in all mycoplasma13 tested of uridine
phosphorylase activity.22 Although mammalian tissues in vivo have this
activity, cells in culture do not. Activity is detected by paper chromatography.
22 E. M. Levine and B. G. Becker, in "Mycoplasma Infection of Cell Cultures" (G. J.
McGarrity, D. G. Murphy, and W. W. Nichols, eds.), p. 87. Plenum, New York, 1978.
26
BASIC METHODS
[2]
A total of 2 x 107 cultured cells are used and are lysed with 3 ml of
phosphate--Triton buffer, za Levine states that as few as 3 x 106 cells in 0.5
ml buffer can be used. [14C]uridine (40 /zCi//xmol) is added as the nucleoside substrate, and the mixture is incubated at 37 for 180 min. After
incubation, uridine is separated from uracil at 37 by paper chromatography. The solvent system consists of 70 ml boric acid (4%, ca. 0.65 M),
1 ml concentrated ammonium hydroxide, and 430 ml n-butanol. Some
phase separation occurs at room temperature but is cleared by warming to
37 and shaking.
Samples are obtained at 30 and 180 min. Uridine and uracil spots are
located on the developed chromatogram with a short-wave UV light. The
radioactivity in these spots is determined. The conversion rate at 180 min
in uninfected cells is generally lower than 10%; mycoplasma-infected cells
have much higher rates, typically 20-90%.
h. Uridine/Uracil Uptake. This procedure was developed by Schneider
and Stanbridge, 14,24 and based on the marked alterations in the incorporations of free bases and nucleosides into the nucleic acid of
mycoplasma-infected cells. Mammalian cells incorporate relatively large
amounts of uridine and only negligible amounts of uracil. Mycoplasma
can incorporate uracil. Mycoplasmal uridine phosphorylase can convert
uridine to uracil, preventing uptake of uridine by cultured cells. The net
effect is that the ratio of uptake of uridine to uptake of uracil is
significantly reduced in mycoplasma-infected cells.
Cells to be tested are inoculated into each of six plastic 25 cm 2 flasks,
2.5 x 105 cells per flask. After 24 hr incubation at 37, three flasks are
inoculated with [3H]uridine, 5 /zCi/ml; three flasks are inoculated with
[3H]uracil, 5 /zCi/ml. After 18-24 hr additional incubation at 37, the
labeled nucleic acids are extracted and counted. Cells are removed from
the flask by trypsinization and pelleted at 200 g for 10 min in a refrigerated
centrifuge. The pellet is washed twice with 1 ml of phosphate-buffered
saline (PBS, pH 7.4) free of calcium and magnesium. Centrifuge at 200 g
for 10 min. Decant and suspend the pellet in 1 ml 5% trichloracetic acid
(TCA). Precipitate the nucleic acids by placing the tube in ice for 30 min.
Centrifuge at 900 g for 15 rain. Decant supernatant liquid and add 1 ml of
23 The phosphate-Triton buffer is made from three stock solutions. A 0.5 M solution of
Na~HPO4 is prepared and adjusted to pH 8.1 with 1 N HC1 (approximately 2.9 ml/100 ml).
Triton X-100 (Rohm and Haas, Inc., Philadelphia, Pennsylvania) is prepared as a 10% (v/v)
solution in water. Nonradioactive uracil is prepared in a 0.01 M water solution for use as a
tracer in the chromatography system. All solutions are stored at 4; Na2HPO4 must be
warmed before use. The final buffer is prepared as follows: 10 ml Na2NPO4, 10 ml uracil, 5
ml Triton X-100, 75 ml water. The final buffer is stored at 4.
24 E. L. Schneider and E. J. Stanbridge, Methods Cell Biol. 10, 277 (1975).
[2]
DETECTION OF CONTAMINATION
27
28
BASIC METHODS
[2]
B. Electron Microscopy
Since the concentration of mycoplasmas in infected cultures is usually
on the order of 106 to 108 per milliliter,electron microscopy can be used as
a method of detection.12 True infection must be distinguished from artifacts that can render diagnosis difficult.Use of an indicator cell culture
known to be free of mycoplasma can minimize this difficulty.
Although transmission electron microscopy (TEM) is generally more
available, sample preparation is more prolonged. In the examination of
sections, only one plane through a cell is examined. Therefore, multiple
sections are required. Because of the geometry of sectioning and the level
of mycoplasma adsorbed to cells, false negatives are possible.
Scanning electron microscopy (SEM) is more suitable for routine
screening. Van Diggelen et al. 27 and Phillips 12 have used this to detect
mycoplasma infection. Cells are grown on glass coverslips for 3-4 days
and fixed in 2.5% glutaraldehyde buffered at pH 7.4 with 0.2 M Collidine.
Cells are dehydrated in ethanol series to absolute alcohol and transferred
to acetone. Coverslips are critical point dried with liquid CO2 in a Sorvall
Critical Point Drying System, coated with gold using an Edwards 306
coater, and viewed in a scanning electron microscope.
Cultures examined by SEM are usually so heavily infected that interpretation is easy. Mycoplasma species vary in their ability to cytadsorb
onto cultured cells. In cultures infected with M. hyorhinis, most mycoplasma organisms are cytadsorbed onto cells. Little cytadsorption occurs
in A-9 mouse cells infected with Acholeplasma laidlawii, with most of the
mycoplasmas attached to the coverslip.
SEM has worked well in examination of fibroblast cultures. Some
difficulty has been observed in interpretation of lymphocyte cultures
grown in suspension (D. M. Phillips and G. J. McGarrity, unpublished
results). As with other test systems, use of an indicator cell culture may
alleviate some problems in interpretation.
Virus Testing
Bovine serum has been shown to contain several bovine viruses .28.29 It
is not always feasible for individual laboratories to screen for them unless
specialized facilities are available. The viruses most commonly isolated
from bovine sera are bovine virus, diarrhea virus, infectious bovine
rhinotracheitis virus, parainfluenze virus, and bovine herpes virus.
2T O. P. Van Diggelen, S. Shin, and D. M. Phillips, Exp. Cell Res. 106, 191 (1977).
2s C. W. Molander, A. Paley,C. W. Boone, A. J. Kniazeff, and D. T. Imagawa, In Vitro 4,
148 (1968).
29 C. W. Molander, A. J. Kniazeff,C. W. Boone, A. Paley, and D. T. Imagawa, In Vitro 7,
168 (1972).
[3]
29
[3]
Mammalian cells serially cultivated in vitro are in an artificial environment which differs in many known and unknown respects from the normal
habitat in vivo. No culture medium can duplicate the in vivo environment,
30
BASIC METHODS
[a]
and we have learned by experience that cells taken from different organs
require different culture media for growth and/or preservation of specialized function. Some cell types inevitably die out in any given culture
medium, and the cells which persist and continue to proliferate also lose
or gain characteristics which were not apparent in vivo. Examples of
changes frequently observed include: microbial contamination, chromosome abnormalities, loss of antigens, loss of ability to proliferate, and
death of the cell culture. Many schemes have been employed with varied
success to avoid or delay these undesired changes. Use of primary cell
cultures and the use of organ cultures are examples which function well
for selected short-term studies. Incubation of cell cultures at reduced
temperature slows the metabolic rate and the frequency of refeeding and
subculture. Storage of cell cultures in the frozen state following the general procedures used commercially for storage of bovine semen 1 was
reported by Scherer and Hoogasian in 19542 and by Swim et al. in 1958.3
These techniques used glycerol as an adjuvant with storage in Dry Ice at
- 7 0 . Viable cell cultures could be recovered for many months, but there
was gradual loss of viability when stored at - 7 0 . Storage of frozen cell
cultures in liquid nitrogen ( - 196) is the accepted procedure at this time,
and when properly carried out it seems to be compatible with prolonged
preservation of viability and other characteristics of mammalian cell
cultures.4-6
Without the presence of an adjuvant, freezing is lethal to most mammalian cells. Damage is caused by mechanical injury by ice crystals,
concentration of electrolytes, dehydration, pH changes, denaturation of
proteins, and other factors not well understood. These lethal effects are
minimized by: (1) adding an adjuvant such as glycerol or dimethylsulfoxide (DMSO) which lowers the freezing point; (2) a slow cooling rate
which permits water to move out of the cells before it freezes; (3) storage
at a temperature below -130 which retards the growth of ice crystals;
and (4) rapid warming at the time of recovery so that the frozen cell
culture passes rapidly through the temperature zone between - 5 0 - 0
where most cell damage is believed to occur.
Different cell strains may vary widely in their ability to withstand
physical and chemical insults during the freezing and thawing process.
[3]
31
Preservation
Nutrition
Cell cultures survive cryogenic storage best if they are frozen in a good
state of nutrition and maximal viability. This is achieved as follows:
Select cells in the logarithmic phase of growth and r e m o v e the culture
medium. Assess sterility by culture on a blood agar plate. Replace with
fresh m e d i u m and harvest 24 hr later for storage in liquid nitrogen. The
cell culture m e d i u m should be the one which best supports growth and
p r e s e r v a t i o n o f desired characteristics of the cell. Cell cultures grown in
synthetic m e d i u m without s e r u m have been successfully frozen and recovered. 'a'14 H o w e v e r , m o s t workers e m p l o y a culture m e d i u m for preservation containing 5 - 2 5 % fetal bovine s e r u m which provides unknown
7 B. Athreya, E. Grimes, H. Lehr, A. Greene, and L. Coriell, Cryobiology 5, 262 (1969).
8 L. Coriell, unpublished.
D. G. Wittinghorn, S. P. Liebo, and P. Mazur, Science 178, 411 (1972).
,0 E Mazur and J. J. Schmidt, Cryobiology 5, 1 (1966).
" P. Mazur and J. Schmidt, Cryobiology 5, 1 (1968).
,2 A. P. Rinfret, Fed. Proc., Fed. Am. Soc. Exp. Biol. 22, 94 (1963).
,3 V. Evans, H. Montes de Oca,J. C. Bryant,E. L. Schilling, and J. E. Shannon, J. Natl.
Cancer Inst. 29, 749 (1962).
,4 C. Waymouth and D. Varnum, Tissue Cult. Assoc. 2, 311 (1976).
32
BASIC METHODS
[3]
Harvest
Suspension cell cultures are counted to determine the number of viable cells, centrifuged at 800-1000 g for 10 min, and suspended in inactivated fetal bovine serum (56 for 15 min) containing 5% dimethylsulfoxide. Dilute to 10 million cells/ml, test for sterility, dispense into
ampules, and process as described below.
For monolayer cultures each flask is inspected under the binocular
microscope before harvest. Any flasks containing cells of abnormal appearance are discarded. It is desirable to release the cells from the
monolayer surface with a minimum of chemical and physical trauma. Add
to each T75 flask, 5 ml of 0.02% ethylenediaminetetraacetic acid (EDTA)
and let stand for 8 min. Remove the liquid and discard it. Add 3 ml of
EDTA in dilute trypsin 15 and observe under the binocular miscroscope
until the cell sheet starts to lift from the surface around the edges. Tap the
flask gently to loosen all the cells. This process usually requires 1 to 7 min.
Stop the action of trypsin-EDTA by adding 3 ml of growth medium containing 20% fetal calf serum. Transfer these cells to a common pool in a
centrifuge bottle that is chilled in a cracked-ice bath. Mix thoroughly,
count in a haemocytometer, and dilute with growth medium to 5 105
viable cells per milliliter as determined by trypan blue exclusion. Culture
aliquots on a blood agar plate, trypticase soy broth, Sabouraud dextrose
broth, fluid'thioglycolate broth, mycoplasma agar and broth, and by one
of the indirect methods to detect Mycoplasma hyorhinis (see this volume
[2]). Sediment the cells in a refrigerated centrifuge at 800-1000 g, discard
the supernatant, and resuspend in complete culture medium with 20% fetal
calf serum plus 10% sterile glycerol. Dispense the chilled cell suspension
into 1.2 ml borosilicate glass ampules with an automatic syringe. The
ampules should be previously marked with printer's ink and sterilized in
the dry air oven to insure a label that will not come off when stored in
liquid nitrogen. The ampules must be closed in an oxygen flame with a
pulled seal. To detect leaks, ampules are placed on aluminum canes m and
tested by immersion in a cylinder of alcohol stained with methylene blue
and chilled to 4. If a pinhole leak exists the contents of an ampule will be
stained blue and should be discarded. Sealed ampules should be kept at 4
and frozen as soon as possible. Significant loss of viability of WI-38 cells
was observed when stored at 4 for more than 6 hr. t7
15 T. Puck, J. Exp. Med. 1114, 6t5 (1956).
m Available from McCracken & Sons, Inc., 636 N 13 Street, Philadelphia, Pennsylvania,
19123.
17 A. Greene, B. H. Athreya, H. B. Lehr, and L. L. Coriell, Cryobiology 6, 552 (1970).
[3]
33
Freezing
Place the sealed ampules on labeled canes and position in a controlled
cooling rate apparatus. Set the controls to achieve a cooling rate of 1-2
per minute. When the heat of fusion is released at about - 10 the cooling
rate should be increased briefly to exchange the heat of fusion and to
reestablish the cooling rate. When the temperature reaches -25 the cooling rate can be increased to 5-10 per minute. When the temperature of
the specimen reaches - 100 the ampules can be transferred quickly to a
liquid nitrogen refrigerator for storage in the vapor or in the liquid phase.
Success with cell cultures has been achieved by many different slow
freezing techniques. 1'17'18'19 When large numbers of ampules are frozen
conditions and cooling rate settings must be adjusted to provide the desired cooling rates. The optimal cooling rate and procedures should be
determined for each cell type. Mouse embryos, for example, survive
poorly if cooled faster than 1 per minute 9 whereas many fibroblast or
epithelial cell cultures tolerate a faster cooling rate TM. As a general guide to
the adequacy of the whole process, the trypan blue viability of the cell
culture after recovery from liquid nitrogen storage should not be more
than 5% below the viability determined before storage. If it consistently
exceeds 10% loss of viability every step in the process should be critically
evaluated.
Storage
Permanent storage should be in liquid nitrogen whether liquid or vapor
phase. This will ensure a temperature well below -150 and prevent ice
crystal growth and enzyme activity. A few investigators have described
changes in cells stored in liquid nitrogen, 2-22 but most investigators have
not been able to observe detectable loss or gain of properties when cells
are properly stored in it. Twenty-two cell cultures stored in liquid nitrogen
in our laboratory have been recovered periodically and observed for viability as determined by trypan blue exclusion, plating efficiency, and cell
yield in milk dilution bottles and roller tubes after 7 days of incubation.23
No significant change in these parameters has been observed after storage
in liquid nitrogen for up to 12 years. 23'24
Is V. Perry, C. Kraemer, and J. L. Martin, Tissue Cult. Assoc. Man. 1, 119 (1975).
19 R. Moklebust, N. Diez, and I. Goetz, Tissue Cult. Assoc. Man. 3, 671 (1977).
2o W. P. Peterson and C. Stulberg, Cryobiology 1, 80 (1965).
21 H. T. Meryman, "Cryobiology," p. 65. Academic Press, New York, 1966.
22 L. Berman, M. P. McLeod, and E. P. Powsner, Lab. Invest. 14, 231 (1965).
23 A. E. Greene, B. H. Athreya, H. B. Lehr, and L. L. Coriell, Proc. Soc. Exp. Biol. Med.
124, 1302 (1967).
24 A. E. Greene, M. Manduka, and L. L. Coriell, Cryobiology 12, 583 (1975).
34
BASIC METHODS
[3]
Shipment
In design of a standard procedure for shipment of cell cultures consideration must be given to (1) safe delivery of viable cells, (2) cost, and (3)
regulations imposed by government and carders.
Regulations
Most cell cultures are free of microbial contaminants and etiologic
agents and therefore can be shipped without meeting any special stan-
[3]
PRESERVATION,
35
BIOMEDICAL
MATERIAL
I
NOTICE TO CARRIER
This package coatalm LESS THAN 50 ml OF AN ETIOLOGIC AGENT, N.O.S., is
packaged and labeled in accordance with the U.S. Public Health Service Interstate
Quarantine Regulations (42 CFR, Section 72.25 (c) (1) and (4)). and MEETS ALL
REQUIREMENTSFOR SHIPMENT BY MAIL AND ON PASSENGERAIRCRAFT.
This shipment is EXEMPTED FROM ATA RESTRICTED ARTICLES TARIFF
6-D (see General Requirements 388 ( d ( l ) ) and from DOT HAZARDOUS MATERIALS REGULATIONS (see 49 CFR, Section I73.386(d)(3)). SHIPPER'S
CERTIFICATES, SHIPPING PAPERS, AND OTHER DOCUMENTATION OR
LABELING ARE NOT REQUIRED.
Date
Signature o f Shipper
Institute f o r Medical Research
Copewood Street
Address
Camden, New Jersey 08103
dards as to volume, labels, and packaging except that they arrive safely
and in an undamaged condition. However, some cell cultures may be
contaminated and some do contain etiologic agents, e.g., lymphocyte cell
lines transformed with Epstein-Barr virus. If known etiologic agents are in
the cell culture a package and labeling procedure for shipment of cell
cultures that will meet all requirements for etiologic agents should be
followed. 25 The packaging requirements for etiologic agents are as
follows:
T h e y shall be placed in securely closed, watertight primary receptacles which shall be
enclosed in a durable, watertight s e c o n d a r y packaging. Several primary receptacles m a y be
enclosed in a single s e c o n d a r y packaging, providing that the total v o l u m e of all the primary
receptacles so enclosed does not e x c e e d 50 ml. The space at the top, b o t t o m and sides
b e t w e e n the primary receptacles and s e c o n d a r y packagings shall contain sufficient nonparticulate absorbent material s u c h as cotton wool, to absorb completely the contents of the
~5 U. S. Public Health Service, " I n t e r s t a t e Quarantine Regulations" (42 CFR, Sect. 72.25).
U S P H S , W a s h i n g t o n , D.C., 1972.
36
BASIC
METHODS
[4]
primary receptacle in case of breakage or leakage. Each set of primary receptacle and
secondary packaging shall then be enclosed in an outer packaging constructed of corrugated
fiberboard, wood, or other material of equivalent strength. The maximum amount of
etiologic agents that may be carried in any one package is 50 milliliters.
Packages containing an etiologic agent must resist breaking or leakage of
the contents including: (1) a water spray test for 30 min, (2) a free drop test
through a distance of 30 feet, and (3) a puncture test of a 3.2-cm steel
cylinder weighing 7 kg when dropped 1 m onto the exposed surface of the
package. The outside of the package should display the two labels shown
in Fig. 1 if it contains etiologic agents.
Packaging Recommendations
The T25 plastic flask is filled to the top with complete culture medium,
and the screw cap is tightened firmly and anchored in place with masking
tape. The flask is placed in a liquid-tight polyethylene envelope # 101. 26
The loose space around the flask is filled with absorbent cotton, and this
envelope is placed in a larger liquid-tight polyethylene envelope #302 and
sealed; this is then placed in a tongue and grooved Styrofoam utility
mailer #3731-3227 and sealed with Fiberglas tape. The outer c a n o n is of
heavy corrugated cardboard sealed with vinyl sealing tape. The completed package, ready for mailing, weighs approximately 5 ounces and has
sufficient insulation to withstand subzero temperatures without freezing
the contents.
26 Briton liquid tight bags distributed by Medical Associates Int., Inc., P.O. Box 123, Topeka, Kansas 66601.
27Utility Mailer #3731-32 obtained from Cole Plamer, 7425 Noah Park Ave., Chicago,
Illinois 60648.
[4] S a f e t y C o n s i d e r a t i o n s
By W. EMMETT BARKLEY
The basic tenet of safety in research that involves potentially hazardous organisms is strict adherence to good laboratory practice. This tenet
demands an awareness of the possible risks associated with the research
materials that are handled, knowledge of mechanisms by which exposure
may occur, use of procedures and techniques that reduce the potential for
exposure, and continuous vigilance to guard against compromise and error. The cell culture w o r k e r is not unfamiliar with these basic principles,
although the primary intent o f their application has been the protection of
METHODS IN E N Z Y M O ~ Y ,
VOL. LVIII
[4]
SAFETY CONSIDERATIONS
37
the cell culture rather than the cell culture investigator. Indeed, the experienced cell culture worker is aware that cell cultures are susceptible to
contamination; knows the potential sources of contamination and the
means by which contaminants can be introduced into cell cultures; is
proficient in the use of procedures and techniques for preventing cell
culture contamination; and well understands that a relaxation in good
laboratory practice often results in cell culture contamination. Fortunately, the methods of the successful investigator are also applicable to
protecting the individual from potential hazards that may be associated
with research involving cell cultures.
Biohazards and Cell Cultures
Cell cultures represent a potential biohazard because of their capacity
to infect the laboratory worker with unrecognized viruses. The
significance of this potential biohazard is emphasized by the few but serious cases of laboratory-acquired infections associated with the preparation and handling of primary monkey cell cultures. The most notable
examples involve infections with Herpesvirus simiae (B virus) and Marburg virus. 1
No laboratory-acquired infections have been associated with the use
of continuous cell culture lines that are assumed to be free of infectious
virus. These cell lines, however, should not be considered free from
hazard since they may harbor latent viruses. For example, transformed
cell lines may spontaneously produce viruses with oncogenic potential in
animals, and human lymphoid cell lines may harbour Epstein-Barr
virus.2,3 Lymphoid cell lines may also represent a unique potential hazard
if they are obtained from persons who plan to maintain them in culture. In
the event of accidental ingestion or injection, these cells may not be
rejected since they would possess common histocompatibility antigens. If
the cells were transformed in culture, they may have the capacity to
create transformed foci in the cell culture worker which may subsequently progress to true malignancy)
Cell cultures used in virological studies should be assessed to present
the same degree of hazard as the infectious virus under study. In such
studies, actual hazards can be assessed easily and appropriate safeguards
R. N. Hull, in "Biohazards in Biological Research" (A. Heilman, M. N. Oxman, and R.
Pollack, eds.), p. 3. Cold Spring Harbor Lab., Cold Spring Harbor, New York, 1973.
2 G. J. Todaro, in "Biohazards in Biological Research" (A. Hellman, M. N. Oxman, and R.
Pollack, eds.), p. 114. Cold Spring Harbor Lab., Cold Spring Harbor, New York, 1973.
3 j. A. Schneider, in "Biohazards in Biological Research" (A. Hellman, M. N. Oxman, and
R. Pollack, eds.), p. 191. Cold Spring Harbor Lab., Cold Spring Harbor, New York, 1973.
38
BASIC METHODS
[4]
[4]
SAFETY CONSIDERATIONS
39
tial for creating aerosols. There are a variety of pipetting aids available
that range from simple bulb- and piston-actuated systems to more sophisticated devices which contain their own vacuum pumps. In selecting a
pipetting aid, consideration should be given to the kind of pipetting procedure that is to be performed, the ease of operation, and the accuracy of
delivery that is needed. It is likely that several pipetting aids will be
necessary; no universal aid is available that is appropriate for all pipetting
procedures.
Pipetting technique that reduces the potential for creating aerosols
should be developed. Pipettes should be plugged with cotton. Rapid mixing of liquids by alternate suction and expulsion through a pipette should
be avoided. No material should be forcibly expelled out of a pipette, and
air should not be bubbled through liquids with a pipette. Pipettes that do
not require expulsion of the last drop are preferable to other types. Care
should be taken to avoid accidentally dropping cultures from the pipette.
When pipetting infectious materials, place a disinfectant-soaked towel on
the working surface. Liquids from pipettes should be discharged as close
as possible to the fluid or agar level of the receiving vessel, or the contents
should be allowed to run down the wall of the receiving vessel. Dropping
of the contents from a height into vessels should be avoided. Contaminated pipettes should be carefully placed in a pan containing enough suitable disinfectant to allow complete immersion.
Hygienic Practice. The cell culture worker should avoid eating, drinking, and smoking in the laboratory. Hands should be washed after handling cell cultures and before leaving the cell culture laboratory. To avoid
contact contamination, the investigator should develop the habit of keeping hands away from mouth, nose, eyes, and face. Laboratory gowns or
other suitable clothing should be worn when handling cell cultures; the
same clothing should not be worn to eating areas or outside of the
laboratory,
Decontamination. Work surfaces should be disinfected with a suitable
chemical disinfectant following the completion of procedures involving
cell cultures. Cell culture wastes which may be contaminated with virus
should be disinfected with a suitable chemical disinfectant or sterilized by
autoclaving before disposal. Contaminated items such as glassware and
laboratory equipment should be disinfected or autoclaved before washing,
reuse, or disposal. Contaminated materials that are to be autoclaved or
incinerated at a site away from the laboratory should be placed in a
durable, leak-proof container that is closed before removal from the
laboratory.
Control of Aerosols. Most techniques used in a cell culture laboratory
have the capacity to create aerosols. Therefore careful attention should be
40
BASIC METHODS
[4]
given to measures that can reduce the potential for inhalation exposures
to aerosols that are produced by laboratory operations. Although the
extent of aerosol production can often be controlled by good technique,
efforts to eliminate exposure require the use of containment equipment.
Such special containment equipment for use in preventing the creation
of aerosols is available. For example, capped safety centrifuge cups and
sealed centrifuge heads can prevent the production of aerosols while centrifuging.
The most important measure for controlling aerosols is the use of
ventilated safety cabinets for containment of operations that produce considerable aerosols. The cell culture worker is encouraged to use this containment equipment for operations that involve grinding, blending,
homogenizing, and resuspending tissues or cell cultures. This should be
standard practice if the cell culture materials are suspected of being contaminated with pathogenic microorganisms or viruses. Specific recommendations for the effective use of this equipment are presented in the
following section.
[4]
SAFETY CONSIDERATIONS
41
dard for Class II cabinets. 1~ The selection of a Class II cabinet that has
been certified by NSF provides an assurance of performance capability.
The performance capability, however, should be validated by measuring
airflow and filter efficiency once the cabinet has been installed in the
laboratory.
Cabinets should be located in an area of the laboratory away from
doorways, supply air diffusers, and spaces of high activity. This is done to
reduce the adverse effects of room air currents that could compromise the
operation of the cabinet. Generally, the best location for the cabinet is
along a side wall at a position farthest from the laboratory door.
The operational practices listed below are recommended when using
laminar flow safety cabinets. Their use will provide excellent protection
for cell cultures against contamination. These practices are most important, however, when the cell cultures are experimentally infected or contaminated with viruses.
1. Cabinet users should be encouraged to wear long-sleeved gowns
with knit cuffs and gloves. This reduces the shedding of skin flora into the
work area of the cabinet and protects the hands and arms from contact
contamination.
2. The work surface of the cabinet should be decontaminated with a
chemical disinfectant before placing equipment and material into the
cabinet.
3. The cabinet should not be overloaded. Ideally, everything needed
for the complete cell culture procedure should be placed in the cabinet
before starting work so that nothing passes in or out through the air
barrier until the procedure is completed.
4. Nothing should be placed over the exhaust grills located at the front
and rear of the work surface. This practice will help maintain uniform
airflow within the cabinet.
5. The arrangement of equipment and material on the work surface is
an important consideration. Contaminated items should be segregated
from clean ones and should be located so that they never have to be
passed over clean items. Discard trays should be located to the rear of the
cabinet.
6. After all equipment and material has been placed in the cabinet, the
cabinet user should wait a few minutes before beginning work. This time
period allows the cabinet air to remove any contaminants that may have
been introduced into the cabinet during the loading process.
7. Always use aseptic techniques when performing cell culture work
within the cabinet. This is important in preventing contact contamination.
13 "National Sanitation Foundation Standard No. 49 for Class II Biohazard Cabinetry."
Natl. Sanit. Found., Ann Arbor, Michigan, 1976.
42
BASIC METHODS
[4]
[4]
SAFETY CONSIDERATIONS
43
mal Viruses and Cells. ''15 These guidelines recommend that good laboratory practice be complemented by special facility safeguards for certain
cell culture work. The guidelines require that cell culture work involving
(1) the co-cultivation or fusion of human cells with cells of nonhuman
mammals and (2) short-term nonhuman primate cells be confined to segregated rooms or cubicles that have exhaust air ventilation. The exhaust air
from these laboratories is to be discharged outdoors. However, recirculation of air is allowed providing that the air is treated by filtration with
HEPA filters. These requirements have been established to prevent the
dissemination of cells and viruses, which the exhaust air may contain, to
uncontrolled areas of. the laboratory facility in the event that they are
accidentally released within the segregated laboratory area.
The facility precautions are based on a possibility of hazard that is
considered to be greater than that associated with most cell culture work.
For co-cultivation and fusion studies with human cell lines, the principal
concern is the possibility of unmasking a virus with oncogenic potential
for man. The hazard concern for short-term nonhuman primate cells is the
evidence that these cells may contain Herpesvirus simiae (B virus).
44
BASIC
[5]
Media
By RICHARD G.
METHODS
and Growth
[5]
Requirements
Introduction
This brief review o f culture media and their applications is addressed
primarily to the use of cell culture as an experimental tool in fields of
r e s e a r c h other than analysis of cellular growth requirements. Our emphasis is on the t y p e s of m e d i a available, and on principles involved in selection of the m e d i u m that is best suited for the e x p e r i m e n t that is to be done.
Environmental requirements for cellular multiplication and theoretical aspects of m e d i u m d e v e l o p m e n t are reviewed in greater detail elsewhere. 1-4
T y p e s of Cultured Cells and Culture Systems
The choice of which culture m e d i u m to use and what supplement (if
any) to add to it is strongly influenced by the type of cell that is to be
grown and by the w a y that cell is to be grown. Thus, it will be helpful to
begin with a brief outline of the types of cultured cells and culture s y s t e m s
that can be e m p l o y e d in various kinds of research.
Classification of Cells
The following criteria are often e m p l o y e d to describe cells:
[5]
45
46
BASIC METHODS
[S]
the term "defined," since in most cases growth in such media appears to
be dependent on trace impurities such as inorganic trace elements that are
not included in their formulas, as will be discussed in greater detail
below.)
The growth requirements of normal cells and nontransformed lines are
not as well characterized, and at the present time, it is necessary to use
poorly characterized supplements of serum proteins or other biological
materials to obtain satisfactory multiplication of such cells. The data are
still incomplete, but it currently appears highly probable that most such
cells have specific requirements for protein growth factors that cannot
easily be replaced by improvements in the low-molecular-weight portion
of the culture medium or in other aspects of the culture system (this
volume [6]).
Types of Culture Systems
The major distinctions among culture systems are based on cellular
population density and on whether the cells are grown attached to a
substrate or in suspension. When the cell density is so low that the descendants of single cells form discrete colonies (clones), the procedures
arc described as "clonal." The term "cloning" should be used only for
procedures in which a new culture is established from the progeny of a
single cell. All other experiments involving formation of colonies from
single cells are referred to as "clonal growth." Experiments involving
cloning or clonal growth are normally done either with the cells attached
to a solid substrate under a liquid medium, or else with the cells suspended in a semisolid medium, e.g., in soft agar.
Dense cultures of cells attached to a substrate arc often called
"monolayer" cultures. Cellular yield in monolayer cultures is limited by
the surface available on which cells can grow. This can be increased by
use of roller bottles, which are rotated in such a way that their entire inner
surfaces are bathed with medium (this volume [16] and [17]), or by use of
capillary perfusion systems (this volume [14]) in which medium is circulated through tightly packed artificial capillaries, or by use of suspensions
of particulate microcarriers on which the cells can attach and multiply
(this volume [15] and [16]).
True suspension cultures, in which the cells are not attached to a
substrate, are generally done with continually stirred or shaken culture
vessels of various sizes, ranging from small spinner flasks with magnetic
stirrers to huge industrial-scale fermentors (this volume [16] and [17]).
Suspension cultures offer the special advantages of case of handling of
large numbers of cells and ability to harvest cells without the use of
[5]
47
enzymes. For certain types of cells that do not readily attach to each other
or to substrates, suspension cultures without agitation are also possible.
Both the cellular population density and whether the cells are attached or
suspended have significant effects on cellular growth requirements, as is
discussed below.
Cellular Population Density. Early in the history of cell culture, it was
observed that large populations of cells would often multiply under conditions where single isolated cells would not. Sanford and co-workers 5 isolated single cells in capillary tubes with small volumes of medium and
demonstrated that the critical variable was the volume of medium per cell
and that growth was due to "conditioning" of the medium. Subsequently,
Puck and Marcus 6 used a feeder layer of nonmultiplying heavily irradiated
cells to condition the medium in a petri dish so that it would support
multiplication of single isolated nonirradiated cells to form macroscopically visible colonies. (See this volume [21]).
At least three separate mechanisms are involved in "conditioning" of
culture media: (1) Inhibitory materials that bind to the cells are neutralized without reaching a dose per cell that is large enough to block
growth. (2) Low-molecular-weight metabolic intermediates that diffuse
readily out of cells accumulate in the extracellular medium in amounts
which, by equilibration, maintain adequate intracellular levels for biosynthesis and metabolism. Such substances, which must be for biosynthesis
and metabolism. Such substances, which must be supplied from an
exogenous source when the volume of medium per cell is too large for
effective conditioning, are referred to as "population-dependent" growth
requirements. The most common examples are carbon dioxide, pyruvate,
and the so-called "nonessential" amino acids, particularly serine. (3)
Macromolecules synthesized by the cells sometimes must build up to a
critical level before growth will occur. In addition to cases where a particular type of cell produces the macromolecules needed for its own multiplication, there are numerous cases in which macromolecules released
by one type of cell are beneficial for another.
Density-Dependent Inhibition. Most types of cells stop or slow their
multiplication at a characteristic population density in any particular
medium. The effect tends to be more pronounced with nontransformed
cells than with those that have undergone a malignant transformation. The
exact mechanisms involved are not fully worked out and remain quite
controversial. However, it is likely that for most types of cells, some
combination of cell contact, localized or generalized depletion of nutrients
K. K. Sanford, W. R. Earle, and G. D. Likely, J. Natl. Cancer Inst. 9, 229 (1948).
6 T. T. Puck and P. I. Marcus, Proc. Natl Acad. Sci. U.S.A. 41, 432 (1955).
48
BASIC METHODS
[5]
[5]
49
50
BASIC M E T H O D S
[5]
[5]
51
gently to suspend the cells. Break up clumps by gentle pipetting and dilute
the cells as needed for subculture. If a hemocytometer count is made for
clonal growth, the suspended cells should be kept on ice during that
procedure. The cells are not warmed above ice-bath temperature until
they are placed in their final culture medium and returned to the cell
culture incubator. As long as they are maintained in the cold, the cells do
not round up, but rather remain elongated and irregular in shape. All
mechanical procedures involving the cells are done as gently as possible.
For other types of cells, and for cells with different culture histories, it
is necessary to vary the amount of trypsin and time of exposure. For
example, for human-foreskin fibroblasts, previously grown under the
same conditions as the human fetal lung cells described above, about 500
/zg/ml crystalline trypsin will be needed, and the digestion time must be
increased to 15 min.
Cellular Growth Requirements
Many different variables combine to determine whether or not cells
multiply in vitro. We have already considered the fact that different types
of cells have different growth requirements, and the fact that many aspects of the culture system that have nothing directly to do with medium
composition affect growth requirements, including cellular population density, substrate attachment or its absence, the nature of the substrate, and
the way the cells are handled during subculturing. In this section, we will
consider in sequence the following aspects of cellular growth
requirements:
1. The medium must supply all essential nutrients. These include all
raw materials needed for the synthesis of new cells, substrates for energy
metabolism, vitamins and trace minerals whose function is primarily catalytic, and bulk inorganic ions whose functions are both catalytic and
physiological.
2. Physiological parameters, such as temperature, pH, osmolality, and
redox potential, must be kept within acceptable limits.
3. The culture system must be free from toxic or inhibitory effects,
including those due to excess amounts of essential components.
4. Precise quantitative adjustments, including balance relationships
among the components of the culture system, are very important.
5. Serum, which is frequently added to defined basal media to stimulate multiplication, interacts with virtually every other variable in the
culture system, and it also serves as a source of macromolecular growth
factors that are essential for multiplication of many, but not all, types of
cells in currently available basal media.
52
BASIC M E T H O D S
[S]
[5]
53
54
BASIC METHODS
[5]
[5]
55
phatidylcholine (lecithin) suspensions 14 may provide a soluble and relatively nontoxic source of lipids for cells grown in adqueous media.15,16
Human diploid fibroblasts and chicken embryo fibroblasts both appear to
have definite requirements for lipids.l'2
Putrescine or other polyamines are required for growth of Chinese
hamster ovary cells in protein-free media and are beneficial to certain
other cells. Many types of cultured cells exhibit a population-dependent
requirement for pyruvate or other small o~-oxo acids. The requirement is
relatively nonspecific and may reflect the need for a substrate that can be
used to oxidize NADH, rather than the need for a specific metabolic
intermediate. A variety of other organic compounds have been incorporated into some of the more complex media on the basis that they might be
beneficial, and it probably does no harm to include them. However, this
philosophy can be a serious barrier to the development of serum-free
media since substances that do not appear to be harmful in the presence of
large amounts of serum sometimes prove to be quite inhibitory in media
with lower concentrations of protein.
Physiological Requirements
In addition to satisfying nutrient requirements, the culture environment must have physicochemical properties that fall within physiologically acceptable limits for cellular survival and multiplication. Important
parameters include the following:
Temperature. Cellular multiplication rate typically increases with temperature until a limiting value is reached, above which further increases in
temperature rapidly become inhibitory. Most laboratories do not have
enough incubators available to perform properly controlled experiments
on the effects of temperature on cellular multiplication, and except for
studies involving temperature-sensitive mutations of cells or viruses, little
attention is usually given to this variable. However, different species of
warm-blooded animals do have different body temperatures, and certain
tissues and organs such as skin and testes are normally maintained at
temperatures below the overall body temperature. Therefore, studies of
temperature optima could prove to be very profitable in certain cases.
Typically, cultures of cells from warm-blooded animals are grown at 37
and cultures from cold-blooded animals at temperatures near the upper
limit of optimum body temperature for the intact animal.
24 G. Poste, D. Papahadjopoulos, and W. J. Vail, Methods Cell Biol. 14, 33 (1976).
15 N. N. Iscove and F. Melchers, J. Exp. Med. 147, 923 (1978).
le W. L. McKeehan and R. G. Ham, Abstracts 1978 TCA Meeting, In Vitro 14, 353 (1978).
56
BASIC M E T H O D S
o_
_ r,-~
- ~ ~
"o
~
[S]
~o
_~
.'~
~,~o.,o~.~-~
~'~"~ ~ 2 , ~
~ ~
"
~ ~ - ~ .~'~
~~ ~.~ B~
,,
~.~
I
m
ZZr~
J
~z
Z
t-
.I
0
~,
~
~
~":
.~
,~
g
S
~-.
_~i
.~-~
.~
~.0
~r.~
i~
[5]
MEDIA
AND
GROWTH
REQUIREMENTS
.~
.~'=o
"~-
=,.,
57
.~
~- ~
~..
.=_
4-
-F
,,~
.~ ~
~
~ ~
.
..~ ~ ~
~.-~
4-
'~
"5
b~
4-
,..1 . l
F-
~-
.1
ZZ
.=
E
E
[,,-,
F-
["-
.4
=..~
's
.=
",~
~-
58
BASIC
METHODS
[S]
~.
o_~i ~ ~ ~
-
~-~
~
~.~
.o ~,
~o.~
.~oo i . ~
...~
~ '~
~ ' ~ .~
x:
~ r~
~o~
.~
+
6t~
. ~
6
t~
mt
t~
6
Z
a~
[5]
MEDIA
.'
AND
.'
GROWTH
"~
~'
'~
E
.=.~
59
REQUIREMENTS
,a
"~
"El "=
.~
.~
.=
=
._=
.=_ ~
~a
~
e
=~
,~
.-
....
.=
._=
=
_
~.=
~=-~
g
o'=
.~,.= ~
).~-~
~:~
++l
.) . 1 Z
<
~=
~:'a
~.=~.~
= ~'~'~
I+
,.az
~ - ~
.=_
.=_
m
r,.
E.= ~ ~.~-
..=
E
..~
~ ; ~ ~.~'~'~
r~L)
.)
L)
L)
x.t
.~-
. ~. ~ ~,=~. ~
BASIC METHODS
60
.~
-~~
.:
~:
.~
o '~-
.~ ~ - ~
~.,~~-.-~ , .~~, , ~
~'0~
=~-.=
. a . ~
~ r ~
~,~,~
~ ~..~
~ 4~d,<~
~,,~
~.~.e~-~_~a'~z=
~ ' 4 ~
.~
..~.~
[S]
.~
4~
",4~,,-;~
~:~ ~
",~,4-q
.-
~.~ g~"
=,,
o~
"
[5]
&
f~zo~
~ =
.....
~o
~ ~
~-~
II
'i
2~
"==g=
~'~
o
-~ > > -
~
=
61
62
BASIC METHODS
[S]
oo
~
m
6
o o o o o ~ o ~ o ~
t-
in
m
m
0
0
0
~ o o o o o o o o o ~ o o
[5]
MEDIA
AND
GROWTH
63
REQUIREMENTS
;<
,z= .-
II
~111111
I I
IIIIIII
I I
I I il
~.~
ii
~lll~li
I I I
itlitiliili
.r-
o
~
o o o o o o
.<
],<
I I I
~ ~
I ~
I~
I ~ ~
I I
64
BASIC METHODS
~<
[5]
IIl,lllll::~lllllll,llllllll
tttltt
tii
t~t~titttlttltlttttt
i i ~t~l~t
it~ttt~
i ~ iii
i i iiIi
ii
t~ltti~ttttitltttt
tltu~,l
I,Illl,ll,l~
~,~,~
o(
u~
it
. . . .
Illll
I III
I I I II~l
~4
I ~ l t ~ t ~ ~
~ ~ = = ~
IS]
MEDIA
AND
GROWTH
REQUIREMENTS
65
~~
~o~
.e
~eg
.N
o.
~q
m
~
I~
Illllll~l~
rqoi
I~11
~,~
IIIIIIIIIIIIIIII
Im~m~m~m
~
. . . . . . .
Illllllllllll
II I
IIIllllllllll
II I
IIIIIll~lllllllllllllllllll
"0
66
BASIC METHODS
[S]
r~
III
c~
IIIIII
IIIIIIIIII
IIIIII
e~
I III
II Ill
llll~illlllll
II
III
lllIlllllllll
I II~l
II III
III
I~IIIIII~II
I IIII
IIIIIIIII
llllll~Itlllllillllllrltli
IrllII~II~IIIIIIII~II1~tl
r--
~S
,~
~ o
....
~.~'~
~'
~ ~ .
[5]
67
Illlllll
e4
e,i ,.-:
I I
II
~l
iI
IIIIIIif
II
II
IIIIIIPI
r,i
~1
r4
I I I
III
III
~1
II
III
eq
~ o 4
r I I
~1
II
z~
II
IIII
I I
Ir
lit
68
BASIC METHODS
I-~o
[S]
. . . . . . . .
t ~
II~lll~lllllllll~llllllllll
I I I I I I I I I I I I I I t I I~ t I ~ t I I I r I
6
I I I I~
I I I I I I I I I~
I I I I I
I~
Illlllllllllllllll~llllllll
-.I
",
'~
II,lll
Jl
llll,lll
llllt,ll
[5]
69
q
b~
III
>- I"O
III
II
II
eq
-6
-r"
III
II
I II
IIII
III
III
I I
III
I~111
I I
[.h
r-O
L)
o~
o
z
L~
IIIIIIIIII1~1111
70
BASIC METHODS
I I
I I
II
II
[S]
cD
-:
-:1
II
I
m
,..r.
I I
IM
I~
[5]
~:
:..~
"~
=o
~.~
.~ . .~ . ~
.~
:~
o.
o_
. ~
.-
o.o~
71
=o
!ill
ilil
il!iiiii
ii
~=
.-
.~_
~.
72
BASIC METHODS
IS]
[5]
73
to adjust the pH at room temperature and in air so that it will shift to the
correct value in the incubator. It is unnecessary to include bicarbonate in
the formulation of a medium whose pH is adjusted in this manner, since
bicarbonate is formed from dissolved carbon dioxide as the buffered
medium equilibrates in the incubator. One other potential area of concern
is possible inhibitory effects of large quantities of artificial buffers. In the
case of HEPES, there does not appear to be a problem up to at least 50
mM, provided that appropriate osmotic corrections are made.
Osmolality. The optimum range of osmotic pressures for cellular
growth is quite narrow and varies with the type of cell and the species.
For human fetal lung fibroblasts, optimum clonal growth occurs between
about 250 and 325 mosmol/kg, with some growth from about 200-375
mosmol/kg. For chicken embryo fibroblasts, the optimum is about 275325 mosmol/kg, and the range of suboptimum growth is about the same as
for the human cells. Cells from amphibia require significantly lower osmolality than avian or mammalian cells, and this is often achieved by
diluting standard culture media with distilled water. Cells from parts of
the body such as the kidneys that are exposed to differing osmolalities
may also have different requirements.
The narrowness of the optimum range of osmolality makes it necessary
to adjust the concentration of sodium chloride when major additions are
made to a culture medium, such as the use of 30-50 mM HEPES buffer (plus
enough NaOH to bring it to a physiological pH). Osmotic considerations
may also be important when media are used with high concentrations of
carbon dioxide. With an atmosphere of 10% carbon dioxide and a pH of
7.4, the combined concentrations of carbonic acid and bicarbonate ion
generated by carbon dioxide dissolving in a buffered medium add up to
about 30 mM. (See also this volume [16] for a discussion of osmolarity.)
Humidification of Incubators. Since rapid equilibration between the
culture medium and the gaseous carbon dioxide-air mixture in the culture
chamber is frequently important, culture dishes usually cannot be sealed
tightly. The narrow limit of acceptable osmolality requires that essentially
all evaporation be prevented during the time that the dishes are in the
incubator, which may be 2 weeks or longer for some clonal growth experiments. Limitation of evaporation is accomplished by maintaining the
relative humidity very close to saturation in the incubation chamber. This
can be a difficult task in a dry climate. One of the specific requirements is
that the water that is used for humidification be kept at the same temperature as the rest of the incubator. Each time that the incubator door is
opened, dry air enters and cools the water by evaporation. If there is
inadequate heat transfer, the water may remain cool for a prolonged
period and fail to humidify the incubation chamber adequately to prevent
evaporation and maintain the osmolality of the culture medium. Similarly,
74
BASIC METHODS
[S]
if there is a cool spot anywhere in the incubator, such as a poorly insulated door, it will serve as a site of condensation and prevent the partial
pressure of water vapor from reaching the needed level. To avoid these
problems, there must not be any surface in the incubator, including that of
the water used for humidification, that is more than a few tenths of a
degree centrigrade below the temperature of the medium in the culture
dishes.
One of the best ways to monitor humidification is to measure the
osmolality of control dishes of medium at the beginning and the end of the
incubation period for the experiments being performed. If there is a
significant increase in osmolality, the humidity is not high enough and
evaporation is occurring. Excessive humidification that leads to condensation on the culture dishes is also undesirable, since it increases the risk of
microbial contamination.
Oxygen Tension. A minimal amount of oxygen is essential for the multiplication of most types of cells in culture. However, the partial pressure
of oxygen in normal body fluids is significantly less than that of air. Systematic studies of the effects of oxygen tension on cultured cells are
technically difficult due to diffusion gradients, leakage, and release of
dissolved oxygen from plastic culture vessels. However, there are numerous reports that growth of cultured cells can be improved by reducing the
percentage of oxygen in the gaseous phase to between 1 and 10%. The
trace element, selenium, is a component of the enzyme glutathione peroxidase, which destroys metabolically generated peroxides. The inhibitory
effects of excess oxygen are much more severe when cells are marginally
deficient in selenium than when adequate amounts are supplied.
There have been a number of attempts to correlate cellular growth
with redox potential measured by inserting electrodes into the culture
medium, but the results are difficult to interpret because a cell growth
medium is not an equilibrium system. In fact, for cellular growth to occur
at all, the cell must be able to catalyze an energy-yielding reaction among
components of the culture system (normally glucose and oxygen). Cysteine and various other reducing agents are present in large amounts in
some of the older culture media, but there is little evidence that the
reducing conditions that they were intended to generate are actually
needed, and in at least some cases, very high concentrations of cysteine
are clearly inhibitory.
[5]
75
However, there are many subtle types of toxic and inhibitory effects that
are not immediately obvious. A sufficiently large excess of any component
of the culture system is inhibitory, even if only for osmotic reasons, and
many essential nutrients become inhibitory at surprisingly low levels.
Contaminants in the chemicals or water used in medium preparation can
be toxic. Membrane-type sterilizing filters may introduce substantial
amounts of detergents into culture media. Because of this, detergent-free
filters should always be specified. Short-wavelength visible light interacts
with certain components of culture media (riboflavin, tyrosine, tryptophan) to generate toxic photoproducts. ~7 Therefore, it is necessary to
keep exposure of media, to ordinary laboratory fluorescent lights at a
minimum. Toxicity can sometimes be generated by interactions among
components of the culture medium that are not individually toxic. Thus,
for example, bovine serum cannot be used in media that contain spermine
or spermidine because its high content of spermine oxidase will generate
toxic oxidation products from these polyamines. Putrescine is safe to use
with bovine serum, however, as it is not affected by the enzyme. In
certain cases, inhibitory contaminants may alter responses to other components of the culture system. For example, the presence of traces of
cadmium may increase substantially the amount of zinc needed for optimal multiplication. Similarly, in certain cases, a substantial portion of the
requirement for serum protein may be to overcome inhibitory effects.
Quantitative Requirements
The full significance of quantitative requirements for cellular multiplication is just beginning to be realized. For classically studied permanent
cell lines, precise quantitative adjustments do not seem to be very important, although there is some evidence that balance relationships affect
growth. TM For normal cells that are now receiving increasing study, however, such adjustments are proving to be critically important.l'2.9
For every required component in a culture medium, a three-part
growth response curve, similar in principle to the idealized example
shown in Fig. 1, can be obtained. In the first region on the left, multiplication is roughly proportional to the amount of the component added. In the
second region, in the center, the growth response is "saturated," and the
response curve exhibits a "plateau," within which growth is virtually
independent of concentration of the component. Finally, on the right,
above a critical threshold, the component becomes inhibitory and the
growth response is negative.
a7R. J. Wang,In Vitro 12, 19 (1976).
as R. G. Ham, In Vitro 10, 119 (1974).
76
BASIC METHODS
[5]
Plateau of
Optimum Growth
i-
Stimulation?
o
o.
\-
~lnhibition
Midpoint of Plateau
on Semilog Plot
is Selected as
"Optimum"
Concentration
o
o
/ . . . . . . . I , , , : , , i.d
,
10-2
i 0 -t
, , li,.,l
i0 o
* I
tl..I
i0 i
,~'~.,,I
......
I ,.
,
102
103
[5]
77
clonal growth of 26 of those 56 components (Table II). None of the "standard" media developed over the years for permanent established lines
will support clonal growth of these cells with comparably small amounts
of serum protein.
Another aspect of quantitative requirements that must be considered is the balance among interrelated components of the culture system.
For example, in addition to osmotic requirements and absolute requirements for sodium and potassium, the Na+/K + ratio appears to be important, with different optimum values for different types of cells. Another
set of balance relationships that appears to be very important occurs
among the amino acids.18 The ratios of amino acids in media optimized for
various normal and nontransformed cells differ significantly. (Compare
MCDB 105,202, and 401 in Table II.)
Serum and Its Relationship to Other Growth Requirements
The reason that serum is such an effective supplement for promoting
cellular multiplication appears to be that it contains a large number of
different growth-promoting activities in a physiologically balanced blend.
The multitude of different growth-promoting roles that serum is capable of
playing can perhaps best be appreciated in terms of the many different
ways in which the requirement for serum can be partially replaced. In a
more detailed review, we have listed and discussed 24 different ways in
which serum could conceivably contribute to cellular multiplication.2
Only a few of the more significant ways are described here.
Whole serum contains most of the low-molecular-weight nutrients
needed for cellular multiplication, and at one time the culture of permanent cell lines such as Mouse L and HeLa was routinely done in mixtures
of serum, embryo extract, and saline, with no defined organic nutrients
added to the medium other than glucose. Dialyzed serum retains many
small molecular nutrients in bound form, and it also possesses many
other growth-promoting activities. Large amounts of serum protein allow
cells to multiply with concentrations of essential nutrients that are inadequate for multiplication with smaller amounts of serum protein.
Dialyzed serum is also able to reduce the inhibitory effects both of contaminants and of essential nutrients that are present in excess amounts. In
some cases, serum macromolecules may buffer toxic nutrients by binding
them and releasing them in small amounts as their free concentration in
the medium is reduced by cellular metabolism. Serum protein neutralizes
trypsin and other proteases and also makes cells better able to overcome
the damage done to them by harsh subculturing procedures. Serum has an
undefined effect on the interaction between cells and their substrate,
78
BASIC METHODS
[S]
[5]
79
factors that have not yet been fully characterized chemically, such as
"platelet" factor and "multiplicaton stimulating activity" from conditioned medium. Gordon Sato's research group has recently reported
complete replacement of the serum requirements of several permanent
cell lines with mixtures of hormones and peptide growth factors z3 (see also
this volume [6]).
Some of the peptide growth factors, such as epidermal growth factor
and fibroblast growth factor, are beginning to be produced commercially.
Such factors will undoubtedly play increasingly important roles in the
study of cellular growth requirements during the next few years, as will
numerous factors that are currently under study, but not yet fully purified
or characterized. It currently seems reasonable to anticipate that growth
of certain normal diploid cells i n " defined" media supplemented only with
nanogram per milliliter quantities of purified macromolecular factors will
become possible within a few years.
80
BASIC METHODS
[5]
related to the experimental objective that first must be made include: (1)
what kind of cell to use; (2) what type of culture (monolayer, suspension,
clonal, etc.) to use; and (3) what degree of chemical definition is needed.
[5]
81
82
BASIC METHODS
[5]
[5]
83
84
BASIC METHODS
[S]
[5]
85
were designed for use with dialyzed serum, as well as a number of media
that support growth of permanent lines without serum supplementation.
The latter have been reviewed in detail by Higuchi 24 and by Katsuta and
Takaoka. z~
3. The third era (part G of Table I) has been under way for many
years, but is just now beginning to pick up momentum. It represents a
return to cells that are far more " n o r m a l " in their properties for the
measurement of growth responses. The classical permanent lines generally underwent extensive nutritional adaptation during the process of becoming established in culture, and media developed to satisfy their needs
generally do not support growth of normal cells without heavy serum
supplementation. Current research on cell growth requirements is focusing increasingly on normal cells and on nontransformed lines whose
growth requirements are more like those of normal cells. At the present
time, the growth requirements of normal cells are not yet fully understood, and the limited number of improved media for such cells that have
been described in the literature are just beginning to become available
commercially.
86
BASIC M E T H O D S
[5]
[S]
87
The most widely used of these is medium L-15 of Leibovitz, which substitutes galactose for glucose and contains high concentrations of the basic
amino acids. This formula has been used with serum for a variety of cells,
both permanent lines and diploid primary cultures. A second such
medium is the CMRL 1415-ATM modification of CMRL 1415, which was
designed for primary cultures of mouse embryo cells. It is also useful for a
variety of other types of cells, and growth can sometimes be improved by
reducing its unusually high concentration of cysteine.
Media for Normal Cells. As described above and in Table I, many of
the media developed originally for established lines are widely used with
serum for growth of normal diploid cells and permanent lines of nontransformed cells. In addition, there are a number of media that have been
developed specifically for such cells (parts A and G of Table I). With the
exception of Medium 199 and similar media from the era of development
that preceded the availability of permanent cell lines, all of these formulas
are modifications of previous media for permanent lines. However, some,
such as the MCDB series, represent complete reworkings of the formulas
to satisfy the needs of the nontransformed cells.
A number of the formulas listed in Table IG were developed primarily
for survival and maintenance of function of normal cells without serum
supplements, and will be discussed below under the defined medium heading. Dulbecco's DME and Richter's IMEM-ZO are both modifications of
original formulations by Eagle. As already discussed, DME is one of the
most widely used media for primary cultures of mouse and chicken cells.
IMEM-ZO has been reported to support normal rat and human cell
growth, including clonal growth with relatively low concentrations of
serum proteins. CMRL 1415 is used with serum for growth of mouse cells,
and CMRL 1969 for human and monkey cells. F12K is a further modification of Coon's modification of F12, and it has been reported to work well
for a number of types of differentiated rat cells.
The MCDB series of media is the result of a process of qualitative and
quantitative optimization for clonal growth with minimal amounts of
serum protein. In each case, they will support clonal growth of diploid
fibroblast-like cells from the species they were designed for (MCDB 104,
105, human; MCDB 202, chicken; MCDB 401, mouse; MCDB 501, duck)
with far less dialyzed serum than conventional media. The composition of
MCDB 202 is such that it will also work reasonably well with cells from a
variety of other species. These media are specifically recommended for
clonal growth of their respective species, although they will not necessarily work for all types of cells from a particular species. They also can be
used for monolayer growth with modest amounts of dialyzed serum. At
the time of this writing, only MCDB 104 is available commercially.
88
BASIC METHODS
[5]
[5]
89
lines in defined media. One such report is growth of the C6 glial tumor line
by Donta in medium 924 MAP/l, developed originally by Waymouth. A
second is the growth of monolayers of C1300 mouse neuroblastoma cells
in medium MCDB 411, which has been optimized qualitatively and quantitatively for growth of the C 1300 neuroblastoma, and will support multiplication without delay or apparent adaptation of cells previously grown
exclusively in serum-containing media. It can also be used without supplementation to initiate cultures directly from transplantable C1300
tumors that have not previously been grown in culture, again without
delay or the need for adaptation. Another set of media developed in the
same manner are F12 and its more recent replacement, MCDB 301, which
are discussed below under clonal growth, although they also support
monolayer growth of Chinese hamster ovary lines. F12 and MCDB 301
have an unusually high concentration of zinc, which should be reduced if
they are used for other types of cells, such as mouse L. 21
Defined Media for Suspension Culture. A number of completely synthetic media for suspension culture have been described in the literature,
including several designed specifically for high cellular yield, but none
have become widely used. Suspension cultures in defined media require
very close attention to details, and it appears that in most cases, the
addition of serum does not interfere sufficiently with experimental objectives to justify the use of defined media. Media that are available for
defined suspension cultures include minor modifications of NCTC 109 and
MB 752/1, and various special-purpose formulas (Table IE). Birch and Pirt
adjusted concentrations of nutrients for efficient utilization and high yields
in suspension cultures of LS cells. Ling et al. used unusually high concentrations of nutrients to develop a medium (7C's) that would yield a maximum number of cells. Nagle and Brown developed a suspension culture
medium that is free of glutamine, autoclavable, and effective for cells from
many different species. However, none are widely used or generally available commercially. RPMI 1640 and several other formulas developed by
Moore and his co-workers for lymphoblastic cells in suspension culture
are commercially available. They are usually used with serum, but will
also support growth of some lines without supplementation.
Defined Media for Clonal Growth. Clonal growth in defined media has
not been widely pursued, but there are a few cases in which it is possible.
Medium F12 was developed for clonal growth of Chinese hamster ovary
and lung lines without protein supplementation, but later failed to support
such growth. Recent studies have shown that the main problem was a
requirement for selenium, which was apparently provided as a contaminant in the original studies.I The current medium of choice for such growth
is MCDB 301, which corrects for the selenium deficiency and also incor-
90
BASIC METHODS
[S]
porates other improvements. However, it is not yet commercially available. Commercially prepared F12 can be used with a selenium supplement, but in many cases it will also be necessary to add linoleic acid,
which is extremely labile to oxidation and is often not present in an active
form in commercial F12, including dry powder formulations.
Defined Media for Normal Cells. A number of defined media have been
developed specifically for maintenance of normal cells. Although little or
no growth occurs without serum supplementation, the cells remain fully
viable, and in some cases they continue along developmental pathways
during prolonged periods in culture. When supplemented with serum,
these media generally support excellent growth of normal cells. The original assay~used for development of medium 199 was long-term survival of
chicken embryo heart fibroblasts in the absence of serum supplementation. When supplemented with serum, medium 199 supports good growth
of many types of cells, and it is still widely used. In our own laboratory,
we have recently found it to be the best starting point among commercially available media for the development of a medium that is specifically
optimized for human epidermal keratinocytes.2a
Medium CMRL 1415 was developed for survival of mouse embryo
fibroblasts and for their growth when supplemented with partially purified
serum fractions. Our experience suggests that clonal growth of mouse
embryo fibroblasts with dialyzed serum can be improved by omitting the
very high level of cysteine in CMRL 1415 and using only the lower level of
cystine,a However, even better growth can be obtained with medium
MCDB 401 plus dialyzed serum. Medium MCDB 401 and most of the other
MCDB media, which were developed for growth of normal or nontransformed cells with small amounts of dialyzed serum, will also support
prolonged survival of their respective cells, even at clonal density, without
protein supplementation.1
Several special-purpose defined media for differentiated cells have appeared in the recent literature, including media E and G of Williams for rat
liver cells; the very complex defined medium of Parsa for maintenance
and differentiation of pancreatic rudiments; medium SM-20 of Halle and
Wollenberger for beating heart cells; and modifications of Waymouth's
media for liver and pancreatic cells (Table IG).
[5]
91
are needed? (2) Is its use feasible on the scale that is needed? The former
is determined by trying the medium, often in competition with other
media, and the latter by practical considerations such as how hard the
medium is to prepare and whether or not it is commercially available.
For laboratories not equipped to prepare their own media, lack of
commercial availability can be a serious obstacle to trying new or unusual
media. It is possible to obtain custom-made batches of media, but the
companies generally require a rather large minimum order to recover their
expenses, and they do not have testing programs for determining whether
the batch that they prepare actually measures up to the claims made about
it in the literature. (Lack of adequate testing can also be a problem with
standard media. Often media are tested only for sterility and ability to
support monolayer growth with whole serum. Special-purpose media,
such as those designed for clonal growth with dialyzed serum, are frequently not tested to see if they will actually do what they are supposed
to. If problems arise, it is desirable to find out from the supplier exactly
what testing has been done.)
For the novice, it is generally desirable to begin with media and culture
conditions that previously have been reported to support growth of the
cell in question. Cell culture is a complex art with many different things
that can go wrong. It is therefore highly desirable to begin with conditions
that are known to work for the cell, and to wait until the initial problems of
getting it to grow have been resolved before attempting innovations. In
many instances, if the original conditions work well and are compatible
with the planned experiment, no further experimentation with media may
be necessary.
If previously reported culture systems are not adequate, or if the cell
type has not previously been cultured from the species that is to be used,
it will be necessary to begin a program of systematic testing of media.
Generally it is desirable to begin by attempting to obtain good growth with
whole serum, and then to proceed as far as needed toward chemical
definition. A generalized outline for that process is given in Table III, and
the details are presented in several other papers. 1,z,9"2
Species is a major factor in determining which medium to use, but
tissue specificity can also have a major effect. Media that tend to have
rather broad species applicability include 199, MEM, DME, F12K, and
MCDB 202. For human and monkey cells, 199, BME, MEM, L15, 5a,
RPMI 1640, CMRL 1969, MCDB 104, and MCDB 202 can be tried. For rat
and rabbit, MEM, 5a, F12, F12K, and MCDB 104 may work. For nontransformed mouse cells, DME, CMRL 1415, MCDB 202, MCDB 401, and
some of the newer Waymouth formulations can be tried. For chicken
cells, 199, DME, F12K, and MCDB 202 are possibilities. Media for certain
invertebrate cells are discussed in this volume [39]. Other formulations
92
BASIC METHODS
[S]
TABLE III
GENERALIZED SEQUENCE OF STEPS FOR MINIMIZING SERUM REQUIREMENTS AND FOR
DEVELOPING DEFINED MEDIA a
1. Obtain growth in vitro of the type of cell of interest by any means necessary.
2. Modify media, supplements, and culture techniques as needed to obtain clonal growth.
3. Survey readily available media and supplements and select the combination that supports the best clonal growth.
4. Replace all undefined supplements, e.g., serum, with dialyzed supplements plus ultrafiltrates, hydrolysates, etc., if they are needed.
5. Identify all low-molecular-weight nutrients needed for clonal growth in thepresence of
dialyzed supplements and add them to the synthetic portion of the medium.
6. Reduce the amount of the dialyzed supplement(s) to a level that supports less than
optimum growth.
7. a. Adjust all components of the synthetic portion of the medium sequentially to experimentally determined optimum concentrations for clonal growth with minimal
amounts of serum protein, beginning with the one found to be most limiting in a
preliminary survey.
b. Systematically test low-molecular-weight compounds considered likely to have
growth-promoting activity, e.g., trace elements and nutrients known to be required
by other types of cells or organisms.
c. Test complex low-molecular-weight mixtures such as hydrolysates, extracts, and
ultrafiltrates for growth-promoting activity.
d. Attempt to separate low-molecular-weight growth-promoting activity from the
dialyzed supplements through the use of dissociating conditions. If activity is detected in steps 7c or 7d, return to step 5.
e. Attempt to improve growth through refinement of the culture techniques (e.g., use of
treated dishes, trypsinization at low temperature, etc.).
8. Whenever growth is improved significantly, reduce the concentration of the dialyzed
supplement until it is again growth-limiting, and repeat all parts of step 7.
9. Keep repeating steps 7 and 8 until no further benefits can be obtained.
10. Isolate and characterize all macromolecular growth-promoting activities that continue to
be required under the "optimum" conditions developed in step 9.
11. Identify all "contaminants" both in the synthetic medium and in the "purified" macromolecular factors, and if any affect growth, add them to the synthetic medium at their
experimentally determined optimum concentrations.
12. Verify experimentally that each component of the "final" medium is at its optimum
concentration, and make any changes that may be needed as a result of adjustments
made in steps l0 and 11.
After all requirements for multiplication of isolated cells have been identified and optimized, it may be necessary to make additional adjustments for monolayer growth,
e.g., by adjusting nutrients to the upper end of their optimum plateaus to avoid depletion and adding factors needed to reduce the effects of density-dependent inhibition.
[5]
93
94
BASIC METHODS
[6]
[6] T h e G r o w t h o f C e l l s in S e r u m - F r e e
Hormone-Supplemented Media
B y JANE BOTTENSTEIN, IZUMI HAYASHI, SHARON HUTCHINGS, HIDEO
REEN WU
Introduction (G. Sato)
A series of experiments from our laboratory indicated that hormonal
depletion of the serum component of the medium is necessary to demonstrate hormone-dependent growth of cells in culture.l-3 From these observations, it was proposed that the main role of serum in cell culture is to
provide complexes of hormones and that it should be possible to substitute hormones for serum in cell culture media. 4 Experimental support for
this hypothesis was first provided in the case of GH3, HeLa, and B H K
cells and extended to other cell lines. 5-7 In this article we present the
practical details for growing a number of cell lines in serum-free
hormone-supplemented medium.
In the absence of serum, greater than usual care must be taken in
preparation of the synthetic portion of the medium. The water used in
preparation of our medium is distilled in glass stills in three steps. In the
first, deionized water is distilled and collected. It is placed in the boiler of
a second still to which is added approximately 10 g of potassium permanganate and 1 ml of concentrated sulfuric acid per liter. The distillate from
the second still is freshly distilled from a third still just prior to the preparation of the medium. Careful preparation of water is essential for consistent results with srum-free medium.
Serum and even dialyzed serum can mask nutritional requirements of
1j. L. Clark, K. L. Jones, D. Gospodarowicz, and G. H. Sato, Nature (London), New Biol.
236, 180 (1972).
2 H. A. Armelin, K. Nishikawa, and G. H. Sato, in "Control of Proliferation and Animal
Cells" (B. Clarkson and R. Baserga, eds.), Cold Spring Harbor Lab., Cold Spring Harbor,
New York, 1974.
3 K. Nishikawa, H. A. Armelin, and G. Sato,Proc. Natl. Acad. Sci. U.S.A. 72, 483 (1975).
4G. H. Sato, Biochem. Actions Horm. 3, 391 (1975).
5 I. Hayashi and G. Sato, Nature (I~ondon) 259, 132 (1976).
6 j. Mather, R. Wu, and G. Sato, in "Growth Requirements of Vertebrate Cells in Vitro"
(R. G. Ham and C. Waymouth, eds.). Cambridge Univ. Press, London and New York. (in
press).
r S. E. Hutchings and G. H. Sato, Proc. Natl. Acad. Sci. U.S.A. 75, 901 (1978).
[6]
95
cells in culture. For this reason we use one of the more complex media,
Ham's F12, s as the basic synthetic medium. We also supplement the
medium with the trace elements recommended by Ham. a Ham's F12 medium, supplemented with trace elements, is sufficiently complex to satisfy
the nutritional requirements of cells in the absence of serum. However,
the concentration of the components is often not optimal. This is remedied
by using mixtures of F12 and DME. 1'11
Commercially available powdered media are adequate for serum-free
work although each batch must be checked for suitability.
Serum also serves the function of a trypsin inhibitor in conventional
tissue culture procedures. In the absence of serum, we use soybean
trypsin inhibitor to neutralize the trypsin after subculture. After trypsinization, the cells are suspended in serum-free F12 medium containing
soybean trypsin inhibitor (1 mg/ml), centrifuged, and resuspended in
serum-free F12 for plating.
Hormones, growth factors, and transferrin are maintained as 100
stocks and added to the medium just prior to plating the cells. Stock
cultures are usually kept in a 1:1 mixture of Ham's F12 and Dulbecco's
modified Eagle's medium (DME), supplemented with sodium bicarbonate
(1.2 g/liter), N-2-hydroxyethylpiperazine-n '-2-ethanesulfonic acid, pH 7.2
(HEPES) (15 mM), penicillin (192 units/ml), streptomycin (200 /zg/ml),
ampicillin (25 ttg/ml), 5% (v/v) horse serum, and 2.5% (v/v) fetal calf
serum. The cells are grown on plastic tissue culture dishes in a humidified
atmosphere of air containing 5% CO2.
In those cases in which subculture involves trypsinization, two techniques for growing cells in serum-free medium are used. The first involves
the transfer of cells by trypsinization from stock medium containing
serum, directly to serum-free medium. In the second procedure, cells are
first plated into serum-containing medium and, after a day, the medium is
replaced with serum-free medium after washing the cells with serum-free
medium. In those cases in which this procedure is followed, the initial
incubation in serum-containing medium may be essential. There are two
likely explanations for the requirement for serum pretreatment: (1) The
serum may be necessary for the repair of trypsinization damage after
subculture; or (2) the residual serum left after its removal, even after one
or more washes, may be furnishing unknown growth factors. The latter
possibility seems unlikely in view of the extensive growth which is possi8 R. G. Ham, Proc, Natl. Acad. Sci. U.S.A. 58, 288 (1965).
aW. L. McKeehan, W. G. Hamilton, and R. G. Ham, Proc. Natl. Acad. Sci. U.S.A. 72,
2023 (1976).
10 H. Eagle, Science 122, 501 (1955).
11 j. D. Smith, G. Freeman, M. Vogt, and R. Dulbecco, Virology 12, 185 (1960).
96
BASIC METHODS
[6]
[6]
97
lar cells TM4, TM rat glioma C6,2 mouse embryonal carcinoma, 21,z~ Swiss
3T3, 23 and a line of cells isolated from rabbit aortic intima. 24
Note
98
BASIC M E T H O D S
[6]
TABLE I
EFFECTS OF GROWTH FACTORS ON BI04 CELLS a
Cell number 103
Addition
None
4F
4F + NasSeOa
1 0 % f e t a l calf serum
Day 3
179
493
573
1307
_+ 19
-+ 11
-+ 14
-+ 3 0
Day 5
78
24
1856
6395
-+ 18
--- 5
_ 97
--- 186
--28
21
a Data are expressed as the mean -+ SD of triplicate samples. Initial inoculum is 7 x 104.
[6]
SERUM-FREE
HORMONE-SUPPLEMENTED
MEDIA
99
get good hormone response. The stocks are subcultured about once a
week.
For growing GHa cells in serum-free condition, exponentially growing
stock cells are trypsinized (0.1% trypsin in PBS) and then treated with
soybean trypsin inhibitor (0.1% of the inhibitor in PBS). The treated cells
are washed by centrifugation, resuspended in serum-free F12 medium,
and inoculated into hormone-containing, serum-free F12 medium at 0.5
105 (35-mm dishes) or 1.0 10~(60-mm dishes) per plate. Unlike the other
lines described here, we have found that GH3 cells prefer F12 medium to
any other media, or the combination of F12 and DME. These cells have
not responded to hormones under serum-free conditions unless F12 was
used. The hormones and their final concentration in the medium that are
required for growth of GH3 cells in the serum-free condition are as follows: 3,3',5-triiodothyronine (3 10-11 M), thyrotropin-releasing hormone (1 ng/ml), parathyroid hormone (0.5 ng/ml), transferrin (5 /zg/ml),
insulin (5 ttg/ml), fibroblast growth factor (1 ng/ml), and somatomedin C (1
ng/ml). 26 When these hormones are added to the serum-free F12 medium,
the cells not only grow as well as in serum-supplemented medium, but
also can be maintained for a long period by serial propagation. Requirement for parathyroid hormone, which has the least effect on short-term
growth of these cells, will become obvious in such long-term culture in
serum-free condition. For long-term growth in serum-free, hormonesupplemented medium, trace metals such as selenium and cadium are
added at the final concentration of 50nM and 0.5/JM, respectively. Some
of the other trace metals, especially manganese, are strongly inhibitory to
GH3 cells and, therefore, are not used for these cells.
One of the immediate interests in defining hormone requirements of a
cell line and growing it in a serum-free condition is the question of the
universality of the requirement. For instance, will the hormone combination that allows the growth of GH3 cells also allow the growth of normal
rat pituitary cells which produce growth hormone and/or prolactin? To
examine these possibilities, we made primary cultures from young normal
rat pituitaries. The pituitaries were washed several times in serum-free
medium, minced in the presence of trypsin inhibitor, and plated in serumfree F12 medium supplemented with the hormones mentioned above.
Comparison of such cultures with those in serum-supplemented (10% fetal
calf serum) F12 medium showed that there is at least a significant suppression of fibroblast growth in serum-free, hormone supplemented media. At
present, it is too early to determine whether the cells in serum-free,
hormone-supplemented medium are the normal equivalent of GH3 cells
which produce growth hormone and/or prolactin, but the results indicate
~ I. Hayashi, J. Larner, and G. Sato, In Vitro 14, 23 (1978).
100
BASIC METHODS
[6]
[6]
101
Serum-free medium supplemented with insulin, fibroblast growth factor, transferrin, and a crude extract prepared from female rat submaxillary gland, rat submaxillary gimmel factor, supports continuous division
cycles of Balb/c 3T3 mouse fibroblasts washed free of serum. Under these
serum-free conditions, the growth rate and saturation density are similar
to those observed for the same cells cultured in DME containing 10% fetal
calf serum. Morphologically, these cells appear more epithelioid than
their serum-grown counterparts.
Trypsinized stock cultures of logarithically growing Balb/c 3T3 cells,
maintained at an early passage number, are subcultured at 5 104 cells
per dish in DME plus 10% fetal calf serum and incubated ovemight at 37
in a humidified atmosphere containing 5% CO2. On day 1, after washing
each culture twice with serum-free DME, the medium is changed to
serum-free medium, consisting of three parts DME and one part F12, plus
trace elements. Hormone additions are made directly into each culture
dish from freshly prepared concentrated stock solutions. The final concentrations of insulin, transferrin, hydrocortisone, EGF, and NIH-LH ( a
source of FGF) are 2/xg/ml, 5/xg/ml, 10-7 M, 50 ng/ml, and 1 /xg/ml,
respectively. Rat submaxillary gimmel factor is added at a concentration
of 2 /zg/ml. The preparation of gimmel factor is described in the section
on C6 glial cells.
Balb/c 3T3 fibroblasts, washed free of serum and incubated in serumfree media supplemented with insulin, transferrin, hydrocortisone,
NIH-LH (FGF), EGF, and a crude preparation of rat submaxillary gland,
grow from a low cell density (3 x 103 cells/cm2) to form a confluent
102
aASIC METHODS
[6]
[6]
103
104
BASIC M E T H O D S
[6]
Cell no. x 10 -5
4.7
48.2
10.3
8.6
12.2
8.4
15.0
36.8
53 8
-+ 0.9
_+ 0.5
+ 0.1
0.4
-+ 0.7
_+ 0.1
-- 0.6
-+ 0.3
0.7
[6]
SERUM-FREE
HORMONE-SUPPLEMENTED
, MEDIA
105
106
BASIC METHODS
[6]
conflttency. The medium is changed every other day with cultures maintained at 37 in a humid atmosphere containing 5% CO2. The cells can be
grown in a serum-free medium consisting of 75% DME and 25% F12
enriched with a mixture of trace elements; 9 insulin (0.5 /Lg/ml); NIHLH-B9, a biological standard of bovine luteinizing hormone (2/~g/ml); and
rat submaxillary gimmel factor prepared as described in the section on
C6 cells.
In this medium, the cells have the same morphology as those cultured
in the presence of 10% fetal calf serum. In serum-rich medium, they divide
with a generation time of 20 hr and reach a saturation density of 75 x 103
cells/cm2; in hormone-supplemented medium, their generation time is 28
hr and they reach a saturation density of approximately 36 10 a cells/cm ~.
The efficiency of this serum-free medium for supporting the growth and
viability of 3T3-L1 depends on the presence of gimmel; its omission results
in an 80% inhibition of growth.
The nature of the factor responsible for the multiplication of 3T3 is
under study. Experiments show that purified EGF is unable to replace
gimmel in supporting the growth of 3T3. The omission of NIH-LH-B9,
insulin, or transferrin results in growth inhibition of 26%, 40%, and 72%,
respectively. In the work described above, the cells were plated and
incubated with DME/10% fetal calf serum for 15 hr after trypsinization
before being cultured in serum-free medium. This "serum-incubation"
appears to be necessary for the cells to recover from the trypsinization
and to spread on the dishes, a process that seems important in the response of the cells to the hormones and growth factors. Attempts to
eliminate the step of serum incubation are in progress. Addition of serum
fractions such as fetuin, and Cohn fractions I and IV, seem to improve
greatly the plating efficiency and the spreading of 3T3-L1 in a serum-free
medium just after trypsinization.
Another clone of mouse Swiss 3T3, 3T3-C2, that does not differentiate
into adipocytes also can be grown in the absence of serum in a medium
similar to that established for 3T3-L1.
Rabbit Intimal Cell Line in a Serum-Free Medium (G. Serrero)
The cell line discussed in this section was established by Dr. Vincenzo
Buonassisi from cells isolated by trypsinization of the subendothelial portion of the intima of rabbit aorta. Cells are routinely cultured in F12
medium supplemented with 10% fetal calf serum and kept at 37 in a
humidified atmosphere containing 5% COs. The cells have a fibroblast-like
appearance in a subconfluent stage and become enlarged and oriented in a
parallel fashion in the culture dish when they reach confluency. The cells
grow in a serum-free medium that consists of F12 enriched with trace
[6]
SERUM-FREE HORMONE-SUPPLEMENTED
MEDIA
107
108
BASIC M E T H O D S
600
e) 500
'f
..~---
[6]
"~ck
~ 400
0
",.
300
200
100
60
120
180
240
MINUTES
FIo. 1 Time course of 1zsI-EGF binding. Six hundred microliters of standard binding
mixture [SF-4X(-EGF), 8 ng/ml 125I-EGF] are applied to the dishes at 37 for various times.
n , cells grown in the hormone-supplemented medium; , cells grown in 10% FCS.
[6]
SERUM-FREE HORMONE-SUPPLEMENTED
MEDIA
109
110
BASIC
METHODS
[7]
[7] U s e o f A n t i b i o t i c s in Cell C u l t u r e M e d i a
By D. PERLMAN
Like Death and Taxes, contamination or the threat o f contamination
is always with us and we need all the weapons to combat them.1
Introduction
Bacterial, yeast, and fungal contamination are hazards to those studying various phases of metabolism and growth of mammalian cellsin vitro.
Although we have used antibiotics for more than 30 years to eliminate or
suppress unwanted microbial contaminants, many who add them to tissue
cultures for this purpose are unfamiliar with the products used, the limitations for their use, and the practical value of this strategy.
The basic requirements are:
1. The antibiotic must eliminate the microbial contaminant. (Bactericidal compounds are preferred over bacteriostatic.)
2. The antibiotic must not inhibit growth and metabolism of the
mammalian cells in tissue culture.
3. The antibiotic must provide "protection" for the complete experimental period.
4. The antibiotic should not affect any ultimate use intended for the
mammalian cells, e.g., virus production or the preparation of antigens.
5. The antibiotic should be nontoxic and safe as far as handling by
laboratory personnel is concerned.
6. The antibiotic should be compatible with other components of the
culture media.
7. The antibiotic should be inexpensive and should not contain excipients, e.g., buffers, which affect cell growth or metabolism adversely.
Materials
[7]
111
Those available from the local pharmacy are products that are usually
intended for parenteral administration in the therapy of acute infectious
diseases. They frequently contain buffers, organic acids, and other excipients which aid their usefulness as therapeutic dosage forms, but might
lead to problems in mammalian tissue culture, e.g., the presence of ascorbic acid in the tetracycline formulation. The purity of the antibiotic in
these preparations is usually very high, and the manufacturer frequently
can supply information on the quantity and identity of the impurities.
However, the presence of buffers may result in the potency of the material
on a weight basis being as low as 10% that of the pure material. The
parenteral dosage form usually is sterile and the container fabricated of
materials such that there is minimal carry-over of plasticizers and other
minor chemical contaminants. The capsular dosage form usually is
"semisterile" and the filler and other excipients are likely to be reasonably water soluble. This formulation should be dissolved in the basal
medium and then sterilized by filtration through Millipore or equivalent
filters; asbestos filters should not be used since the antibiotic may react
with the filter.
The antibiotics available from biological and chemical supply houses
include some primarily intended for use in mammalian culture media and
others which are sold on a "user's risk basis." The former should have
been tested for cytotoxicity in mammalian cell cultures, and the vendor
should have data to support claims of low or minimal cytotoxicity; each
manufacturing batch should have its own documentation. The latter materials are sometimes of uncertain origin and not infrequently contain
cytotoxic contaminants and/or microbial contaminants. The user should
be warned to evaluate these materials for cytotoxicity over a period of
time before adding them to a critical experiment. This evaluation might
include study of the interaction of the antibiotic (or mixture o f antibiotics)
with constituents of the medium and other factors.
Several antibiotics may be obtained directly from the manufacturer.
These are usually in the form of nonsterile powders, and the administrative difficulties in obtaining them frequently outweigh the advantages of
obtaining them through this route. Most of the manufacturers are reluctant to sell a few grams of material, and most laboratories are not interested in purchasing kilogram amounts. The result is often a gift of a few
grams, and no guarantees are involved with regard to potency, presence
of microbial and/or chemical contaminants, or cytotoxicity.
Some of the antibiotics which may be useful in the elimination or at
least suppression of contaminating microorganisms from mammalian cell
cultures are listed in Table I together with information on the useful concentration and the length of time protection might be expected under
112
BASIC METHODS
[7]
TABLE I
SOME ANTIBIOTICS USEFUL FOR ELIMINATION OR SUPPRESSION OF CONTAMINATING
MICROORGANISMS FROM MAMMALIAN CELL CULTURES
Antibiotic
Amoxicillin
Amphotericin B
(as deoxycholate
complex)
Amphotericin B
methyl ester
Ampicillin
Carbenicillin
Cephalothin
Chloramphenicol
7-Chlortetracycline
6-Demethyl-7chlortetracycline
Dihydrostreptomycin
Doxycycline
Erythromycin
Gentamycin
5-Hydroxytetracycline
Kanamycin
Lincomycin
Neomycin
Nystatin
Antimicrobial spectrum
Gram-positive and gramnegative bacteria
Fungi and yeasts
Recommended
concentration
in #g/ml
mediaa
Stability in
media at
37 days b
100
2.5
1.0
100
100
100
10
100
20
100
50
100
100
50
4
5
50
[7]
113
Antibiotic
Paromomycin
Polymyxin B
Penicillin G
Penicillin V
Tetracycline
Streptomycin
Tylosin
Viomycin
Antimicrobial spectrum
Gram-positive and gramnegative bacteria
Gram-negative bacteria
Gram-positive bacteria
Gram-positive bacteria
Gram-positive and gramnegative bacteria and
mycoplasma
Gram-positive and gramnegative bacteria
Gram-positive bacteria and
mycoplasma
Gram-positive and gramnegative bacteria
Recommended
coricentration
in/xg/ml
media"
Stability in
media at
37 days ~'
100
50
100
100
10
5
3
3
4
100
100
50
Concentration effective in controlling "light" infection and at the same time not cytotoxic to L~29 and KB cells in serum-containing media.
b Length of time in which at least 10% of initial activity could be demonstrated in
incubation at 37 up to 5 days in serum-containing media.
114
BASIC METHODS
[7]
TABLE II
SOME COMMERCIALLY AVAILABLE ANTIBIOTIC PREPARATIONS WHICH MAY BE USED
AS TISSUECULTUREMEDIA SUPPLEMENTS
Antibiotic
Amoxicillin
Amphotericin B
Ampicillin
Carbenicillin
Cephalothin
Chloramphenicol
7-Chlotetracycline
6-Demethyl-7-chlortetracycfine
Dihydrostreptomycin
Doxycycline
Erythromycin
Gentamb~cin
5-Hydroxytetracycline
Kanamycin
Lincomycin
Neomycin
Nystatin
Penicillin G
potassium
Penicillin V
potassium
Commercial source
Beecham Laboratories
Roche
E.R. Squibb and Sons
Bristol Laboratories
Ayerst Laboratories
Pfizer Laboratories
E. R. Squibb and Sons
Wyeth Laboratories
Beecham Laboratories
Pfizer Laboratories
Eli Lilly Company
Park, Davis and Company
Lederle Laboratories
Lederle Laboratories
Eli Lilly Company
Pfizer Laboratories
Pfizer Laboratories
Abbott Laboratories
Bristol Laboratories
Eli Lilly Company
Upjohn Company
Schering Corporation
Pfizer Laboratories
Bristol Laboratories
Upjohn Company
S.B. Penick Company
Pfizer Laboratories
E.R. Squibb and Sons
Upjohn Company
E.R. Squibb and Sons
Bristol Laboratories
E.R. Squibb and Sons
Pfizer Laboratories
Wyeth Laboratories
Bristol Laboratories
Abbott Laboratories
Eli Lilly Company
Wyeth Laboratories
Comments
Take capsules and dissolve
contents and sterile filter
Available as power for
reconstitution; contains
deoxycholate and buffer
Use injectable formulation
Distributed as sterile
solution
Use injectable formulation
Use injectable formulation
Use injectable formulation
Distributed as sterile
solution
[7]
115
TABLE II (Continued)
Antibiotic
Commercial source
Paromomycin
Polymyxin B
Streptomycin
Tylosin
Viomycin
Pfizer Laboratories
Tetracycline
Comments
Take capsules and dissolve
contents and sterile filter
Use injectable formulation
Use injectable formulation
Use injectable formulation
and note composition of
buffer
Distributed as sterile
solution
Use injectable formulation
116
BASIC
METHODS
[8]
Our studies have shown that many antibiotics may be fairly well tolerated by mammalian cells and chick tibroblasts growing in various media.
For practical purposes we recommend benzylpenicillin (penicillin G) and
dihydrostreptomycin for elimination or suppression of bacteria, and gentamicin or tylosin to prevent growth of mycoplasmas. Amphotericin B is
fairly useful in controlling fungal or yeast contamination but we question
whether it should be used on a continuous basis since it is somewhat toxic
to most cells.
If the bacterial contaminants turn out to be resistant to the combination of penicillin with dihydrostreptomycin, treatment of the contaminated mammalian cell cultures with tetracycline or chloramphenicol may
be one approach to rescuing the cell line. Cross-resistance among antibiotics should be given some consideration when resistant contaminating organisms are the enemy: organisms resistant to penicillin G are likely to be
resistant to ampicillin, penicillin V, and p~rhaps the cephalosporins,
cephalothin and cephalexin. Organisms resistant to dihydrostreptomycin
may be resistant to kanamycin, gentamy~in,lneomycin, paromomycin,
and ribostamycin.
[8] L a b o r a t o r y M a n a g e m e n t
of Cell Cultures
By KEN WOLF
Introduction
Animal cell or tissue cultures--widely used in different scientific
disciplines--are biological systems that are seldom completely predictable or amenable to absolute control. Accordingly, cell culture has some
attributes of a science while other characteristics resemble those of an art.
As such, some practitioners are consistently more adept or successful
than others. However, anyone who has had more than passing experience
with cell cultures soon learns that things can and do go awry; in fact,
catastrophes sometimes happen.
Contamination is an ever-present threat; bacteria, yeasts, and molds
can flourish in tissue culture media. The presence of such microbial contaminants, fortunately, is usually quickly and readily visible. Moreover,
in many situations, antibiotics are used to minimize the risk of contamination. Cell cultures may also become contaminated but show little or no
visible signs of a problem; virus is one example of nonapparent infection,
but mycoplasmas are by far the most frequent culprits. Detection of
mycoplasma contamination with any degree of confidence is a challenging
feat. A variety of methods have been developed for this purpose and for
METHODS IN ENZYMOLOGY, VOL. LVIII
[8]
117
118
BASIC METHODS
[8]
are grown to maintain the lineage under the best possible conditions and
working cultures that are to be used for the actual work applications. Propagation of stock cultures must be accorded the highest work priority and
be carded out with strict aseptic technique and without antibiotics.
3. The second element of dichotomy of the system is in carrying two
sets of stock cultures. To minimize the risk of culture loss due to equipment failure, the two sets should be housed in different incubators. Each
set of cultures should have its own pretested, reserved, single-use portions of frozen, antibiotic-free media; separate records; and separate
schedules for handling. Ideally, the division of risk and responsibility
should be between two persons, but the principle can be employed by an
individual who does the work as two independent activities.
4. Each stock culture should be maintained on two different lots of
appropriate medium. Stock culture medium stored at - 2 0 or lower remains useful for several years, and single-use portions minimize risk of
contamination and obviate formation of precipitates from repeated
freeze-thaw cycles. In practice, stock cultures can be kept at reduced
temperature and may require handling only once a month and, in some
cases, only several times a year.
5. Serum should be obtained from a supplier who certifies it to be free
of mycoplasma. Several different samples are obtained prior to purchase
and tested to determine which gives the best plating efficiency for the cells
to be used. For this test, a suspension containing about 200 cells is added
to duplicate or triplicate 25 cm 2 flasks containing 5 ml of medium with at
least a 20% level of the several samples of serum to be tested. Incubation
is allowed to proceed for 2-3 weeks, after which the flasks are drained,
rinsed with balanced salt solution, stained with 1% aqueous or alcoholic
crystal violet, wasl~ed, and the cell colonies (clones) counted. That serum
giving the most clones should be purchased. Additional measures for
quality control of serum are considered in this volume [2]. Serum should
be stored at - 2 0 or lower and may safely be kept for at least a year
without noticeable loss of potency.
6. Medium and other solutions are prepared only with water of
tissue-culture quality. A freshly prepared borosilicate glass distillate of
deionized water is usually satisfactory. Preferred storage is under sterile
conditions in borosilicate glass. For the small user, USP water for injection is suitable; it is available from pharmacies and hospital supply houses
in liter bottles.
7. Locally prepared media or solutions must be thoroughly tested for
sterility before use. Samples of at least 5% are suggested as is incubation
for 7-10 days at the usual incubation temperature and at 37 . Details are
presented in this volume [2].
[9]
DISPERSION
AND DISRUPTION
OF T I S S U E S
119
8. To minimize adulteration of one cell line with another, only one cell
line should be opened and handled at any given time. All cultures must be
identified at the time of handling.
9. Stock cultures should be propagated in multiples. Because plastic
vessels permit slow gas exchange, 16 x 125 glass tubes with rubber-lined
screw caps, small flasks, or bottles are advantageous since they assure gas
retention, provide multiple cultures in reduced space, and are easily examined microscopically.
10. Stock cultures are to be handled with minimal frequency. To accomplish this use reduced seeding levels and low-temperature incubation.
I1. Maintain detailed records of medium and solutions that are prepared. Include relevant data on all components and notations of when,
how, and by whom cells are handled.
12. Stock cultures should be tested at least once a year and preferably
twice for mycoplasma and other possible contaminants.
13. Replace regular working cultures annually with newly expanded
stock cultures.
14. Attempts should be made to disperse the cells from stock cultures
with an autoclaved solution of EDTA that does not contain trypsin. Trypsin
solutions are not sterilized, and they represent a possible source of culture
contamination. Accordingly, for greatest security, stock cultures should
be dispersed with an autoclaved solution of EDTA.
15. Frozen stocks of unique cell populations or those that are epecially
valuable should be maintained in liquid nitrogen and at more than one
facility. The American Type Culture Collection, Rockville, Maryland
20852 offers such a safe-deposit service.
Initial studies in tissue culture carried out near the beginning of this
century involved the explantation of fragments of tissue from an organism
into an artificial in vitro environment. Propagation of such explants usually
involved aseptically cutting or trimming the original explant to reduce its
size so that appropriate gas and nutrient diffusion could occur, and so that
multiple cultures could be obtained. From these pioneering efforts have
developed the closely related disciplines of cell, tissue, and organ culture.
i T h e opinions or assertions contained herein are t h e private views o f the author and are not
to be c o n s t r u e d as offacial or as reflecting the views o f t h e D e p a r t m e n t o f the A r m y or the
D e p a r t m e n t o f Defense. Citation o f trade n a m e s in this report does not constitute an
offacial e n d o r s e m e n t or approval o f the u s e o f s u c h items.
METHODS IN ENZYMOLtX~Y, VOL. LVIII
120
BASIC METHODS
[9]
[9]
DISPERSION
AND
DISRUPTION
OF
TISSUES
121
,O
"O
;>..
e-.
'
m
t.
:::t
~.
--
--.
~t
',D
O~
%
z
-/
[-
0
r~
~D
<
t::
e'~
O~ ON
"O
mm
,.--:.~
m
J
,.
~
tt3
,.-2,
0
Z
["
oe~
O",
~ . ,eeq q~
t..
~mo
~ _ ~ : ~ , . ~
<
..,:
~.~
o ~
122
BASIC METHODS
[9]
which is not buffered with bicarbonate should be used, since loss of CO2
will cause the solution to become too alkaline. On the other hand, if some
or all steps will be done under an air/COs atmosphere, then a BSS using a
bicarbonate buffer will have to be employed to prevent acidification of the
medium by dissolved COs. Phenol red may be included, usually at a
concentration of 20-50 mg/liter, as a visual indicator of pH. Glucose may
be included or omitted, depending upon the time elapsing between extirpation of the tissue and the final dispersion of the cells in complete growth
medium. The final consideration in the choice of BSS composition is the
inclusion or exclusion of the divalent cations calcium (Ca) and magnesium
(Mg). As early as 1900, it was recognized that these ions increased the
stability of the intracellular matrix, and many investigators have confirmed this in tissue culture experiments. For this reason several of the
BSS are also commercially available (or are prepared in the laboratory) as
calcium-and magnesium-free solutions (CMF-BSS). The further addition
of chelators, or the readdition of Ca or Mg when certain enzymes are used
as dispersing agents, is discussed below.
It is not the intent of this section to present an extensive discussion of
growth media, as this is covered elsewhere in this volume [5]. Many
formulations are available commercially as sterile solutions and in some
instances as sterile concentrates or dry powders. Most such media were
originally formulated for a particular cell type, such as RPMI-1640 for
lymphocytes or Dulbecco's modified Eagle's medium (DMEM) for fibroblasts. In almost all instances these media are further supplemented with
an animal serum or sera (horse, calf, fetal calf, human; 2-20%). Regardless of the type or composition of the growth medium, or the nature of the
supplements which are added to it, all represent no more than an elaboration of nutrients, hormones, or "growth factors" to a BSS. Occasionally
these media, usually without a serum supplementation, are used in place
of BSS during the intermediate stages of tissue dispersal. More frequently,
however, they are used only as a fnal rinse, immediately prior to suspension of the cells in the complete (serum-supplemented) growth medium
and plating in the culture vessels as described in a later section.
The use and choice of antibiotics is discussed seperately from several
points of view in this volume [2] [3] and [7].
Procurement and Preparation of the Tissue
The source or sources of tissue to be cultured will vary with the design
and needs of a particular laboratory. Two precautions should be observed
in all cases insofar as is possible. The tissue must be kept moist, and a
minimal amount of time should elapse between extirpation of the tissue
and initiation of culture. Under ideal circumstances the culture laboratory
[9]
123
will be situated adjacent to the surgical rooms. Recognizing that this ideal
is seldom realized, the following suggestions are made. Samples obtained
from a hospital operating room may be placed by the surgeon in sterile
dishes containing a BSS that one supplied. Once this is accomplished, an
assistant can move them to the culture laboratory. When a considerable
time may elapse between excision and culture, it may be advisable to
place the tissue in a BSS containing glucose or in a complete culture
medium. If extensive transportation is involved, as between a
slaughterhouse and the laboratory, chilling with ice may be required. For
small samples a 100-150 mm sterile, disposable tissue culture dish may be
employed. These are conveniently sealed for transportation by stretching
a length of Parafilm " M " (American Can Co., Neenah, Wisconsin) around
the circumference two or three times. The closest situation to ideal is
when tissues are removed from laboratory animals, in which case the
excision of tissue can be accomplished readily in or very near the culture
laboratory. The animal is anesthetized or sacrificed, the area of incision
shaved (if necessary), sterilized by liberal application of 70% ethanol, and
the sterile field isolated by draping the animal with sterile cloth or gauze.
The incision is then made, the tissue located, removed aseptically, and
placed immediately in sterile BSS or complete medium. The instruments
should be chosen and the surgery accomplished in such a way as to
minimize mechanical trauma to the tissue.
When the tissue is of fetal origin, the uterus is first removed in toto to a
suitable sterile dish, the fetuses dissected out, decapitated, transferred to
clean sterile dishes, and the tissue(s) then removed. Fresh sterile BSS is
used whenever the material needs to be moistened or rinsed free of blood
or small pieces of debris; excess BSS can be aspirated using sterile Pasteur pipettes connected by tubing to a vacuum flask. Small surgical instruments which are used repeatedly may be cleaned and sterilized readily by keeping at hand a beaker of detergent solution and a small brush (a
toothbrush is adequate), one or two beakers of distilled water for rinsing,
and a container in which the instruments can be immersed in 70% ethanol.
Before use, instruments are removed and dried by evaporation or flaming
(carefully).
Dispersion of the Tissue
It is only necessary to consider the extreme differences in tissue architecture exhibited by tissues such as brain, heart, skeletal muscle, liver,
lung, and kidney to appreciate that there is no "standard" procedure for
preparing cell suspensions from these or any other tissues. Perhaps as we
learn more about the nature of the cell surface and cell-cell interactions
we will be able to devise better, more uniform methods for disrupting the
124
BASIC METHODS
[9]
[9]
125
become very popular. Several other enzymes have also been used, notably pronase, u-13 collagenase,14'15 hyaluronidase,14-17 elastase, 15'~7 pancreatin, 17,1s papain, 17 and deoxyribonuclease.17.18 It should be mentioned
that virtually all of these enzymes normally contain several activities.
Crude trypsin may contain, in addition to trypsin, chymotrypsin, elastase, ribonuclease, deoxyribonuclease, and amylase. This may be of
considerable importance in designing a protocol for preparing cell suspensions, since in some instances the efficacy of a trypsin-induced dissociation has been attributed to the presence of contaminating activities and
substitution of more highly purified crystalline trypsin has proven less
effective or even harmful. On the other hand, some investigators have
found that crude trypsin contains toxic or harmful substances, whereas
the highly purified enzyme is suitable. A statement of the rationale for
including deoxyribonuclease in some dispersing media may be instructive
at this point with regard to "contaminating" enzyme activities. Essner et
al. 19 found that clumps of a rat ascites hepatoma could be dispersed by
several treatments. Disaggregation by trypsin or chymotrypsin proceeded
in two phases, the first being dissolution of the clumps into single cells, the
second phase being the agglutination of freed cells by a mucous-like
coagulum which formed. Auerbach and Grobstein, 2 studying dissociation
and reaggregation of embryonic mouse tissues, found that certain lots of
trypsin produced a "gummy" material which enmeshed the cells and cell
clumps, preventing complete disaggregation. This material was not
formed when crude pancreatin was included in their trypsin solutions.
SteinbergTM subsequently observed the appearance of a "slimy" material
which trapped cells that had been obtained by trypsin dissociation of heart
and liver cells from 5-day chick embryos. This material was shown to be
DNA which was released from damaged cells, hydrated, and uncoiled to
form a viscous material which readily enmeshed the cells. Steinberg
found that inclusion of crystalline deoxyribonuclease in the dissociation
medium would completely prevent the appearance of the gel.
In recent years better preparations of several enzymes have become
available for tissue culture work. As an example, Worthington Biochemiu D. Weinstein, Exp. Cell Res. 43, 234 (1966).
12 j. C. Sullivan and I. A. Schafer, Exp. Cell Res. 43, 676 (1966).
13 j. F. Foley and B. Aftonomos, J. Cell. Physiol. 75, 159 (1970).
14 p. O. Seglen, Exp. Cell Res. 76, 25 (1973).
P. O. Seglen, Exp. Cell Res. 82, 391 (1973).
16 L. M. J. Rinaldini, Exp. Cell Res. 16, 477 (1959).
17 L. L. Wiseman arid W. R. Hammond, J. Exp. Zool. 197, 429 (1976).
is M. S. Steinberg, Exp. Cell Res. 30, 257 (1963).
19 E. Essner, H. Sato, and M. Belkin, Exp. Cell Res. 7, 430 (1954).
so R. Auerbach and C. Grobstein, Exp. Cell Res. 15, 384 (1958).
126
BASIC METHODS
[9]
[9]
127
128
BASIC METHODS
[9]
Adult rat liver is an organ which is particularly amenable to mechanical perfusion, both in situ and in vitro. Seglen 14,15has described a method
for preparihg hepatocyte suspensions from adult rat liver which has been
widely used and occasionally modified. The procedure is unique in that
the dispersant solutions are perfused through the liver rather than treating
minced tissue. The method also involves sequential treatment of the tissue
with different solutions. The method as used by Williams et ai. a4 is briefly
31Availablefrom Tetko, MontereyPark, California91754.
32Availablefrom PerforatedProducts Inc., Brookline,Massachusetts02146.
oa Disposable, sterileplastic centrifugetubes, with caps, are availablein 15- and 40-ml sizes
from several manufacturersof tissue culture plasticware.
34G. M. Williams,E. Bermudez,and D. Scaramuzzino,In Vitro 13, 890 (1977).
[9]
129
stated as follows. The liver of an adult rat is perfused for 4 min with
Hanks's CMF-BSS buffered with 0.05 M HEPES 35 and containing 0.5
mM EGTA. 2z After this treatment the liver is perfused for 10-12 min with a
coilagenase solution (100 units/ml) in Williams' s Medium E a6 buffered with
0.05 M HEPES. The cells are combed into fresh collagenase solution,
centrifuged (50 g for 4 min), and resuspended in complete growth medium
before inoculating into culture vessels.
130
BASIC METHODS
[9]
[9]
131
the advantages are that the method is more rapid than conventional trypsinization; that the cells are released directly into the growth medium,
myoblasts having a much shorter recovery period before fusion occurs;
and that a lower level of fibroblast contamination is achieved.
Similar procedures have been used for other types of muscle4~ and for
neural tissue, 4a and they may prove useful for other tissues as well.
Determining Cell Yield and Viability
Judging a procedure for the preparation of cell suspensions from living
tissues as successful or.unsuccessful depends to a certain extent upon the
needs and requirements of the investigator and the purpose for which the
cell cultures are being established. Important considerations in making
such a judgement are: the adequacy of the cell yield, i.e., the number of
cells obtained per gram or milligram of tissue; the viability of the cells;
and the ability of the cells tO retain in culture the properties and functions
expected of them.
The cell yield is determined by counting the number of ceils in a
known volume of the final suspension and calculating the total number of
cells obtained from the original mass of tissue (See this volume [11]).
The most commonly used criterion for cell viability is based on the
assumption that viable cells will exclude certain dyes such as trypan blue,
whereas nonviable cells will take up the dye (See this volume [13]). Other
criteria for viability include measures of the ability of the cells to attach to
the surface of the culture vessel, i.e., the fraction of cells inoculated into a
vessel that will attach to the surface. Occasionally, the ability of the cells
to take up or incorporate a radioactive compound may be useful. The
ability of cells to survive normal culture conditions or clonal growth conditions is a further criteria of viability. The integrity of the cell membrane
and the general appearance of the suspended cells should be monitored by
phase-contrast microscopy. If possible, the ultrastructure of the cells
should be examined by scanning and transmission electron microscopy.
132
BASIC METHODS
[10]
[10]
B y JAMES A . M C A T E E R a n d W I L L I A M
H. J.
DOUGLAS
Introduction
Monolayer cell cultures are frequently established from single cell
suspensions prepared by the enzymic dissociatibn of organ fragments.
The cell preparation is inoculated into a culture vessel containing fluid
medium and incubated in a controlled atmosphere. Viable cells settle and
attach to the substrate within several hours. The resultan t primary culture
is a mixed cell population which contains many of the cell types present in
the tissue of origin. In order to obtain monolayer cultures consisting of a
single cell type, it is necessary to isolate that cell from the primary preparation. Cell separation and isolation methods such as density gradient
centrifugation, ~ electrophoresis, 2 or affinity column separation a can be
applied to the initial cell suspension prior to culture. Alternatively, the
cell population can be enriched using in vitro methods that select cell types
based on their attachment or growth characteristics. For example, fibroblasts and macrophages attach to culture surfaces more rapidly than other
cells and thus they can be removed by selective adherence techniques. 4
Fibroblasts replicate faster than most cell types. In mixed cell cultures
they tend to overgrow the population and obscure other cells of interest.
Dilution plating at clonal density 5 is often used to separate cell types so
that colonies which develop may be physically isolated and then subcultured as pure populations.
The cells routinely studied in monolayer culture can be derived in the
laboratory from primary cultures, or obtained from commercial suppliers
and nonprofit organizations such as the American Type Culture Collection 6 and Institute for Medial Research. 7 The most commonly studied,
well-characterized cell lines include the WI-38 and IMR-90 (human diploid, fibroblastic), 3T3 (mouse embryonic, fibroblastic), and HeLa (hu1 T. G. Pretlow II, Int. Pathol. 16, 42 (1975).
2 R. C. Boltz, Jr., P. Todd, M. J. Streibel, and M. K. Louie, Prep. Biochern. 3, 383 (1973).
a S. F. Schlossman and L. Hudson, J. lmmunol. 110, 313 (1973).
4 F. H. Kasten, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, Jr., eds.), p. 72. Academic Press, New York, 1973.
5 R. D. Cahn, H. G. Coon, and M. B. Cahn, in "Methods in Developmental Biology" (F. H.
Wilt and N. K. Wessels, eds.), p. 493. Crowell (Collier), New York, 1967.
The American Type Culture Collection. Rockville, Maryland, 20852.
r The Human Genetic Mutant Cell Repository. Inst. Med. Res., Camden, New Jersey,
08103.
METHODS IN ENZYMOLOGY,VOL.LVIII
[10]
133
134
BASIC METHODS
[10]
since essential metabolites are rapidly depleted at high cell density. Some
cell types will not replicate beyond confluency and are described as being
contact inhibited. "Contact inhibition" is a widely misused term which
encompasses the phenomena of contact inhibition of movement TM and contact inhibition of replication. 11 These are separate, probably unrelated,
aspects of cell behavior. Contact inhibition of movement describes the
well-documented condition that occurs when the directional movement of
a cell changes upon contact with another cell. Contact inhibition of replication is the cessation of growth due to physical crowding of cells at
confluency. This occurrence is not well documented and in most cases
may be a misinterpretation of a well-known cellular response to nutrient
depletion more correctly described as a density-dependent inhibition of
growth, lz
Cultured cells can become " t r a n s f o r m e d . " Cellular transformation is
a spontaneous or induced change, e.g., viral transformation, in the
characteristics of a cell type expressed as an abnormal karyotype, a
change in morphology, or alteration in cell behavior or growth potential.
Spontaneous cellular transformation often occurs with extended time in
culture. Since a transformed cell population will no longer respond
characteristically to experimental conditions, the age of the cells at the
onset of transformation is an important parameter. The age of a culture is
usually expressed as either its passage number or number of population
doublings. Passage number refers to the number of times the cells have
been subcultured since the time of explantation. This is not a precise
estimate of cell age for it is affected by numerous variables including: the
number of cells replanted at each passage, cell survival after inoculation,
and the cell density (i.e., confluency, semiconfluency) at subcultivation.
The number of population doublings is a more critical estimate of cell age
since it accounts for the time required for the cells to double in number.
The number of population doublings a culture has undergone is calculated
by knowing the number of cells originally planted and the cell number at
the next subcultivation.
Normal diploid cells in vitro have a finite lifespan, while transformed
cells apparently possess unlimited proliferative potential. The process of
in vitro cellular aging is considered to be a deterioration of the cellular
processes that are necessary to support continued replication. As a cultured cell population ages its population doubling time progressively increases until proliferation eventually ceases. During the process of senelo M. Abercrombieand J. E. M. Heaysman,Exp. Cell Res. 6, 293 (1954).
11L. N. Castor, J. Cell. Physiol. 72, 161 (1968).
12M. G. P. Stoker and H. Rubin, Nature (London) 215, 171 (1967).
[10]
135
136
BASIC METHODS
[10]
[10]
137
138
BASIC METHODS
[10]
within an incubator or controlled-temperature room. The bottle is partially filled with medium, and as it turns the cells adherent to its walls are
alternately bathed in the fluid medium and exposed to the gas phase.
Dissociation
[10]
139
3. Add warm enzyme solution (collagenase 0.1%, trypsin 0.1%, chicken serum 1% in Moscona's saline) (1 ml/5 cm ~ culture surface). After 1
min withdraw the solution, leaving only a very thin layer of fluid coveting
the cells.
4. Observe the cells with an inverted phase-contrast microscope; note
distribution and predominant cell morphology.
5. Cap the flask tightly and incubate at 37 for 3-5 min.
6. Again observe the cells with a phase microscope; look for freefloating cells and attached cells which have pulled in their processes and
are rounding up. If the majority of cells do not appear to be detached
return the flask to the incubator.
7. After several minutes of incubation hold the flask horizontally and
give its side a sharp rap with the hand. This will help detach loosely
adherent cells. Often the cells loosen in sheets and these can be seen to
drift across the substrate surface when the flask is tipped at an angle.
Observe the cells by phase microscopy. Repeat if necessary.
8. Add several milliliters of culture medium to the flask. Use a
pipette to gently triturate the cells.
Seeding
9. A viable cell count 25 (dye exclusion) is then performed on the suspension so that other flasks can be seeded at a known cell density.
10. Cell dilutions are made and aliquots seeded to new culture vessels
containing warm (equilibrated) medium.
11. The newly seeded culture vessels are incubated undisturbed to
facilitate cell attachment.
Morphological Evaluation of Monolayer Cultures
Many cultured cells possess distinctive morphological characteristics
observable by phase-contrast microscopy. This enables the investigator
to identify specific cell types in mixed cultures and to carry out procedures such as clonal isolations. Routine observations by phase microscopy are essential for proper assessment of a cell population. The morphology of cultured cells may change in response to slight alterations in
culture conditions. An abnormal morphological change in the cell population is often the first indication that a problem exists within the system.
Therefore, it is useful to frequently monitor culture morphology and to
keep a record of the observations. A Polaroid-type camera back affixed to
an inverted microscope is very useful for this purpose.
25 H. J. Phillips, in '*Tissue Culture: Methods and Applications" (P. F. Krusc, Jr. and M. K.
Patterson, Jr., eds.), p. 406. Academic Press, New York, 1973.
140
BASIC METHODS
[10]
Fixed and stained cell preparations of various types are used for morphological analysis of monolayer cultures. Clonal plates are prepared for
gross visual examination by fixing the cultures in situ (buffered 2.5%
glutaraldehyde) and then staining directly with 0.5% crystal violet. For
light microscopical analysis of monolayer cultures/n situ cells can be
grown on coverslips, fixed with Bouin's solution, stained with hematoxylin and eosin, and mounted on microscope slides. 26 This provides good
resolution, and cell structure is well preserved.
Critical morphological analysis of cultured cells by high-resolution
light microscopy and transmission electron microscopy necessitates embedding and sectioning of the cultures. The cells need not be removed
from their substrate for processing, and all steps through embedding can
be carded out within the culture vessel. Since monolayer cultures are only
one to several cell layers in thickness, fixation and dehydration are rapid,
and infiltration with resin is uniform. Convefitional preparation methods
can be used with the precaution of avoiding solvents that might dissolve
the tissue culture vessel. Processing cells in situ does not hinder sectioning
since resin will infiltrate between the cells and substrate allowing them to
be separated following polymerization. 27 The tissue block can then be
oriented for enface or transverse sectioning. Certain types of resins may
work better with specific brands of plastic culture ware. One reliable
combination is the use of an Epon mixture 28 with Lux or Coming plastic
surfaces. Processing cells grown in glass dishes is more difficult because
polymerized resin does not readily separate from the dish. Cells grown on
glass coverslips are easier to work with. If the coverslip is embedded so
that resin contacts only the side which bears cells the coverslip can be
separated from the polymerized plastic by immersing them repeatedly in
liquid nitrogen and then boiling water.
The preparation of cultured cells for scanning electron microscopy
presents no unusual difficulties. Processing by the critical-point drying
method is recommended. 29 One necessary precaution is to assure that the
cells are grown on a substrate that can be conveniently placed in the
chamber of the critical-point dryer. Alternatively, pieces of the culture
surface must be cut from the culture vessel without damaging the cells.
The cells may then be fixed, critically point dried, and coated by routine
methods.
26 j. Fogh and J. A. Sykes, In Vitro 7, 206 (1972).
27 W. H. J. Douglas, E. P. Dougherty, and G. W. Phillips, ~ssue Cult. Assoc. Man. 3, 581
(1977).
2s C. C. Haudenschild, R. S. Cotran, M. A. Gimbrone, Jr., and J. Folkman, J. Ultrastruct.
Res. 50, 22 (1975).
29 K. R. Porter, D. Kelley, and P. M. Andrews, Proc. Annu. Stereoscan Syrup., 5th, 1972 p.
1 (1972).
[11]
MEASUREMENT
OF G R O W T H
AND VIABILITY
141
[11 ] M e a s u r e m e n t o f G r o w t h a n d V i a b i l i t y o f Cells in C u l t u r e
By MANFORD K. PATTERSON,JR.
142
BASIC METHODS
[11]
TABLE I
BALANCED SALT SOLUTIONS a
CaCI2
CaCI2 2 H20
KC1
KH2PO4
MgCI~ 6 H20
MgSO4 7 H20
NaCl
NaHCOa
Na~HPO4 7 H20
NaH~PO4 - H20
Glucose
Phenol red
Dulbecco b
Earlec
Geyd
Hankse
0. l0g
0.20
0.17g
0.14#
0.20
0.20
0.I0g
0.40
0.37
0.03
0.21
0.07
7.00
2.27
0.226
8.0
0.20
6.80
2.20
2.16
0.14
1.00
0.01#
i.00
Puck Ff
Puck Gs
0.40
0.06
0.016
0.285
0.083
0.016
0.40
0.150
0.20
8.00
0.33
0.09
0.154
7.40
1.20
0.29
0.154
8.0
1.00
0.01#
1.10
0.0012
1.10
0.0012
0.29
a Usually prepared as a x 10 solution and diluted prior to filtration. Cell suspensions are
generally washed with calcium- and magnesium-free solutions. Concentrations given
are g/liter, x 1 solution.
bSterilize by autoclaving; R. Dulbecco and M. Vogt, J. Exp. Med. 99, 167 (1954).
cSterilize by filtration; J. C. Bryant, Tissue Cult. Assoc. 1, 185 (1975).
d Sterilize by filtration; G. O. Gey and M. K. Gey, Am. J. Cancer 27, 45 (1936).
e Sterilize by filtration; J. H. Hanks, Tissue Cult. Assoc. 1, 3 (1975).
rSterilize by filtration; T. T. Puck, S. J. Ciecura, and A. Robinson, J. Exp. Med. 108,
945 (1958).
v Prepare as separate solution and 'mix prior to use; may be omitted if solution is to be
used for wash.
b l e m a t e r i a l s . P r o p e r h a n d l i n g o f c u l t u r e s r e q u i r e s c a r e f u l a t t e n t i o n to p H ,
t e m p e r a t u r e , a n d o s m o t i c p r e s s u r e d u r i n g w a s h i n g , a n d a b a l a n c e bet w e e n a d e q u a t e r e m o v a l o f g r o w t h m e d i a w i t h o u t loss o f cells.
Choice of washing solutions appears to be a simple matter of prefere n c e . T h e b a s i c f o r m u l a s f o r t h e m o s t c o m m o n l y u s e d are g i v e n i n T a b l e I.
I n g e n e r a l t h e c h o i c e s h o u l d b e d i c t a t e d b y t h e salt s o l u t i o n u s e d as the
b a s e for the g r o w t h m e d i u m . M o r e o v e r , c a l c i u m - f r e e salt s o l u t i o n s are
g e n e r a l l y p r e f e r a b l e f o r all cell s u s p e n s i o n s s i n c e this d i s c o u r a g e s cell
a g g r e g a t i o n . A c o m p r e h e n s i v e listing o f b a l a n c e d salt s o l u t i o n s h a s b e e n
published, z
Growth Measurements
Visual Methods
T h e m o s t c o m m o n l y u s e d m e a s u r e m e n t o f g r o w t h is d i r e c t e n u m e r a t i o n o f cells e m p l o y i n g a h e m o c y t o m e t e r . T h i s m e t h o d is b e s t a p p l i e d
n c. Waymouth, in "Cell Biology" (P. L. Altman and D. D. Katz, eds.), Vol. I, p. 61.
FASEB, Bethesda, Maryland, 1976.
[11]
143
Reagents
Citric acid-O.1 M (1.9212 g/100 ml distilled water) to which a few
crystals of thymol are added
Crystal violet----0.2% dissolved in 0.1 M citric acid
Balanced salt solution--see Table I
Procedure. Monolayer cultures are rinsed 3 times with balanced salt
solution (this is optional depending upon media and subsequent use, e.g.,
if citric acid extract is to be used also for protein determination) and
4 M. Absher, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, Jr., eds.), p. 395. Academic Press, New York, 1973.
144
BASIC METHODS
[1 1]
drained. Two milliliters of 0.1 M citric acid are added to a T-255 flask (5.0
ml for T-75) and allowed to stand at room temperature. The time can vary
but a minimum of 10 min has been found adequate for disruption of all
cells that have been tested. Frequent shaking of the flask during incubation can aid in disrupting the cells but is not always necessary, since the
final suspension is prepared by scraping the flask surface.
Note: Protein analysis may be performed on this extract by adding
Lowry's reagent directly to an aliquot (see "Chemical Methods").
A 0.1-ml aliquot is diluted to 1.0 ml with 0.1 M citric acid and 1.0 ml of
the crystal violet solution added. The resulting suspensions of muclei have
been found stable for up to 1 week at 4 .
The suspended nuclei are counted in a hemocytometer; calculations
are made as described.
Chemical Methods
The relationship of protein and nucleic acid biosynthesis to the growth
of cells has led to the measurement of th~se components as an expression
of cell growth. However, variations in their content occur, and a linear
relationship is not always found. The validity of this type of assay should
be established by comparison with the more direct visual methods.
PROTEIN DETERMINATION
Oyama and Eagle s were the first to adapt the Lowry 7 assay for protein
to measurement of cells in tissue culture. Subsequent studies have shown
wide variations in ratio of protein per cell values (Table II).
Reagents
Balanced salt solution--see Table I
Lowry solution A---20 g Na~CO3 and 4 g N a O H pellets dissolved to
give a final volume of 1 liter (store in plastic bottle)
Lowry solution B--Equal parts of 1% CUSO4.5 H~O and 2.7%
sodium potassium tartrate
Lowry solution C m l 0 0 ml solution A, 2 ml solution B
Phenol reagent (Folin and Ciocalteau)ml N (usually purchased as a 2
N solution)
Human serum albumin, crystallized--Stock solution containing 200
/zg/ml
s Tissue culture flasks are available in various sizes constructed of glass or plastic. The
"25" represents 25 cm ~ surface area.
e V. I. Oyama and H. Eagle, Proc. Soc. Exp. Biol. Med. 91, 305 (1956).
r O. H. Lowry, N. J. Rosebrough, A. L. Farr, and R. S. Randall, J. Biol. Chem. 193, 265
(1951).
[11]
145
TABLE II
PROTEIN CONTENT OF CELLS IN TISSUE CULTURE
Days after plating
3
References
480
500
450
500
370
330
350
330
b
o
930
810
890
1010
850
960
920
780
1010
740
640
790
940
700
320
360
220
175
800
710
300
350
180
150
400
720
320
410
175
150
390
400
310
400
175
100
872
860
787
678
498
588
545
446
Cell line
L-5 (monolayer)
L-5 (suspension)
HeLa
Human leukemia
Intestinal epithelium
WI-38
Jensen sarcoma
WISH
HEp-2
Chang (suspension)
HeLa (suspension)
CF-3
IMR-91
Human fetal lung
20-29 doublings
30- 39 doublings
40-49 doublings
50- 59 doublings
320 a
350
550
500
pg/cell
1150
1100
350
390
400
230
c
c
c
410
390
320
350
a
a
d
d
e
s
s
g
490
480
883
1287
Italicized values are extrapolated from curves; data given in picograms per cell.
b A. Tsuboi, T. Kurotsu, and T. Terasima, Exp. Cell Res. 103, 257 (1976).
c V. I. Oyama and H. Eagle, Proc. Soc. Exp. Biol. Med. 91, 305 (1956).
d E. Miedema and P. F. Kruse, Jr., Biochem. Biophys. Res. Commun. 20, 528 (1965).
e p. Voipe and T. Eremenko-Volpe, Cur. J. Biochem. 12, 195 (1970).
i R. T. Dell'Orco, unpublished.
g E. L. Schneider and S. S. Shorr, Cell, 6, 179 (1975); days not specified; cell cultures
either in logarithmic growth (<1.3 104 cells/cm~) or when the monolayer reached
light confluency (1.3-2.6 104 cells/cm2).
146
BASIC METHODS
[11]
2- to 3-ml aliquots are required; a deep blue color suggests a 1:2 or 1:10
dilution with solution C. A final volume of 5 ml of sample in solution C
plus 1 ml distilled water or standard (0-200/zg) receives 0.5 ml of phenol
reagent, jetted to obtain rapid admixture. A digest from cell-free controls
is best used as a blank. Thirty minutes after addition of the phenol reagent, the absorbance of the samples is measured with a spectrophotometer at 660 nm. Color is stable for at least 2 hr. Absorbance is compared
with the absorbance values of standards in the range of 0-200/zg.
Limitations. A large number of compounds including organic buffers,
sucrose, s glycerol, s,9 Tris, ~ and certain reducing agents u interfere with
the Lowry protein method, and special sample treatment may be necessary when such reagents are used in cultures.
D N A DETERMINATION
As with protein, the DNA content per cell depends upon the stage of
growth and the phase of the cell cycle.n-Is It is therefore advisable to
equate the results of DNA determination with other methods of evaluation. The procedures in general use for tissues are adapted to cells derived
from cell culture systems. Several critical steps exist in the estimation of
nucleic acid, TM the majority of which concern extraction and hydrolysis.
Since one is dealing with small amounts of tissue, special care must be
exercised if quantitation is to be achieved. The disintegration of the cell
sample is a critical step in obtaining an accurate determination of nucleic
acids because of the widespread presence of nucleases. Two general procedures exist: (1) removal of the cells from the solid matrix, or (2) in situ
extraction. An example of the former uses a freeze-thaw procedure which
separates a particulate fraction containing the cellular DNA and a soluble
fraction containing cytoplasmic enzymes TM prior to removal of interfering
substance by cold acid precipitation of the nucleic acids. An example of in
situ extraction can be found in the procedure to be described (cf. also
Setaro and MorleylZ).
s A. Bensadoun and D. Weinstein, Anal. Biochem. 70, 241 (1976).
g P. Bl'fimel and W. Uecker, Anal. Biochem. 76, 524 (1976).
to R. Rej and A. H. Richards, Anal. Biochem. 62, 240 (1974).
11 F. Higuchi and F. Yoshida, Anal. Biochem. 77, 542 (1977).
lz E. L. Schneider and S. S. Shorr, Cell 6, 179 (1975).
la A. Leyva, Jr. and W. N. Kelley, Anal. Biochem. 62, 173 (1974).
14 V. J. Cristofalo and D. Kritchevsky, Med. Exp. 19, 313 (1969).
t5 R. Yanishevsky, M. L. Mendelsohn, B. H. Mayall, and V. J. Cristofalo, J. Cell. Physiol.
84, 165 (1974).
16 H. N. Munro and A. Fleck, Methods Biochem. Anal. 14, 113 (1966).
1 F. Setaro and C. G. D. Morley, Anal. Biochem. 71, 313 (1976).
[11]
147
Reagents
Balanced salt solution--see Table I
Perchloric acid, 1.0N; dilute 8.62 m170-72% reagent grade to 100 ml
Sodium hydroxide, 0.3 N; 1.2 g/100 ml
Diphenylamine--dissolve 1.5 g in 100 ml glacial acetic acid; add 1.5
ml concentrated H2SO4; mix fresh and store in dark until used
Acetaldehyde, 16 mg/ml; dilute 0.5 ml to 25 ml with distilled water;
store at 4
Burton reagent--just before use add 0.1 ml of acetaldehyde solution
to each 20 ml of diphenylamine reagent required
DNA standard--sodium desoxynucleate, dissolve 0.2 mg/ml of 5 mM
NaOH; store at 4; mix equal volume with 0.5N perchloric acid for
a final concentration of 100/.~g/ml
Extraction Procedure (for T-25flask). Wash cell layer 3 times with balanced salt solution. Wash once with 5 ml cold 0.2N perchloric acid. Add 5
ml cold 0 . 2 N perchloric acid and let stand 10 min at 4; decant. Add 2 ml
of 0.3N N a O H and let stand 1 hr at 37; transfer to 15-ml centrifuge tube
and cool in ice. Add 2 ml of cold 1.0N perchloric acid and let stand 10 min
at 4 to precipitate DNA and protein; centrifuge (supernatant contains
RNA fraction). Wash the pellet with 2 ml cold 0.2 N perchloric acid;
centrifuge. Resuspend pellet in 2 ml of 0.5 N perchloric acid and heat in
water bath for 15 min at 90 to hydrolyze DNA. Centrifuge and retain
supernatant liquid. Wash the pellet with 2 ml 0.5 N perchloric acid; centrifuge and combine supernatant fluid with that obtained in the immedila B. T. Hill and S. Whatley, FEBS Lett. 56, 20 (1975).
19 B. T. Hill, Anal. Biochem. 70, 635 (1976).
z0 U. Karsten and A. Wollenberger, Anal. Biochem. 46, 135 (1972).
21 U. Karsten, Anal. Biochem. 77, 464 (1977).
22 M. J. Blackburn, T. M. Andrews, and R. W. E. Watts, Anal. Biochem. 51, 1 (1973).
2a W. Y. Fujimoto, J. Teague, and R. H. Williams, In Vitro 13, 237 (1977).
24 H. L. Hosick, Tissue Cult. Assoc. Man. 1, 45 (1975).
148
aASXC METHODS
[1 1]
TABLE III
DNA CONTENT OF CELLS IN TISSUE CULTURE
Cells
DNA (pg/cell)
Method
Reference
9.6 +- 3.7
9.7 _+ 2.8
10.6 -+ 3.0
8.4 _ 2.5
7.4
9.8
7.5
7.94 ___ 0.55
9.0 + 0.4
8.3 -+ 0.45
8.5
8.0 -+ 0.1
7.5 _+ 0.4
7.8 +- 0.4
7.5 -+ 0.27
8.62 -+ 0.67
13.7
Colorimetric
Colorihaetric
Human iymphocytes
Granulocytes
Granulocytic leukemic
3T3 mouse
Colorimetric
Fluorometric
Fluorometric
Fluorometric
Colorimetric
Absorbance
Fluorometric
Fluorometric
Absorbance
Absorbance
d
:
g
ately preceding step. (Note: The pellet may be dissolved in 0.3 N NaOH
and used for protein determination.)
Analytical Procedure (Colorimetric-Burton). Standard curve: Prepare
duplicate standards of 5, 10, 25, and 50/zg of D N A per tube by adding
0.05, 0.10, 0.25, and 0.50 ml of working standard (100/~g/ml) to individual
tubes and adding 0 . 5 N perchloric acid to a final volume of 2.0 ml. Prepare
a standard blank of 2.0 ml 0.5 N perchloric acid. To 2 ml of tissue extract
and tubes containing the D N A standards, add 4.0 ml of Burton's reagent.
Incubate 16-20 hr at room temperature. Read absorbance at 600 nm and
calculate by comparing with the absorbance of the D N A standards.
Recent modifications suggest substitution of paraldehyde for acetaldehyde and its inclusion with diphenylamine to produce a more stable
[11]
149
Efforts to enumerate cells in situ have been directed mostly toward the
use of individual frames from time-lapse cinematography: a4,a~ Others
have enumerated cells in a microscopic field, ao-as These methods are limited to low cell counts.
25 G. M. Richards, Anal. Biochem. 57, 369 (1974).
26 j. R. Decalionne and J. C. Weyns, Anal. Biochem. 74, 448 (1976).
27 M. Harris, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, Jr., eds.), p. 400. Academic Press, New York, 1973.
zs W. F. Daly, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, Jr., eds.), p. 398. Academic Press, New York, 1973.
29 M. E. Kaighn and D. J. Merchant, Tissue Cult. Assoc. Man. 3, 649 (1977).
30 H. A. Crissman, P. F. Mullaney, and J. A. Steinkamp, Methods Cell Biol. 9, 179 (1975).
31 W. P. Drake, P. C. Ungaro, and M. R. Mardiney, Transplantation 14, 127 (1972).
a2 H. A. Crissman, M. S. Oka, and J. A. Steinkamp, J. Histochem. Cytochem. 24, 64 (1976).
aa C. Waymouth, J. Natl. Cancer Inst. 17, 305 (1956).
a4 L. N. Castor, Exp. Cell Res. 68, 17 (1971).
as G. Froeze, Exp. Cell Res. 65, 297 (1971).
36 C. W. Jamieson, D. Russin, E. Benes, C. W. DeWitt, and J. H. Wallace, Nature (London)
222, 284 (1969).
37 G. Martinez-Lopez and L. M. Black, In Vitro 9, 1 (1973).
as G. F. -Y. Lee and D. L. Engelhardt, J. Cell. Physiol. 92, 293 (1977).
150
BASIC METHODS
[11]
OTHER
Measurements of Viability
Numerous criteria are used to determine cell viability. A survey by
Malinin and Perry 41 listed 40 viability assay methods. These may be
grouped into six broad categories: (1) survival and growth in tissue culture, (2) membrane integrity, (3) metabolite incorporation, (4) enzyme
spectrum, (5) trafisplantation potential, and (6) structural alteration,
chemical composition, and electroconductivity.
The ability of cells to withstand the rigors of dispensing agents,
changes in environment, freezing, and thawing is indicative of cell integrity but is not sufficiently quantitative for description. Beating heart cells
39 C. Peraino and W. J. Eisler, Jr., in "Tissue Culture: Methods and Applications" (P. F.
Kruse, Jr. and M. K. Patterson, Jr., eds.), p. 351. Academic Press, New York, 1973.
40 D. J. Merchant, R. H. Kahn, and W. H. Murphy, in "Handbook of Cell and Organ
Culture," p. 160. Burgess, Minneapolis, Minnesota, 1964.
~1 T. I. Malinin and V. P. Perry, Cryobiology 4, 104 (1967).
[11]
151
152
BASIC METHODS
[12]
[12] C l o n i n g
By LOLA C. M. REID
A. Introduction
A cloned cell population is one derived from a single parental cell.
Cloned populations provide the experimenter with the distinct advantage
of minimal genetic variability in the model system under study. Methods
for isolating single cells and propagating them into a population of cells
are called cloning, and the majority of them fall into one of three categories of techniques:
1. Dilution Plating. Suspensions of cells are diluted with a sufficient
volume of medium to permit addition of single cells to dishes.
2. Cloning of Anchorage-Dependent Cells. Dilute concentrations of cells
are seeded onto one of several possible substrates. The cells are allowed to
attach and to grow into colonies of cells. The colonies of cells are then
individually subcultured and transferred to other dishes.
METHODS IN ENZYMOLOGY,VOL. LVIII
[12]
CLONING
153
i54
BASIC METHODS
[12]
Equipment
Desk-top centrifuge
Inverted phase microscope with a total magnification of x 50 to x 100
Incubator, maintained at 37 with a water-saturated atmosphere containing 5% CO2 in air
Biogaard or laminar flow hood. The hood minimizes contamination of
cultures. If a hood is not available, one can do reasonably well with
an inexpensive, plastic-sheltered area in which an ultraviolet
light is placed to minimize pathogens.
Hemocytometer for counting cells. Cell counting also can be done
with a Coulter counter if it is available.
Bunsen burner
Supplies
Plastic culture dishes and flasks can be obtained from a number of
suppliers. However, the plating efficiency and growth of cells may
vary somewhat on the plastics from different commercial suppliers.
Therefore, one should select plastics from one company and use
them exclusively for all experiments in which data are to be compared.
Pipettes:
Sterile Pasteur pipettes (7" and 9"). Some pipettes should be
plugged with cotton prior to sterilization.
Sterile 1- and 5-ml volumetric glass or plastic pipettes
Sterile test tubes holding 10 ml
Basal media: See Table I for a listing of possible cloning media.
Serum: Fetal bovine serum is most commonly used in cloning since it
has high concentrations of growth factors and pregnancy hor14K. K. Sanford, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and
M. K. Patterson, eds.), pp. 237-241. Academic Press, New York, 1973.
15 A. C. Hildebrandt, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and
M. K. Patterson, eds.), pp. 244-254. Academic Press, New York, 1973.
16 p. K. Dougall, in"Tissue Culture: Methods and Applications"(P. F. Kruse, Jr. and M. K.
Patterson, eds.), pp. 261-264. Academic Press, New York, 1973.
[12]
CLONING
155
TABLE I
CLONING MEDIAa
Name
Eagle's Minimal
Essential Medium (MEM)
Ham's Cloning Media
F10
F12
F12M
F12K
MCDB104
RPMI
CMRL- 1066
Medium 199
Use
References
2~
5-9
4
6
a
9
20
~1
n
a The compositions of the media listed can be found in this volume [5] and in the
specific references noted. Media should be freshly prepared every 2-3 weeks and
stored at 4. Longer storage results in the loss of labile components such as
glutamine. Selection of a medium for a call population to be cloned should be
made after assaying the plating efficiency and growth behavior of the cells in the
medium. Ham's F12K medium, in particular, has proven to be an excellent cloning
medium for many cell types and is used routinely in many laboratories.
156
aASl METHODS
[12]
Procedures
1. The cell cultures to be cloned should be recently subcultured, be in
an active state of growth, and show no signs of ill health.
2. Cloning of freshly explanted normal or neoplastic tissue is difficult
and almost always requires use of feeder layers or conditioned media
(techniques presented in subsequent sections). Tissue or tumor to be
cloned should be sterilely dissected from the animal.
3. Single cell suspensions are prepared from monolayer cultures as
follows. Remove the medium from the culture plates and rinse the plates
twice with PBS. Add 1 ml of the trypsin solution to a 60-mm plate (2-3 ml
for larger plates or flasks) of cells and incubate the plates at 37 for 5-15
is A. Lwoff,R. Dulbecco, M. Vogt, and M. Lwoff,Virology 1, 128 (1955).
1~j. A. Robb, Science 170, 857 (1970).
~oj. A. Robb, in "Tissue Culture: Methods and Applications"(p. F. Kruse, Jr. and M. K.
Patterson, eds.), pp. 270-274. AcademicPress, New York, 1973.
[12]
CLONING
157
min. The plates should be watched carefully to ensure that the time of
contact with trypsin is minimized. As soon as the majority of cells are
rounded and mostly detached from the plates, squirt the ceils with the
conditioned medium supplemented with serum. Most serum contains a
trypsin inhibitor (chicken serum is an exception). By squirting the cells
with the medium, trypsin is inactivated and detachment of the cells from
the plates is completed. To completely inactivate trypsin requires a 9:1
dilution of the trypsin solution with serum-supplemented medium. One
can also remove trypsin by centrifuging the cells, removing the supernatant fluid, and resuspending the ceils in the cloning medium.
Single cell suspensions are prepared from solid tissues, either normal
or neoplastic 42 (see this volume [9]), as follows. The dissected tissue is
minced finely with scissors sterilized by dipping them in 95% ethanol.
Treat the mince with 0.1% collagenase (Sigma) in PBS at a ratio of 10 ml
of enzyme solution to 1 ml of mince. Enzymic digestion should take place
in a shaker bath at 37. After 15 min of enzymic treatment, allow the
chunks of tissue to settle to the bottom of the test tube, and pipette the
supernatant fluid, containing suspended cells, into another test tube. Centrifuge the cells in a desk-top centrifuge at 900 rpm. Resuspend the pellet
in cloning medium. The chunks and tissue mince can be repeatedly
treated with collagenase solution until they are totally disaggregated.
4. Count the cells by the trypan blue exclusion assay in order to
determine the number of viable cells (see this volume [11]!.
5. Dilute the cell suspensions, whether from the monolayer culture or
from the solid tissue dispersion, to a concentration of 10 cells/ml.
6. Use 0.1 ml of the medium containing the cells per well. Repeatedly
agitate the cell suspension to ensure homogeneous dispersion of the cells
throughout the medium. For rapid inoculation of cells into microtiter
wells, a Hamilton repeating dispenser (Hamilton Company, Whittier, California, # PB600-1) is useful. 2
7. Carefully observe each well and score those containing only one
cell.
8. Incubate the cultures in 37 incubators with 5% CO2 in air and ~vith
95% relative humidity.
9. During the growth of the colonies, it should be unnecessary to
change the medium. However, if colony formation is unusually slow, and
a medium change is required, the spent medium can be gently aspirated
from the cultures, and fresh medium added. Leave a residue of fluid over
the cells while changing medium to prevent desiccation of the cells.
10. When colonies of 500-600 cells have formed, usually in 2-3
weeks, remove the medium from the wells. Rinse twice with phosphate
buffered saline. Add 0.1 ml of trypsin solution per well. Allow the cells to
158
BASIC METHODS
[12]
round, and then add cloning medium with 10% serum to suspend the cells.
Transfer the cells to an appropriate tissue culture dish (35-60-mm plate)
or flask.
D. Cloning of Anchorage-Dependent Cells
The techniques in this category are slight variations of the ones described above in "Dilution Plating Techniques." The preparation and
dilution of single cell suspensions are the same as described there. The
cells are seeded onto 60-ram tissue culture dishes or on 60-ram petri dishes
coated with either collagen or fibrin. By seeding only 100-200 cells per
plate, one can obtain large colonies of cells that are sufficiently separated
to permit isolation of individual colonies.
The technique can be used only for cells that require anchorage and
spreading on a substrate in order to grow. This includes most normal cells
as well as many established cell cultures.
Procedures
1. Cells vary as to the substrates to which they will anchor. Established cells lines will attach to plastic coated with polycations. However,
freshly explanted tissues usually prefer either collagen-coated plates or
fibrin-coated plates. Preparation of collagen-coated plates is given elsewhere in this volume [21]. Fibrin-coated plates are prepared as follows:
Prepare a solution of cloning medium containing 0.12 units of thrombin
per 100 ml of medium. Prepare a solution of 250 mg bovine fibrinogen, 800
mg sodium chloride, and 25 nag sodium citrate per liter of glass-distilled
water. Sterilize by filtration. Add 1 ml of the fibrinogen solution and 4 ml
of the medium containing thrombin to a tissue culture dish and mix rapidly
with a sterile Pasteur pipette. A clear gel will form within several minutes.
This technique was first introduced by Schindle& 1 who used it to replace
21 R. Schindler, M. Day, and G. A. Fischer, Cancer Res. 19, 47 (1959).
[12]
CLONING
159
agar cloning (described in the next section). Schindler suspended the cells
in fibrin and permitted them to form colonies shaped like morulae. However, one can also attach the cells to the surface of the fibrin and observe
colony formation.
2. Prepare single cell suspensions from monolayer cultures or from
tissues by the methods given in "Dilution Plating Techniques."
3. Depending on the plating efficiency of the cells, select an appropriate concentration of them to yield ultimately 1-10 colonies per plate. For
example, with a plating efficiency of 5-10%, one should plate 100 cells per
plate.
4. Dilute the cell suspensions to an appropriate density so as to add
the correct number of cells per 60-mm dish. The 60-mm dishes hold about
5 ml of medium.
5. The plates are incubated at 37 in 5% CO2 in air and with 95%
relative humidity.
6. The following day, single cells that are sufficiently isolated to permit easy cloning are marked by encircling them on the plastic with a
grease pencil or felt pen.
7. The cells are allowed to grow for several weeks into colonies containing approximately 500-1000 cells.
8. Remove the medium from the plates and rinse them twice with
PBS. Leave a residue of the PBS over the cells to prevent their drying.
9. With forceps sterilized by dipping them into 70% ethanol, dip previously sterilized cloning rings into the petri dish containing stopcock
grease. Place the grease-coated end down against the plastic so that the
ring encircles a colony of cells and so that the greased end forms a seal
with the plastic.
10. Add enzyme solution, e.g., 0.1% trypsin, to each of the rings.
Usually 1-2 drops of solution will fill the rings.
11. Incubate the cultures for 5-15 min at 37, observing the cultures
periodically with a phase microscope. When the cultures are rounded and
ready to detach, use a 9" Pasteur pipette and pipette the solution gently
up and down to complete the detachment process.
12. Pipette the solution of suspended cells into a 35-mm tissue culture
dish or 35-mm petri dish coated with collagen or fibrin. Add 2 ml of
cloning medium per plate.
E. Cloning of Cells that Are Anchorage-Independent for Growth
Most cells require anchorage to and spreading on a substratc for
growth. However, some cells have acquired the ability to grow in suspension cultures. Factors contributing to this cellular capability include viral
transformation (SV40, polyoma), infection of cells with mycoplasma, and
160
BASIC METHODS
[12]
[12]
CLONING
161
Procedures
1. Agar stock. Add 12.5 g of Difco agar to 800 ml of boiling glassdistilled water and heat until the agar dissolves. Make up to 1 liter with
water. Sterilize by autoclaving 80-ml portions of the agar solution in
screw-capped bottles of 250-ml capacity. Autoclave under low pressure
and with the screw caps loosened. Allow the bottled solutions to come to
room temperature. Then tighten the bottle caps. Store at room temperature.
2. Melt the agar stock solution by placing the bottle in boiling water
until the agar liquefies. Cool for several minutes and place in a water bath
at 44 so that the hot water is above the level of the agar.
3. Prepare the complete agar medium by mixing 80 ml of the cloning
medium (double strength) with 20 ml tryptose broth and 20 ml serum in
the bottle with the agar. Mix by swirling.
4. Pipette 1-2 ml of medium into 60-ram petri dishes or tissue culture
dishes and swirl the plate to evenly coat the solution over the bottom of
the dish. Allow the base layers to solidify (10-15 min).
5. Prepare a cell suspension as described under "Dilution Plating
Techniques."
6. Dilute the cell suspension with cloning medium to a concentration
that is 3-fold that of the final, desired concentration. Mix 1 volume of the
cell suspension with 2 volumes of the complete agar solution.
7. Spread the cells over the base layer and swirl the plate to assure
even spreading.
8. Incubate the cultures at 37 in an atmosphere of 5% CO2 in air and
with 95% relative humidity.
9. Allow the cells to form colonies of 500-1000 cells.
10. Colonies are transferred by sucking them from the agar with a
Pasteur pipette and transferring to a sterile test tube containing 1 ml
of medium. The colony of cells is pipetted to dissociate it from the
agar.
11. The cell suspension is transferred to a 35-mm dish to which is
added 2 ml of cloning medium.
F. Preparation of Cloning Media, Feeder Layers, and Conditioned Media
A listing of some of the media used for cloning various cell types is
provided in Table I. All of them employ a basal medium enriched in
162
BASIC METHODS
[12]
Feeder Layers
The low plating efficiency of some cells, especially freshly explanted
cells, prevents or seriously complicates the cloning procedure. As noted,
the low plating efficiency is due in part to the loss of essential nutrients
from the isolated cells. However, in the special cloning media listed in
Table I these nutrients are provided. The other factors, available in highdensity cultures of cells but not in low density cell cultures, are "chemical
messengers" secreted by tho ceils and regulating survival and/or growth.
The chemical messengers playing roles in colony formation are referred to
as Colony Stimulating Activity factors (CSA). To provide such factors, it is
necessary to co-culture the cells to be cloned with a feeder layer of the
same or another ceil type. Many established cell lines requiting feeder
layers under clonal conditions can use feeder layers of the same cell type.
Freshly explanted epithelial cells, whether from normal or neoplastic tissue, often do better when cloned over feeder layers of mesenchymal ceils
(L. C. M. Reid, unpublished data). Many of the CSA factors have been or
are being purified in a number of laboratories. Several of the bettercharacterized CSA are listed in Table II. al-a7 Except for some anaplastic
cell lines, supplementation with only the CSA factors is inadequate for
colony formation, suggesting that other factors are necessary. Corroborating this interpretation are the findings by Sato and his colleagues
that multiple hormones, growth factors, and transfer factors are essential
as supplements to basal media to attain growth of many ceil types 17 (see
this volume [6]).
[12]
CLONING
163
.8
"
.~
"
<
<
<
c~
'~
..~
r,
0
Z
Z
o
c~
0
Z
0
,.d
0
r..)
0
"~V-
~ra
co
0
Z
<
~o
i ~- i ~
<
m
r,..)
,~
.~ ..~
z
"~
ell
o -~
rj
o .=
r..)
164
BASIC METHODS
[13]
Procedures
[13] C e l l C u l t u r e C h a r a c t e r i z a t i o n : M o n i t o r i n g for C e l l
Identification 1
By W A R D D.
F.
SIMPSON,
and
BHARATI HUKKU
Techniques and cell culture support systems now available enable any
bioscience laboratory to successfully propagate animal cells in vitro.
While the relative ease with which cell cultures can now be started or
Supported in part by National Cancer Institute Contract NO1 CP 3-3333.
[13]
165
166
BASIC METHODS
[13]
(1969).
[13]
167
procedures used are readily carded out. Use of both isozymes give sufficient information to determine most cell species. 15
A. Preparation of Isozyme-containing Extracts
Reagents
Trypsin-EDTA solution:
1:300 Trypsin, crude, 2.5 g/1
EDTA (ethylenediaminetetraacetic acid), 0.2 g/1
Glucose, 1.0 g/1
NaC1, 1.0 g/1
NaHCO3, 1.89 g/1
Procedure
1. Grow 5 x 106 cells in appropriate medium and disperse with
trypsin-EDTA if it is an adherent cell line. When cells are monodispersed,
stop the action of trypsin-EDTA by adding growth medium and collect
the cells in a conical centrifuge tube.
2. Centrifuge the cell suspension at 1500 rpm for 10 min, remove the
supernate, and wash the cells 3 times with 10 volumes of 0.9% NaC1.
3. After the last washing, resuspend the cells in an equal volume of
0.9% NaC1. Freeze the cell suspension by placing the centrifuge tube in a
methanol---dry ice solution. When frozen remove the tube and thaw the
cell suspension slowly at room temperature. Repeat the procedure 3
times. This rapid freeze-thaw procedure will disrupt cells.
4. Centrifuge the suspension at 3000 rpm for 10 min and draw up the
supernatant liquid in a Pasteur pipette. Place the sample in a closable 12 x
75 mm tube for storage at - 2 0 . The isozymes are stable for several weeks
when stored in this manner.
B. Electrophoresis of Sample
Reagents
Glucose 6-phosphate dehydrogenase buffer, 0.075 M Tris-citric acid,
pH 7.5:
Tris, 9.08 g/1
EDTA, 1.49 g/1
Citric acid, 15.75 g/1
Add 20 ml of citric acid solution to 1 liter of Tris-EDTA solution.
15j. E. Shannon and M. L. Macy, in "Tissue Culture: Methods and Application" (P. F.
Kruse, Jr. and M. K. Patterson, Jr., eds.), p. 804. AcademicPress, New York, 1973.
168
BASIC METHODS
[13]
10 mg/ml
25 mg/ml
2 mg/ml
2 mg/ml
0.5 M
0.075 M
Proportion
in stain
0.4
0.6
1.0
1.0
0.4
5.0
ml
ml
ml
ml
rnl
ml
toj. K. Turner model 310-211 with electrophoresis chamber 310-120 is used in this
laboratory.
~7 Universal electrophoresis film, agarose; Coming ACI, Palo Alto, California.
[13]
169
PMS solution is made fresh each day and should not be exposed to
light. NAD, glucose 6-phosphate, and NBT solutions are stable at 4
for 1 week. The stain is bright light yellow and will not react if the
color darkens. Prepare stain and incubate for 15 min at 37 immediately
before use.
L D H stain:
Amount/
solution
Proportion
in stain
NAD
10 mg
10 mg
Sodium lactate
1M
1.0 ml
Nitro blue tetrazolium
1 mg/ml
3.0 ml
Phenazine methosulfate
1 mg/ml
0.3 ml
Barbital buffer, pH 8.8
0.5 M
1.0 ml
Stain does not require preincubation before use.
Procedure
1. Prepare stain as indicated. Pour into a clean shallow staining dish.
Remove electrophoresis film, and place agarose side directly onto stain so
that stain comes in contact with area to which isoenzymes have migrated.
2. Incubate the staining dish in a humidified chamber at 37. Color
should be observed within 5 min, and the staining reaction can be developed for 20 min. The staining reaction should be followed every few
minutes so that overstaining, with consequent blurring of reaction bands,
does not occur.
3. When the staining reaction is satisfactory, remove the electrophoresis film from the staining dish and place it in 1:40 formaldehyde
for 10 min to stop the reaction.
4. Comparative electrophoretic mobilities can be determined immediately by removing the film from the formaldehyde solution, shaking the
film to remove moisture, and overlaying it on lined or graph paper. By
measuring the distance from the origin to the reactive enzyme site, and by
noting the patterns, comparison of enzyme mobilities with known and test
isoenzyme extracts can be carried out.
5. A permanent record for the electrophoresis run can be made by
drying the agarose film at 37 overnight or at 60 for 2 hr. The stained film
serves as a record itself, or it may be photographed. To prevent scratching
or dislodging of the electrophoresis film with time, an adherent clear
plastic sheet may be placed over the reactive side of the film.
Comment. The procedure presented above utilizes inexpensive
equipment, offers simplicity of operation, and gives rapid results. The
entire procedure will take 2-3 hr, and eight different samples can be
accommodated. The resolution is not as great as that achieved by starch
gel or acrylamide substrates, nor is its application to other enzyme sys-
170
BASIC METHODS
[13]
tems as facile when direct visual staining is utilized. If other electrophoretic systems are already in use, it will require very little additional work to
develop appropriate cell monitoring regimens.
O'Brien et al. is have shown that appropriate selection of additional
isoenzymes polymorphic in a species can be used to develop a genetic
signature for each cell line. Seven isozymes were used to study 30 human
cell lines. They showed that the enzyme profiles obtained distinguished
between 19 human glucose 6-phosphate dehydrogenase Type B cell lines
and 5 dehydrogenase Type A cell lines, demonstrated that two dehydrogenase Type B cell lines thought to be different were actually the same
line, anti demonstrated that three cell lines suspected of being HeLa cells
by other criteria were confirmed as such since all three had a genetic
signature identical to that of HeLa. The probability of two different cell
lines having the same signature with the seven isozymes by chance alone
was calculated to be less than 5%. Extension of the concept seems possible with regard to other species as a means for intraspecies identification
of individual cell lines.
II. Monitoring of Cells by Chromosomal Examination
Examination of chromosomes is the definitive method for determining
cell species, and if carried out in a thorough way it is one of the definitive
methods for individual cell line identification. The methodology for this is
a fairly complicated and time-consuming task--so much so that cytogenetics tends to be completely avoided by those not in that area of study. In
fact, however, it is not difficult to quickly obtain basic information on cell
cultures by chromosomal examination. Metaphase preparations can be
prepared, stained, and examined in a short time. In terms of cell culture
monitoring, these procedures are extremely useful for determining the
current status of a cell culture. Familiarity with the chromosomal complement of the cells cultured in a laboratory will enable one to quickly
denote similarities or differences in a cell line with passage and time in
culture. On the basis of such findings, a decision to obtain more expert
analysis can be made (see also [27]).
A. Preparation of Cell Metaphases
Reagents
[13]
171
172
BASIC METHODS
[13]
Reagents
Giemsa stain 2:
Giemsa powder, 7g
Glycerol, 482 ml
Methanol (absolute), 462 ml
Mix Giemsa powder and glycerol. Place solution in a 60 oven for 2 hr.
Allow solution to cool and add methanol. Filter through Whatman #1
paper as used.
Giemsa buffer, pH 7.0:
Na2HPO4 (anhydrous), 9.5 g/1
NaH2PO4H20, 9.2 g/1
Add 61.1 ml of dibasic phosphate solution and 38.9 ml of monobasic
phosphate solution to 900 ml of distilled water. The working solution of
z0 Harleco Company, Philadelphia, Pennsylvania. Also available as prepared working solution from other companies.
[13]
173
174
BASIC METHODS
[13]
[13]
175
176
BASIC METHODS
[13]
during this period. Centrifuge the suspension at 3000 rpm for 30 min and
remove the supernatant liquid.
2. Add 3 ml of distilled water to the pellet of gamma globulins to
dissolve them and transfer the solution to dialysis tubing. Tightly knot the
dialysis tubing and dialyze against two changes of 1 liter of distilled water
for 30 min each. Dialyze further against six changes of 0.9% NaC1 during
an 18-hr period.
3. After removing the protein solution from dialysis tubing, sample 0.1
ml to determine the protein concentration; the Biuret method a4 is satisfactory. On the basis of the protein assay, weigh sufficient fluorescein
isothiocyanate (FITC) to have 0.025 mg FITC/mg protein. Mix this with
nine parts by weight of Celite 501 powder, a5
4. The dialyzate is adjusted to pH 9.0 by the addition of 0.5 M
carbonate-bicarbonate buffer (1 volume to 9 volumes dialyzate), and the
FITC-Celite 501 mixture is added slowly with gentle agitation of the
suspension. Continue to gently shake the suspension for 10 min during
which time the FITC becomes bound to gamma globulin. Centrifuge the
suspension at 1500 rpm for 5 min and remove the supernate containing
FITC-coupled globulin.
5. Because there is free FITC in the protein solution the preparation is
charged onto a Sephadex G-25 (fine) column (15 1 cm). Collect the
effluent from the column until it appears colorless. Add an additional 10
ml of saline to the column to remove remaining globulin. Store the
coupled globulin in 0.5-ml portions in ampules at - 2 0 .
C. Titering and Use of Coupled Antiserum
Reagents
Phosphate buffered saline (PBS) pH 7.5:
NaH2PO,H20, 1.38 g/1
NaCI, 9.0 g/1
Adjust to pH 7.5 with 1 N NaOH.
Procedure
1. Grow approximately 3 x 10n cells of the same cell line or a line of
the same species as that against which antiserum was prepared. Disperse
the cells by trypsin-EDTA, if an adherent cell line, and collect the
monodispersed cells. Neutralize the dispersing agent activity by adding 5
ml of growth medium containing serum and centrifuge the cell suspension
s4 j. R. M a r r a c k a n d H. H o c h , J. Clin. Pathol. 2, 161 (1949).
s5 J o h n s Manville Co., Waterville, Ohio.
[13]
177
at 1500 rpm for 5 min. Wash the pelleted cells 3 times with 10 volumes of
PBS. Adjust the concentration of cells to 3 x 106 cells/ml.
2. Thaw an ampule of antiserum and prepare a series of 2-fold dilutions, from undiluted to 1:128 inclusive of antiserum in PBS. Place 0.1 ml
of each dilution of antiserum in a 12 75 mm tube and add 0.1 ml of cell
suspension to each tube.
3. Place the eight tubes on a low-speed rotary shaker to mix the cellantiserum suspension thoroughly for 30 min at room temperature.
4. Add 1.0 ml of PBS to each tube, shake, and centrifuge at 3000 rpm
for 2 min. Remove the supernatant and wash the cell-antiserum mixture
again in the same manner. Remove the supernatant and resuspend the
cells in 2 drops of PBS.
5. Place a large drop of the resuspended cells on a glass slide and
carefully float a 22 x 30 mm #1 coverslip on the drop so as to avoid
trapping air bubbles or crushing the cells in suspension.
6. Place the slide on the stage of a fluorescent microscope and observe
the preparation with a x 16 or 25 objective by dark-field illumination.
When cells are located, switch to fluorescent illumination. Cells reacting
specifically have a halo of peripheral bright green fluorescence delimiting
the cell membrane. Nonreactive cells are difficult to discern because of a
failure to show reaction. Dead cells have bright green fluorescence across
the entire cell. The brightness of a specific reaction will quickly fade (30
sec) if fluorescent illumination continues, so a new microscopic field
should be located by changing back to dark-field illumination and repeating the process.
7. Examine each slide in the dilution series as described to qualitatively determine the dilution at which brightness is less intense. The working dilution of antiserum is of that dilution.
8. Test the working dilution of the antiserum on other species of cells
maintained in the laboratory and compare its reaction with that against
the homologous species of cells. If nonspecific cross-reactions do occur,
absorption with 1 volume of the cells of the cross-reacting species and 9
volumes of the working dilution of antiserum will usually eliminate the
cross-reaction without significantly diminishing the homologous reaction.
Alternatively, use of a higher dilution of antiserum will frequently take
care of the problem.
9. For routine monitoring cells should be tested within 4 hr of harvesting. The procedure works only with living cells. It is not well adapted to
fixed cell preparations or tissue sections. Excess working dilution antiserum can be refrozen and used one additional time. After that, a fresh
working dilution antiserum should be prepared for use. The undiluted
178
BASIC METHODS
[14]
coupled antiserum is stable in frozen storage for several years. After that
period of time the antiserum may require titering.
Comment. T h e procedure described is not difficult to perform. Results
can be obtained in 2-3 hr, and the test can be activated for use as needed.
The only reagent that need be prepared for each day of test is PBS.
Antisera are stable for a long period of time, and they can be used for
other serologic tests of cell identity, including cytotoxicity, hemagglutination, and mixed agglutination. 3e-3a Emphasis is placed on a direct immunofluorescent procedure because, unlike the other methods, it can be
used to follow cocultivation experiments involving cells of two different
species, as well as detecting early evidence of cross-contamination. As
few as one cell of one species can be detected in a mixture of 10,000 cells
of another species. 39 The indirect immunofluorescent procedure can also
be used.
The limitations of the method derive mostly from the limitations of
specificity of antisera that can be prepared. Cross-reactions between
closely related species occur. For example, chimpanzee, orangutan, and
human cells react to substantially equal degree with human antiserum.
Rhesus monkey, African green monkey, and baboon cells react similarly
with rhesus antiserum, so one should carefully consider the monitoring
capability of this procedure in relation to the species of cells being utilized
in the laboratory. However, the method can be adapted for use in intraspecies cell characterization. For example, using reciprocally absorbed
antisera, specific human T cell and B cell antisera have been developed to
analyze T and B lymphoblast cell lines by indirect immunofluorescence. 4
3e A. E. Greene, L. L. Coriell, and J. Charney, J. Natl. Cancer Inst. 32, 779 (1964).
3T K. G. Brand and J. T. Syverton, J. Natl. Cancer Inst. 28, 147 (1962).
3e D. Franks, B. W. Gurner, R. R. A. Coombs, and R. Stevenson,Exp. CellRes. 608(1963).
39 W. F. Simpson and C. S. Stulberg, Nature (London) 189, 616 (1963).
4o j. Kaplan and W. D. Peterson, Jr., Clin. Immunol. lmmunopathol. 8, 530 (1977).
[14]
T i s s u e C u l t u r e o n Artificial C a p i l l a r i e s
By P. M. GULLINO and R. A. KNAZEK
[14]
179
cedures, the CCU preserves the pericellular microenvironment and simulates the physiological conditions in which cells function in vh,o.
Principles
Under standard tissue culture procedures, cells are immersed in a pool
of medium containing essential nutrients and metabolic products. The
concentration of both constituents changes as the cell population grows,
thus influencing cell survival and function. In the CCU the culture medium flows within the capillaries and its composition can be kept constant.
The cell population grows within the extracapillary space, and nutrients
and metabolites are exchanged by diffusion from the perfusate within the
capillaries. A more complex form of CCU embodies a double network of
capillaries whereby differential pressure between the two capillary sets
superimposes extracapillary convection upon diffusion.
In neoplastic tissues, anoxic necrosis was observed in vh, o when the
distrance between the capillary wall and the cells was about 150/zm or
more.1 The CCU was built on the assumption that within a rapidly growing population sufficient oxygen tension could not be maintained at distances greater than 150/xm from the capillary wall. If a bundle of artificial
capillaries of about 300 /~m in outside diameter are tightly packed in
parallel, most of the extracapillary space should be within the oxygen
diffusion distance of 150/xm.
Construction of Culture Units
The standard culture unit consists of polymeric membranes in the
shape of tubes bundled together within a transparent cylinder (Fig. 1A,B).
We refer to these tube-shaped membranes as "capillaries" although their
diameter and wall thickness are much greater than an in vivo capillary.
Several types of artificial capillaries made of polysulfone, acrylic
copolymers, or cellulose acetate are available commercially, as is the
complete CCU. 2 Such units, individualized to suit specific needs, can be
constructed with a variety of available capillaries, as follows:
A bundle of 100-300 capillaries, having inner and outer diameters of
- 2 0 0 and - 3 5 0 ~m, respectively, are arranged in parallel and pulled
tightly into a glass or plastic shell about 1 cm in diameter and about l0 cm
in length. The bundle is closed with a suture tie at both ends of the shell
and immersed in a mixture of 12 g liquid silicone rubber (General Electric
1 R. H. Tomlinson and L. H. Gray, Br. J. Cancer 9, 539 (1955).
2 Amicon Corporation, Lexington, Massachusetts; Gulf South Research, New Orleans,
Louisiana.
180
BASIC METHODS
[14]
FIG. I. (A) Capillary culture perfusion circuit: (a) tissue culture medium reservoir; (b)
peristaltic pump; (c) silastic tubing coil for oxygenation of perfusate; (d) capillary culture
unit; (e) syringes for loading ceils onto capillaries. (B) Schematic presentation of capillary
culture unit: (a) loading ports; (b) cutaway view shows capillaries encased in polymeric
potting material (stippled area). (C) Histologic cross-section of human mammary carcinoma
cells (BT20) cultured for 4 weeks within a culture unit. Note the extracapillary space filled
with cells that also penetrate the porous outer structure of the capillary walls. Inner diameter
of capillary is 200/xm.
RTV-11) a n d 8 g M e d i c a l F l u i d 360 ( D o w C o m i n g C o r p . , M i d l a n d , M i c h i gan) t h a t h a d b e e n f o r m e d b y c a t a l y s i s w i t h a p p r o x i m a t e l y 40 m g stann o u s a c e t a t e ( T e n n e c o , N u o c u r e 28). A n e p o x y r e s i n c a n b e u s e d in p l a c e
of the silicone rubber mixture. After solidifying overnight, the excess
[14]
181
polymer is trimmed flush to the shell to expose the lumen of the capillaries. The process is then repeated for the opposite end. The resultant
seal must be tight so that fluid pumped through one end of the CCU will
flow only within the capillaries while the.extracapillary space, in which
the cells are growing, remains undisturbed. With this type of arrangement
the exchange between inner and extracapillary spaces occurs mostly by
diffusion through the capillary walls.
In the more complex CCU, convective currents are generated within
the extracapillary space. The capillaries are intertwined in a braid-like
fashion rather than arranged in a parallel array. This unit consists of a
central part where the capillaries form a single bundle and two extremities
where the capillaries diverge into two bundles, one designated as the
"artero-venous" (A-V) circuit and the other as the "lymphatic" circuit.
The A-V circuit is filled with medium circulating in a closed recirculating
loop to which pressures up to 300 mm Hg can be applied. The lymphatic
system is filled with medium circulating at the same rate but at atmospheric pressure. Steady convective currents, therefore, move within the
extracapillary space from the artero-venous to the lymphatic networks.
The CCU with lymphatic drainage is not available commercially.
Perfusion Circuit
a. Assembly
182
BASIC METHODS
[14]
tion, but the disadvantage of wearing out the wall of the pumping chambers. Replacement of this chamber every 10 days is advisable. Another
problem is the liberation of microscopic fragments from the inner wall of
the pumping chamber into the perfusing medium. After several weeks of
uninterrupted activity, occlusion of the capillaries may occur with irreparable damage to the culture. 4
A CCU with lymphatic drainage requires two circuits, assembled as
described above, with an additional device to increase the pressure of the
medium circulating within the A-V circuit. This may be attained by
pressuring the A-V reservoir with an air-CO2 mixture. The lymphatic
circuit is connected to a second reservoir, and the medium is circulated at
normal atmospheric pressure. An equal flow rate is usually maintained in
both lymphatic and A-V circuits.
It is convenient, although not indispensable, to insert a filter in the
perfusion circuit downstream from the pump with an optional bypass. In
this manner, sampling sites can be inserted into the circuit.
The recommended sterilization of the CCU is in ethylene oxice for 10
hr followed by extensive aeration; however, certain types of CCUs may
be autoclaved, or the unit may be perfused in a 3% formaldehyde solution
at 37 overnight. 4 The remainder of the perfusion circuit may be autoclaved. All components are then assembled in an aseptic environment. The
assembled circuit must be perfused with sterile, pyrogen-fi-ee water for at
least 2 and preferably 7 days with at least three changes of water prior to
replacing the perfusate with complete medium. The objective of the preliminary flushing is to eliminate residues that are toxic to the cells.
The sterilized circuit should always be handled under rigid aseptic
conditions; one should replace the reservoir in a sterile hood while wearing sterile gloves, and the stopper of the reservoir should be covered by
aluminum foil to be replaced with each change of the medium.
Seeding of the CCU is done with approximately 5 106 cells obtained
either by dissociating tissues or from an established culture. The cell
suspension is injected through one shell port while a slight vacuum is
drawn by a syringe on the second port (Fig. 1A). The fluid must be
transferred slowly to deposit, rather than channel, cells or cell clumps
between the capillaries.
A perfusion flow rate of 0.5-1.0 ml/min is maintained at the beginning
of the culture and then gradually increased as the culture grows. When the
cells fill more than 25% of the extracapiUary space, a perfusion volume of
10 ml/min must be maintained to supply adequate amounts of oxygen and
nutrients. Under these conditions the recirculating medium becomes depleted very rapidly and frequent changes of the reservoir are required.
4 C. F. W. Wolf and B. E. Munkelt, Trans. Am. Soc. Artif. Intern. Organs 21, 16 (1975).
[14]
183
Any culture medium used for a specific cell type, grown in standard in
vitro cultures, can be used for CCU perfusion. In this system the capacity
to supply oxygen to the culture is severely limited by the low solubility of
oxygen in the medium. To improve delivery, hyperbaric conditions may
be employed. Fluorocarbons (3M Corp., St. Paul, Minnesota) or erythrocyte suspensions may also be used as oxygen carders, although they are
not necessary under standard conditions.
Cell growth can be detected by gross observation of the CCU. During
the second week after seeding, a white haze appears and gradually
obscures the capillaries. The amount of tissue present within a CCU can
be estimated by measuring the total DNA content, but this requires destruction of the culture. A less precise estimation can be made by measuring the glucose and oxygen consumption of the growing cell population in
the CCU. In repetitive cultures of the same cell population, the correlation between both metabolic parameters and DNA content of the CCU is
reasonably reliable.
Histologic sections through the capillary bundle can be obtained by
flushing the perfusion circuit for 1-2 rain with warm buffered saline and
then circulating a fixative solution, such as 10% formaldehyde, through
the capillaries for several hours. The extracapillary medium is then replaced with a 2% agarose solution at 37 by slowly injecting the solution
through one shell port while withdrawing it from the other until agarose
has penetrated the whole bundle. After solidification the shell is removed
and the capillary bundle can be processed for histologic examination (Fig.
1C).
Utilization of the Culture Units
The major theoretical reason for developing the CCU was to reproduce tissue-like structures in vitro. In the CCU a high cell density
(100 l0 e cells/era3) is obtained, much higher than under standard in vitro
culture conditions; the pericellular microenvironment is preserved; the
exchange of metabolites occurs through a capillary-like network; and the
same culture can be studied for several weeks. The importance of these
parameters in regard to the biological properties of cell populations has
already been evaluated under a variety of conditions. Prolactin and
growth hormone production were studied in rat and human cells of pituitary origin. 5 Secretory stimulation of human/3 cells was studied, 6,7 and
5 R. A. Knazek and J. S. Skyler, Proc. Int. Syrup. Growth Horm. Rela. Pept., 3rd, 1975 p.
386 0976).
6 W. L. Chick, A. A. Like, and V. Lauds, Science 187, 847 0975).
7 R. A. Knazek, in "Pancreatic Beta Cell Culture" (E. yon Wziselewski and W. L. Chick,
eds.), p. 29. Excerpta Med. Found., Amsterdam, 1977.
184
BASIC METHODS
[15]
[15] M i c r o c a r r i e r C u l t u r e : A H o m o g e n e o u s E n v i r o n m e n t
Studies of Cellular Biochemistry
for
Microcarrier culture is the growth or maintenance of anchoragedependent cells on small beads suspended in a stirred tank. The primary
conception of this technology was made by A. L. van Wezel of the
Netherlands. 1 He and his colleagues were responsible for several important demonstrations of feasibility. ~-4 Other laboratories have confirmed
their observations. 5~ Van Wezel and others noted a marked toxicity of the
original microcarriers which was ascribed variously to binding of essential
nutrients or unidentified toxic contaminants associated with manufacture.
However, recent studies have revealed a simple parameter, surface
1 A. L. van Wezel, Nature (London) 216, 64 (1967).
A. L. van Wezel, Prog. Immunobiol. Stand. 5, 187 (1972).
3 A. L. van Wezel, in "Tissue Culture: Methods and Applications" (P. F. Kruse, Jr. and
M. K. Patterson, eds.), p. 372. Academic Press, New York, 1973.
4 D. van Hemert, D. G. Kilburn, and A. L. van Wezel, Biotechnol. Bioeng. 11,875 (1969).
s C. Horng and W. McLimans, Biotechnol. Bioeng. 17, 713 (1975).
6 R. E. Spier and J. P. Whiteside, Biotechnol. Bioeng. 18, 659 (1976).
[15]
MICROCARRIER CULTURE
185
charge density, which, when optimized, permits the facile attachment and
growth of animal cells on microcarriers without any of the toxic effects
previously noted. 7-9
The advantages of microcarrier culture are based on provision of a
large surface area in relation to the volume of the growth vessel used. This
may be understood by comparing roller bottles to microcarriers in terms
of the surface area afforded. A standard 500 cm z roller bottle requires 100
ml of nutrient medium. One milliliter provides 5.0 cm 2 of growth surface.
In our microcarfier cultures, 1 ml of medium provides a minimum of 30
cm 2 of growth surface, and the opportunity exists for increasing this number by 2 or 3 times. Thus, for most cell types, a 5-liter spinner flask would
replace a facility using 300 roller bottles. While the simple increase in
surface area is important, other advantages are offered as a result of the
suspended, stirred configuration of microcarrier culture. Possibly the
most important advantage is the provision of a homogeneous environment
for the cell population, in which the time-dependent changes in microenvironment which occur in culture plates and minimally agitated roller bottles are reduced by simple mixing of the cellular microenvironment and
the bulk liquid of the culture. We suspect that students of cell physiology
are performing many studies without knowledge of microenvironmental
changes or their effects on cellular behavior. Microcarrier culture permits
the investigator to increase the rate of exchange between the cell surface
and the bulk liquid. Thus, the properties of the cellular macro- and microenvironments can approach identity. The bulk properties (pH, redox
potential, concentrations of excreta and nutrients, etc.) become controllable by the experimenter with the facility afforded in bacterial, yeast, or
suspension-adapted animal cell culture. When needs arise for amounts of
cellular material in the range of 10-100 g, scale-up can be accomplished
easily using very simple equipment.
Source of Microcarriers
Microcarriers are obtained from Flow Laboratories, Inc., Rockville,
Maryland. A usable small-scale procedure for their synthesis is based on
that of Hartman TM in which diethylaminoethylchloride. HC1 is reacted
7 D. W. Levine, D. I. C. Wang, and W. G. Thilly, in "Cell Culture and Its Applications"
(R. T. Acton and J. D. Lynn, eds.), pp. 191-216. Academic Press, New York, 1977.
8 D. W. Levine, J. S. Wong, D. I. C. Wang, and W. G. Thilly, Somatic Cell Genet. 3, No. 2,
149-155 (1977).
9 D. W. Levine, D. I. C. Wang, and W. G. Thilly, Biotechnol. Bioeng. (submitted for
publication).
10 M. Hartmann, U.S. Patent 1,777,970 (1930).
186
aASIC METHODS
[15]
[15]
MICROCARRIER CULTURE
187
rinsed with 10-4N HC1 (4 x 50 ml) to remove the unbound chloride. The
beads are rinsed with aqueous sodium sulfate (10% w/w), and the effluent
is collected. This procedure replaces bound chloride ions with sulfates,
releasing the chlorides into the effluent solution. The chloride is titrated
with 1 M silver nitrate in the presence of dilute potassium chromate as an
indicator. The carders are washed with 30-ml portions of 10% sodium
sulfate solution until the amount of released chloride in the effluent is less
than 0.1 mM. Typically, three to four washes are required. After titration,
the carders are again washed thoroughly with water (500 ml) and then
with phosphate-buffered saline (PBS). The carders are suspended in 100
ml of PBS and autoclaved.
Initiation of the Culture
The microcarrier culture bottle is simply a glass or plastic vessel containing a suspended stirrer in which the provision is made to avoid grinding surface contacts with the liquid culture. Simple bottles which are
particularly convenient for microcarrier work are available from Wilbur
Scientific, Boston, Massachusetts, in the volume range of 50-1000 ml. As
mentioned, microcarriers are sterilized by autoclaving a suspension of
physiological saline buffered at pH 7.
The culture is begun by the combination of microcarriers, e.g., 5 mg
dry weight beads per milliliter culture medium, with the desired volume of
culture medium and the introduction of the cells to be used (freshly trypsinized or otherwise collected) as a suspension into the culture bottle. The
bottle is gassed to the desired COs concentration if necessary and placed
on a magnetic stirrer motor (50-90 rpm) and allowed to incubate at 37.
This process is indeed as simple as it must seem to the reader acquainted
with cell culture practice.
Observation of the Culture
The use of the microcarriers at 5 mg dry weight dextran per milliliter
will provide about 40,000 beads/ml. Seeding at 300,000 cells/ml assures
distribution of cells to all carriers. Within a few minutes, most cells will be
attached to microcarriers as illustrated for secondary chick fibroblasts in
Fig. 1A. Samples of stirred cultures are simply withdrawn by pipetting
using nonwettable pipettes to avoid adherence of beads to hydrophilic
surfaces. Plastic pipette tips (Rainin Instrument Company, Brighton,
Massachusetts) are the simplest solution to bead adherence. Samples are
observed directly under a microscope. In a few hours, cells will have
attained their normal culture morphology as illustrated in Fig. lB. Growth
188
BASIC METHODS
[15]
is easily observed microscopically as seen in Fig. 1C. Confluence is attained as on other culture surfaces (Fig. 1D).
Cell counting is easily performed using a hemocytometer and the
technique for enumeration of nuclei reported by Sanford et al. 11 A 1-ml
u K. K. Sanford, W. R. Earle, V. S. Evans, J. K. Waltz, and J. E. Shannon, J. Natl. Cancer
Inst. 11, 773 (1951).
[15]
MICROCARRIER CULTURE
189
40
i~
FIBROBLASTS
50
'o
x
.j 20
_$ i,
T I M E (DAYS)
FIG. 2. Growth curve of secondary chick embryo fibroblasts. Initial seeding at 300,000
cells/ml with 5 g/fiter microcarriers at 2 meq/g dextran. After an initial lag phase (typically
shorter than observed in this experiment) exponential growth to about 2 10 cells/ml is
followed by linear growth to saturation between 3-4 x l0 e cells/ml. Use of 10 g/liter microcarriers with timed partial medium changes will bring chick embryo fibroblast concentrations to 1.0-1.2 l0 T cells/ml.
190
BASIC METHODS
[15]
A
8
FIG. 3. Appearance of epithelial cell lines on microcarriers. Cultures at 5 g/liter microcarriers, 2.0 meq/gdextran. Both cu.lturesare in exponentialgrowth phase. (A) Chinese
hamster ovary cells; (B) HeLa cells.
state. CHO, HeLa, murine cl 1, and human embryonic lung fibroblasts
(HEL299) have demonstrated vigorou s growth without any refeeding
after seeding. However, some cell types such as African green monkey
kidney (AGMK) or human foreskin fibroblasts (FS-4) have not demonstrated equally vigorous growth (Fig. 4).
To date, no dependence on medium type or formulation has been
observed. At this time, it seems that media suitable for growth of cells in
roller bottles or petri dishes are equally suitable when used with the
microcarriers described in this article.
Harvesting
The mode of harvesting will vary depending on individual research
needs. The simple washing of the microcarriers with buffered saline, trypsin exposure, and separation of cells from beads by differential sedimentation rate or by sieving is easy and effective. Increased shear can be
achieved in a number of ways such as increased impeller velocity, pumping through a small orifice, 6 or other strategems.
Some workers have asked about removing cells without trypsin for
biochemical studies in which loss of external cellular components is undesirable. We have been unable to devise a means of preventing such losses
i n harvest, but note that considerable cellular material remains on microcarriers and standard glass or plastic dishes when either trypsinization
or strictly mechanical means are used to remove cells from the attachment substrate. An alternate approach to this problem might be the em-
[15]
MICROCARRIER CULTURE
i
40
OHILo
/~cl I
3C
echo
i.i11!. 2
191
I-1FS- 4
g
~" 20
N
J
0
TIME (DAYS)
FIG. 4. Growth curves for various cell types. All were seeded at 2-3 x 105 ceUs/mlin
100-mlcultures, 5 gm/litermicrocarriers at 2.0 meq/gdextran. HeLa--human epithelial-like,
aneuploid; CHO--hamster epithelial-like,pseudodiploid; cl 1--murine epithelial-like,aneuploid; FS-4--human fibroblast, diploid; AGMK--African green monkey, epithelial-like,
diploid.
ployment of a synthetic growth surface of which a known component
would be severed by enzymic treatment, leaving the cell surface or its
associated exudate unaltered.
Viral Studies
Viruses of interest to investigators may remain physically within a
cell, be budded continuously from the cell surface, or be expressed into
the liquid support medium by cell lysis. In the case of a cell-contained
virus, e.g., Marek's disease, the cells simply can be stripped from the
beads as in cell harvest and the cell suspension used in biochemical studies or as the basis of a vaccine.
Some budding viruses, e.g., RNA tumor viruses, present a special
challenge to virologists in that loss of infectious units with time is often a
problem necessitating frequent or continuous harvesting. Microcarrier
culture offers a ready solution to these problems. The motor drive of a
microcarrier culture, and a pump connecting the culture to a refrigerated
collection bottle, can be connected to a timer. The culture is grown to an
appropriate cell density, infection is accomplished, and when viral expression commences, the stirring motor is turned off, the microcarriers
192
BASIC METHODS
[15]
allowed to settle for a few minutes, the supernatant pumped into the
collection bottle, fresh medium pumped into the culture, and stirring
begun again. This cycle is repeated at intervals appropriate to the cells
and viruses involved.
Viruses expressed into the culture medium lytically are handled similarly. Stirring ceases, and the supernatant liquid above the settled beads is
pumped into a receiving container.
One consideration in the use of microcarrier culture is that the viral
growth cycles may not be the same in these microcarrier suspension
cultures as they are on roller bottles or plates, possibly because of differences in microenvironmental conditions. Thus, preliminary experiments
optimizing for time of infection, latent period, and harvest are necessary.
Viruses on microcarriers that have been studied in this manner include
Sindbis, polio, Moloney murine leukemia, and vesicular stomatitis. 12
Rabies virus has been produced in excellent yield, ~a and a number of
commercially important veterinary vaccine viruses are apparently produced in good yield.
Interferon and Other Cellular Products
Theoretically, interferon or other secretory products could be produced in a manner directly analogous to the budding viruses on a semicontinuous basis. Initial interferon studies ~4 have found that using the same
conditions of cell type, growth media, and superinduction, the interferon
production on a per-cell basis was the same for cells in microcarrier
culture or in roller bottles.
There seem to be no obvious reasons to expect that other cellular
products such as peptide hormones or secretory enzymes could not be
made or studied to advantage through the use of microcarrier systems. It
should be noted, however, that the microcarriers described in this article
are porous, positively charged hydrogels. Some products having a low
molecular weight or a negative charge may be expected to associate with
the microcarriers and possibly require special elution procedures.
Mitotic Cell Selection
One limitation on study of the mammalian cell division cycle has been
the difficulty of obtaining a sufficient number of synchronously dividing
1~D. J. Giard, W. G. Thilly, D. I. C. Wang, and D. W. Levine,Appl. Environ. Microbiol. 34,
668-672 (1977).
18 B. Mered, personal communication.
14 D. J. Giard, D. H. Loeb, W. G. Thilly, D. I. C. Wang, and D. W. Levine, Biotechnol.
Bioeng. (submitted for publication).
[15]
MICROCARRIER CULTURE
193
194
BASIC METHODS
[16]
[16] M a s s C u l t u r e o f M a m m a l i a n C e l l s
By WILLIAM F. MCLIMANS
Introduction
The purpose of this article is to provide a guide to the mass culture of
mammalian cells for those investigators who require milligram or gram
quantities of cells for biochemical studies. Thus, some points may appear
controversial or even offensive to the "cell culturist" anxious to protect
his domain. The "state of the art" and need are such, however, that an
effort must be attempted, even at that risk. The rewards are great for
those who would exercise control over their own source of cells.
The approach is complicated by the multiple configurations of equipment and methods that are available. Basically, however, there are only
two effective mass-scale systems; one employs the suspension culture of
established cell lines, and the other and newer technique, is that of direct
METHODS IN ENZYMOLOGY, VOL. LVIII
[16]
MASS C U L T U R E OF M A M M A L I A N CELLS
195
Operations
Water
196
BASIC METHODS
[16]
TABLE I
SOURCES: MATERIALS AND EQUIPMENT
Letter ref.
//
Source
Travenol Laboratories, Inc., 1 Baxter Parkway, Deerfield, Illinois 60015
Difco Laboratories, Detroit, Michigan
Flow Laboratories, Inc., 1710 Chapman Avenue, Rockville, Maryland
20852
Grand Island Biological Company (GIBCO), 3175 Staley Road, Grand
Island, New York 14072
Microbiological Associates, 5221 River Road, Bethesda, Maryland 20016
Colorado Serum Company, 4950 York Street, Denver, Colorado 80216
Fiske Associates, Inc., Bethel, Connecticut
Cryogenic Supply Co., Inc., Quinby Park, Hamburg, New York 14075
The Matheson Co., Inc., East Rutherford, New Jersey
Armour Pharmaceuticals, Inc., P.O. Box 511, Kankakee, Illinois 60901
Bellco Glass, Inc., 340 Edrudo Rd., Vineland, New Jersey 08360
Ace Glassware, Inc., Vineland, New Jersey / 54 Muddy St., (P.O. Box
425), Ludlow, Massachusetts 01056
Virtis Co., Route 208, Gardiner, New York 12525
New Brunswick Scientific Co., Inc., 1130 Somerset St., New Brunswick,
New Jersey 08903
Fermentation Design, Inc., Div. of New Brunswick Scientific Co.,
Bethlehem, Pennsylvania 18017
Chemicals
Biologic grade or U S P chemical reagents should be used in the formulation o f all tissue culture preparations. Certification of the source and
purity o f the reagents should be furnished b y the vendor. A certificate of
analysis o f amino acids and vitamin mixtures should be obtained prior to
p u r c h a s e o f bulk lots.
T h e use o f c o m m e r c i a l l y p r e p a r e d liquid culture m e d i a is not recomm e n d e d for the following critical reasons. One has no control o v e r the
quality o f the w a t e r c o m p o n e n t . T h e formulation structure is too rigid to
permit critical adjustment o f p H , osmolarity, and glucose concentration at
various levels. S o m e m e d i a c o m p o n e n t s , such as penicillin and glutamine,
are labile and thus undergo continuous changes in concentration as a
[16]
197
198
BASIC METHODS
[16]
TABLE II
STOCK SALT SOLUTIONS
Stock
number
Stock
reagent
Final (x l)
(mg/liter)
1
2
3
4
5
6
NaCI
CaCI~ (anhyd.)
KC1
MgSO4 7 H~O
NaH2PO4H20
Phenol red
6460
I00
400
97.7
580
2
Glucose
Gentamycin
Mycostatin
10
NaHCO3
11
NaCI (mos M)
Stock (x 10)
(g/liter)
64.6
10.0
40.0
9.77
58.0
20.0 mg
Stock
(ml/liter)
90a
I0~
10
10
10
2
Glucose
1~4 g/liter
Stock of 100 mg/ml or 100 g/liter in water.
Antibiotic
50 mg/ml stock: use 0.5 ml/liter = 0.025 mg/ml =
25 tzg/ml.
500,000 ~g/vial + 10 ml H20 = 50,000 Izg/ml stock:
use 2.0 ml stock/liter for final cone. of 100 mg/ml.
Buffer adjust stock
5 g/100 ml or 5%
Osmolarity adjust stock
1 mg/ml
a Stock number 1: NaCI is added at lower level than that normally used so as to permit
osmolarity adjustment later.
b In some suspension-type cultures the deletion of Mg~+ and Ca2+ ions is recommended
in order to reduce the tendency of some cell types to clump.
Media Preparation
W e p r e p a r e c u l t u r e m e d i a a s f o l l o w s . T h e s t o c k salt s o l u t i o n s , a s
d e t a i l e d in T a b l e I I , a r e p r e p a r e d in a v o l u m e o f 1 l i t e r w i t h d i s t i l l e d w a t e r
as t h e d i l u e n t . T h e s t o c k s o l u t i o n s a r e e i t h e r f r e s h l y p r e p a r e d a n d u s e d
immediately, or stored as sterile solutions. Solutions are sterilized by
filtration in o r d e r t o a v o i d p o s s i b l e c o n t a m i n a t i o n o f t h e s t o c k s o l u t i o n s
during autoclaving with steam containing volatile boiler preservatives, s
S t e r i l i z a t i o n b y a u t o c l a v i n g in u n i t s fitted f o r s t e a m , a s g e n e r a t e d f r o m
d i s t i l l e d w a t e r , is a c c e p t a b l e . C a r e m u s t b e e x e r c i s e d d u r i n g t h e filtration
t o w a s h t h e filter b y d i s c a r d i n g t h e first p o r t i o n o f fluid p a s s i n g t h r o u g h t h e
filter; t h i s p r e c a u t i o n will r e m o v e f i l t e r - b o n d i n g m a t e r i a l , d e t e r g e n t , o r
w a t e r t h a t m a y h a v e b e e n r e t a i n e d in t h e filter.a
a G. E. Gifford, J. Bacteriol. 80, No. 2, 278-279 (1960).
R. D. Cahn, Science 155, 195-197 (1967).
[16]
199
Media pH
A problem in the preparation of tissue culture media is the uncontrolled variability of the final pH under the actual experimental conditions. This, particularly in the instance of bicarbonate buffer systems,
results from: variation in the bicarbonate levels, as found in various serum
components; the intrinsic buffer capacity of the media mixture; variation
in the actual concentration of the CO2 overlay (see section on culture gas);
and the adjustment of pH at room temperature during media preparation
whereas the cells and medium are actually maintained at 36.5 . Thus, if
medium is prepared at 25 with the pH adjusted to 7.30, the actual pH on
incubation at 36.5 will be 7.40. For CO~oicarbonate systems in this range
a differential of 0.087 pH units per degree C increase is observed.
The critical importance of pH shifts to cellular function has been described in detail. 1 For several years, we have used a very simple "tonometer" for pH adjustment that consistently has yielded culture media
poised at any preset or desired pH level (Fig. 1). The tonometer consists
of a 100-ml water-jacketed flask, via which the contained medium is held
at the desired culture temperature--36.5 . A Teflon-coated spin bar is
used to facilitate equilibration. The upper configuration of the tonometer
possesses two ports for the direct flow and circulation of the selected
CO2-air mixture over the stirred flask contents. Normally, the same cylinder of gas is used for the tonometer as for the actual cultures. A third
indwelling port is provided as a thermometer well. Finally, a silicone
stopper is added to both seal the chamber and allow insertion of a combination pH electrode the tip of which is centered in the middle of the
medium. A vaccine stoppered port penetrates the stopper and permits
direct addition via a tuberculin syringe (2-5 ml) of 5% NaHCOa without
loss of equilibration gas.
The bicarbonate is added step by step, being sure that the equilibration
level has been reached at each step before the next addition. Extrapolation from the 50-100 ml tonometer volume to the actual batch volume
yields the correct amount of 5% NaHCO3 required for any desired pH, at
any incubation temperature, with any gas mixture, despite variations in
the buffer capacity of the medium. Its prime disadvantage is that it re10S.
200
BASIC METHODS
[16]
PH
Probe
Il
Thermometer Well
~ _ ~ ~ " ~
Bar"
Osmolarity Adjustment
After adjustment of the pH, medium osmolarity is measured with an
appropriate osmometer. Our instrument (g) has a precision of _-_1.0
mosM/kg water. The balance of NaC1 required to achieve the desired
osmolarity is calculated in the following manner.
1. Desired or normal osmolarity of healthy persons = 291.8 -+ 2.6
mosM/kg water. 11
2. Measure the osmolarity of experimental media and calculate the
amount of a special stock NaC1 (1 mg/ml) which must be added to achieve
the desired osmolarity. Thus: 1 mg NaC1/ml = 1 ml stock (mosM) =
32 mosM increase. Therefore:
D-O
32
=X
[16]
MASS C U L T U R E OF M A M M A L I A N CELLS
201
While the major goal remains the simplification of the tissue culture
"tool," meaningful studies demand that the mammalian cells be cultured
under physiologic conditions that are nontoxic and reproducible. In these
terms, one of the most important parameters of that environment is the
culture gas. Thus, for example, culture buffers operative without bicarbonate utterly fail to provide sufficient CO2 and, therefore, must'depend on
202
BASIC METHODS
[16]
TABLE III
GAS CONTAMINANTS IN COMMERCIAL BOTTLED GAS
Cylinders,
research grade
(ppm)
O~
Carbon monoxide
Nitrous oxide
Hydrocarbons (as methane)
C02
<50
<l
16
Estimated level
in gas overlay of
cell culture (ppm)
5% CO2 < 2.5 ppm
20% O~ < 0.2 ppm
20% O~ < 3.2 ppm
the cells to provide this gas for the environment. TM Indeed, the buffer
capacity of the CO~/bicarbonate system is perhaps the least important of
its many metabolic control configurations. 12Accordingly, in our laboratories we routinely use the CO~oicarbonate buffer system.
Specialty gas mixtures, medical or biologic grade, are available for use
as culture gas. However, wide variations may exist in the actual pC02
levels of commercial gas mixtures. Such variation can be extensive if
certified gas mixtures are not obtained. In our laboratories, infrared analyses of the actual CO2 content of 27 tanks of uncertified 5% COs in air
showed a mean variation of 0.9% from the tank value noted by the
supplier. The actual range of variation was from 0.1-3.3% with an overall
span of 3.2%. Very low values, 2-3%, may be encountered if the tanks
have not been properly mixed by rolling.
Gaseous contaminants may include particulate matter, microbial
agents, and oil. These are easily removed by in-line filtration. Other contaminants such as hydrocarbons, nitrous oxide, and carbon monoxide are
of concern as toxic or potentially mutagenic agents (Table III).
It is recommended that specialty gas mixtures be procured with clear
specifications as to the permissible level of contamination and variation of
component concentration, i.e., less then 3 ppm moisture, less than 2 ppm
CO, etc. Certification by lot to lot analyses is much less expensive than
tank to tank and just as effective.
Suspension Culture--Cell Lines
A number of investigators have demonstrated that established mammalian cell lines proliferate in agitated fluid suspensions. Initial tech~z W. F. McLimans, in "Growth, Nutrition and Metabolism of Cells in Culture" (G. H.
Rothblat and V. J. Cristofalo, eds.). Vol. 1, Chapter 5, pp. 137-162. Academic Press, New
York, 1972.
[16]
203
204
BASIC METHODS
[16]
[16]
205
206
BASIC METHODS
[16]
TABLE IV
A COMPARISON OF COSTS (LEss LABOR) OF BATCH VS. CONTINUOUS PROCESS OF
PRODUCTION OF MAMMALIAN CELLS AS REFLECTED BY CELL POPULATION,
FERMENTOR VOLUME, AND SERUM CONCENTRATION
IN CULTURE MEDIUM a
Batch b
Fermentor volume (liters)
Maximum cell Population (x 106/ml)
Day of first harvest
Number of harvest days
Total cells harvested (x 109)
Grams cell yield/week (g)
Total media used (liters)
Total serum (10%) used (liters)
Cost of serum-free media
(at $1.46/liter)
Cost of serum
Total media costc
Cost/gram of cells ($)
Continuous a
1
1.4
2-3
11
3.85
2.24
6.5
0.65
$8.54
10
1.4
2-3
I1
38.5
22.5
65.0
6.5
$85.41
10
10
5-6
8
800
467
120
12
$157.68
5.20
13.74
6.13
52.00
137.41
6.11
96.00
253.68
0.54
a All calculations are based on 24-hour generation time for 12 days following establishment of the culture on day 0.
Batch system calculations based on harvest of culture every day.
c Media cost based on 66d/liter for Baxter Water, 78.5d/liter for dry mix, 10d/liter for
salts and antibiotics, and $8.00/liter for serum at concentration of 10%, i.e., price
levels obtaining January 1978.
a Continuous systems are calculated to reflect conditions at the same cell population
level as batch and under conditions wherein cell population was maintained at about
x7 this level. Such levels are reafistic with some cell lines. We have operated units
at 8-10 x 10s cells/ml, while Earle has reported short-term propagation at levels as
high as 20 108 cells/ml.
establishing conventional monolayer subcultures and by periodically det e r m i n i n g t h e efficiency o f p l a t i n g w i t h a t t e n t i o n t o h i s t o l o g i c t y p e s o f
colonies.
In "batch-type" systems, the culture pH should be checked daily and
efforts m a d e t o m a i n t a i n it a b o v e 6.9. T h i s is a c h i e v e d b e s t b y t h e d a l l y
a d d i t i o n o f f r e s h m e d i a in a n a m o u n t g e n e r a l l y e q u a l t o 1 0 - 2 0 % o f t h e
t o t a l v o l u m e . E x c e p t f o r v e r y h i g h c e l l p o p u l a t i o n s , t h i s p r o c e d u r e will
help to maintain growth and circumvent possible exhaustion of a substrate
such as glucose.
Suspension Culture--Primary Cells
A critique of suspension culture systems must include observations
r e l a t i n g t o t h e m o r p h o l o g i c a n d b i o c h e m i c a l s i m i l a r i t y o f e s t a b l i s h e d cell
[16]
207
208
BASIC METHODS
[16]
sion system. Details regarding the type of cell growth, kinetics of cell
proliferation, inhibitory effect of high bead concentrations, and growthenhancing activity by a cell factor have been presented, a2 The reported
experimental work appears to have established the necessary background
for a direct approach to the long-term, steady-state culture of primary
differentiated cells at high population densities in suspension culture.
The calf anterior pituitary gland, immediately after removal, was isolated, decapsulated, and soaked in a trypsin mediummMcCoy's 5A containing 6.250 IU/ml of Tryptar (j'). After transport to the laboratory, the
tissue was minced and trypsinized in a stirred 50-ml spinner flask containing 15 ml o f " t r y p t a r " medium. After 6 hr of trypsinization, the cells were
readily dissociated with a Pasteur pipette. Typically, from one pituitary
gland, 2-7 107 cells were obtained with a viability above 95%. In other
instances, e.g., mammary and prostate, cultures were initiated better directly from a fine tissue mince mixed with the prepared " b e a d s " ; trypsin
appeared to be deleterious to cells derived from these organs.
McCoy's 5A medium supplemented with 10% heat-inactivated fetal
calf serum was used as growth medium for the pituitary cell cultures. The
media was adjusted to a pH of 7.4 with a gas overlay of 2% COs in air for
normal tissue and 5% COs for tumor tissue. The osmolarity was controlled
at 291-315 _+ 10 mosM.
DEAE-Sephadex was, in the main, used as a microcarrier, although
other beads are available, a3 Prior to use, the beads are sequentially
washed with 0.5 N NaOH, distilled water, 0.5 N HC1, distilled water, and
a phosphate buffer solution, and sterilized by autoclaving at 121 for 20
min. They are stored at 4 . Just prior to use, the phosphate buffer is
replaced with culture medium.
Suspension cultures are carried out, for example, in a spinner flask of
50-100 ml capacity (k,l). The cultures are incubated at 36.5 with a magnetic spin bar adjusted to a speed of about 100 rpm. The spinner flasks are
continuously gassed via an overlay of 2% COs in air. The usual culture
volume is 30 ml. The pH of the culture fluid is monitored daily. If the pH
decreases below 7.0, the culture is usually fed with 10-20 ml of fresh
growth medium. Every 3-4 days, or daily, depending on the activity of the
culture, I ml of the cell/bead suspension is removed and mixed with 3 ml
of phosphate buffer solution contained in a petri dish. The percentage of
beads with cells growing on their surface is checked by microscopic observation. Subsequently, the beads with attached cells are washed with a
phosphate buffer solution and exposed to 4 ml of 0.1 M citric acid while
being incubated at 37 for 1 hr. Following the incubation, the released
aa D. W. Levine, J. S. Wong, D. I. C. Wang, and W. G. Thilly, Somatic Cell Genet. 3, No. 2,
149-155 (1977).
[16]
209
nuclei are washed from the bead/cell surface with the citric acid solution,
stained with gentian violet, and counted in a hemocytometer or by an
electronic counter; method for counting nuclei follows the procedure originally developed by Sanford et al. 34
Interestingly, "naked beads" added to a monolayer culture will pick
up and anchor cells. Conversely, "beads" with anchored cells when transferred from suspension to a flat culture surface will initiate monolayertype clones and monolayers.
For monitoring the morphology and cell types, petri dish cultures are
established from a dilution of the microcarrier suspension cultures. Feeding is done by replacing the acidic spent medium with 3 ml of fresh growth
medium at 2--4 day intervals. Quantitation of colonies, as well as histologic
evaluation, is made after 14 days of incubation in a COz chamber or bell
jar incubator. This constitutes a critical control procedure!
The DEAE-Sephadex bead is transparent in growth medium. Thus,
the morphology of the attached cells can be fixed by the usual methods
and monitored directly via the light or electron microscope.
As noted, petri dish cultures are employed for preliminary assessment
of cell growth under various conditions. Immediately after inoculation,
cells are observed to attach to the bead surface and form a round mass for
about 2 days. Thereafter, the cells appear to spread out and begin to
proliferate. By the 4th day, a partial monolayer of cell growth on the bead
surface can be observed. After 2 weeks of incubation, nearly all of the
beads are completely covered by cell layers. It appears that cells may
move from one bead to an adjacent bead, leading eventually to the formation of bead clumps with cell layers serving as a bridge; cells are predominantly of epithelial morphology when attached to the bead surface. Electron microscopy of cells from these cultures shows the presence of active
mitochondria, endoplasmic reticulum, and tight cell junctions. Some
cytoplasmic projections have been noted between bead and cell, which
probably indicates an active anchorage site, rather than passive physicalchemical attachment of the cells on the bead surface.
Suspension cultures in spinner flasks have been routinely successful.
Within certain limits of bead concentration, cells proliferated in the suspension system with growth on the order of magnitude of a 10-fold cell
population increase in 8 days. Contrary to the expectation that higher
bead concentration should provide greater surface area for cell growth
and a higher incidence of cell-bead collision and attachment, it appears
that with a constant cell inoculum, higher bead concentrations may inhibit
cell growth. This inhibitory effect, as would be expected, can be reduced
84 K. K. Sanford, W. R. Earle, V. J. Evans, H. K. Waltz, and J. E. Shannon, J. Natl. Cancer
Inst. 11, 773-795 (1950-1951).
210
by using proportionately higher quantities of inoculum. There might be a
critical cell-to-bead ratio below which the cells will not show sustained net
growth. By using a rough estimation of 1.6 105 beads/ml in the bead
bed, this ratio was calculated to be about 1 : 5 for the calf anterior pituitary
cells.
In our hands, direct primary cell culture with the microcarrier suspension system and batch feeding can routinely yield a cell population of
about 5 10e cells/ml. However, with such a dense population, nutrient
feeding becomes the limiting factor. Even feeding 3 times a day, one
cannot keep up with the extreme shifts in pH. To circumvent this problem, we ~ave devised a continuous-feed culture system with automatic
pH monitoring and feedback loop, as based on use of a modified "SpinFilter" (rn). Other systems may be equally applicable (n,o).
Employing the "Spin-Filter," a 500-ml microcarrier (DEAE) culture
of primary anterior pituitary cells was continuously maintained, at a high
cell population (4-7 10e cells/ml), for 57 days with excellent control of
the culture parameters. Repetitive subcultures to petri dishes were routinely successful.
Cell debris does not cause significant problems in healthy cultures.
However, freely suspended cells do appear. Whether or not these represent newly divided cells as released from bead surfaces remains to be
established. At times, freely suspended cells may reach a population as
high as 3 x 105/ml and have a viability above 85%. If such a cell population is truly being continuously released from the beads, it might well be
possible to consider these methods for synchronized cell culture studies.
Other systems and configurations for cell culture have been demonstrated, i.e., capillary tubes, 3s plastic or cellular sheets or tubes, 3e multiplate fermentors, 37 and even columns of Sephadex. a8 They are not considered he~-e, since they all appear to suffer from difficulty in quantitating
growth rates and responses. There is also a marked tendency for formation of clumps or cellular masses. Such clumps may, in turn, serve as foci
of necrosis and transformation. Vital to any system is the facility of meaningful monitoring techniques with which to assess the true kinetic or
histogenic profile of the culture. This, in turn, must depend on obtaining
truly representative aliquots.
It is the author's opinion that the microcarrier technique will, in time,
prove to be the best tool for this task.
s5 R. A. Knazek, E M. Gullino, P. O. Kohler, and R. I. Dedrick, Science 178, 65-67 (1972).
See also this volume [15].
se M. D. Jensen, D. F. H. Wallach, and P. Sherwood, J. Theor. Biol. 56, 443-458 (1976).
s7 R. Weiss and J. B. Schleicher, Biotechnol. Bioeng. 10, 601-616 (1968).
aa B. Bergrahm, Biotechnol. Bioeng. 10, 247-251 (1968).
[17]
PROPAGATION
AND
SCALING-UP
OF
CULTURES
211
Acknowledgment
I acknowledge the invaluable and tireless assistance of Chi-byi Horng, Barbara Kwasniewski, Angeline Wasielewski, Frances O. Robinson, Robert Zeigel, Elizabeth Repasky,
W. "Sam" McLimans, Peter Hasenpusch, and William Beers in the investigative phases that
form the bases of this article. I thank Ann M. Gannon for the editing and other assistance.
The United States Public Health Service supported with these grants-in-aid various facets of
this work: 5 R01-ES00030-05, 89013-01, NIH-NCI-G-72-3866, 1 R0"IGM22532-01, and 5 P,26
CA 1865203.
[17] Propagation
and Scaling-Up
of Suspension
Cultures
I. Introduction
One m a j o r consideration in cell culture is w h e t h e r a given cell line
grows in suspension or m u s t attach to a matrix. Initial attempts at growing
cells in vitro dealt mainly with anchorage-dependent types. Following the
early demonstrations 1~ that cells would multiply in suspension cultures,
m a n y investigators recognized the potential of this a p p r o a c h and sought
to increase the efficiency o f the process (see Perlman et al. 3-6 for review).
F r o m the beginning animal cell suspension culture w a s appealing due to
the anticipation o f utilizing fermentation techniques similar to those used
in cultivating microorganisms. Although the a p p r o a c h is similar in principle the difference in growth p a r a m e t e r s b e t w e e n p r o k a r y o t i c and e u k a r y o tic cells p o s e s unique problems.
An a p p r o a c h will be described here for the growth of cells in suspension culture f r o m 1 ml up to 200 liters, e Although the method was initially
derived for the propagation o f l y m p h o c y t e s , 7 it has been found amenable
to m a s t o c y t o m a , h e p a t o m a , L cells, and various n e r v o u s tissue derived
cell lines. Since cell culture requires a high level of attention to detail,
lo. v. H. Owen, M. K. Gey, and G. O. Gey, Ann. N.Y. Acad. Sci. 58, 1039(1954).
2W. R. Earle, R. L. Schilling, J. C. Bryant, and V. J. Evans, J. Natl. Cancerlnst. 14, 1159
(1954).
3 D. Perlman, Proc. Biochern. 2, 42 (1967).
4 G. E. Moore, Methods Cancer Res. 5, 423 (1970).
5 R. C. Telling and P. J. Radlett, Adv. Appl. Microbiol. 13, 91 (1970).
6R. T. Acton, P. A. Barstad, R. M. Cox, R. K. Zwerncr, K. S. Wise, and J. D. Lynn, in
"Cell Culture and Its Application" (R. Acton and J. Lynn, eds.), p. 129. Academic Press,
New York, 1977.
7 R. K. Zwerner, C. Runyan, R. M. Cox, J. D. Lynn, and R. T. Acton,Biotechnol. Bioeng.
17, 629 (1975).
212
BASIC METHODS
[17]
even the more mundane aspects of the approach are described. Experience has taught that inability to grow a particular cell line frequently lies
in the failure of an individual to adhere precisely to the prescribed steps
rather than in the method itself.
II. Cell Culture
1. Recovery of Cells
The initial procedure for culturing continuous lines depends on the
state in which cells are received. Cells may be transported either as viable
cultures or in the frozen state. If cells are received as a growing culture
one simply determines density and viability and places them in fresh
medium, ff received in the frozen state cells }nay be either stored in liquid
nitrogen or placed directly into culture. The ampule containing the frozen
cells is thawed by immediately immersing in a 45 water bath. The ampule
should be agitated to insure that the thawing process is completed in less
than 1 rain. The ampule is immersed in 70% ethanol to reduce the possibility of contamination and transferred to a vertical flow laminar hood. The
neck of the ampule is scored with a small sterile file and the cells transferred to a sterile 15-ml polystyrene screw-cap disposable centrifuge tube
(Coming #25310) using a sterile Pasteur pipette. Ten volumes of culture
medium at 4 are added and the tube is capped and inverted several times.
The cells are pelleted by centrifugation at 400 g for 5 rain. It is important
to conduct these procedures immediately after thawing the cells in order
to remove the cryoprotective agent as quickly as possible. Regardless of
the designated concentration of cells contained in the ampule the cell
density and viability should be determined at this time. The cells are then
apportioned to sterile 25 cm 2 or 75 cm 2 polystyrene tissue culture flasks
(Coming #25100 or 25110, respectively), and the cell concentration is
adjusted to approximately 1 10n cells/ml with medium at 37. An aliquot
is evaluated for microbial contamination. The flasks with slightly loose
caps are then placed in a humidified CO2 incubator at 37. Each day
thereafter the cells are enumerated and evaluated for viability, and the
cell concentration is adjusted by adding new medium. When the culture
volume allows, an aliquot of cells should be frozen and stored in liquid
nitrogen to serve as a source of future stocks and reference point. In the
event the cells grow slowly special attention must be given to include
centrifugation every other day and resuspension in fresh medium, assessing the use of other media or nutrient supplementation and variation
in cell density. 7
[17]
213
214
BASIC METHODS
[17]
vessel must be sufficient to sustain growth until the cells are evaluated
again. As one progresses to larger volumes the amount of oxygen available to the cells becomes a limiting factor. For volumes between 50 ml and
6 liters the Bellco-type spinner flasks (Bellco Glass, Inc.) offer the largest
volume above the culture and provide the best system for suspension
culture. For volumes above 6 liters the configuration of vessels from most
manufacturers are such that a gas mixture must be added continuously to
obtain optimal growth. For some cell lines a fermentation-type vessel in
which pH and dissolved oxygen are continuously monitored and controlled is more effective. These vessels are available in sizes from 1 liter to
14 liters with various optional instrumentation (New Brunswick Scientific
Co., Inc., Edison, New Jersey).
C. DEFINING GROWTH PARAMETERS
[17]
PROPAGATION
AND
SCALING-UP
OF
CULTURES
215
iJ
8
6
E
i
c
2
o
8
\ \
10 5
I
0
I
24
I
48
I
72
I
96
I
120
1.
216
BASIC METHODS
[17]
I0i
8
6
x_.x~.x..x ~
o
.)
~- 106
o
i'
~
"'x ~"x"x.
)zo~,
7.5
"x''x''x''x'~
6.s
0
(.)
~4
~8
7,z
9,e
Culture Timo (Hours)
FIG. 2. Growth of $49.1 in a 4-liter spinner flask (Wheaton Scientific). The medium was
RPMI-1640 supplemented with 10% HS. The culture was overlayed with a mixture of 5%
COz + 95% air and capped immediately.
A set of conditions has been established which proves useful for the
growth of cell lines in suspension culture. 6,1," When culturing mammalian cells according to this scheme, cells are maintained in concentrations
between late exponential and early stationary phases of growth by the
addition of fresh media.
In practice one may establish a growth curve and continuous culture
conditions by the following method:
1. Begin cultures at several initial densities starting at some concentration, e.g., 10~ cells/ml, using rapidly dividing cells. The optimal inoculation point is the lowest initial concentration that results in a very short or
no lag phase.
~op. A. Barstad, S. L. Henley, R. M. Cox, J. D. Lynn, and R. T. Acton, Proc. Soc. Exp.
Biol. Med. 155, 296 (1977).
, R . T. Acton and J. D. Lynn, Adv. Biochem. Eng. 7, 85 (1977).
[17]
217
i#
8
~i
'
,
'
4OOml
8C'Oml
260_mI 6.~0~I
6000 ~
i
1
1 1200Oral
15Oral 200m
FIG. 3. Scaling-up the growth of $49.1 from a tissue culture flask to growth in a 12-fiter
culture vessel. Growth from the 150-ml stage to the 2600-ml stage was conducted in Bellco
spinner flasks. Growth from 6.5-liter to 12-liter was conducted in a 14-liter New Brunswick
fermentor (New Brunswick Scientific Co.).
218
BASIC METHODS
[17]
Upon removal from the magnetic stirrer and before the cells settle, 0.5
ml of the culture is aseptically removed by use of a sterile 1-ml pipette and
placed in a 12 x 75 mm RTU culture tube (BD #7813 Becton Dickenson
and Co. Rutherford, New Jersey). A 100-/.d aliquot is mixed with 100/zl of
a solution of 0.4% trypan blue in normal saline (#525 Grand Island Biological Co.; Grand Island, New York). This high concentration of trypan blue
is necessary to counteract the affinity of the dye for proteins in solution. TM
An alternative dye is erythrosine B which has a greater alfnity for nonviable cells than the soluble proteins in the culture fluid. ~3 Cells are then
counted in a standard 0.1-mm deep hemocytometer bright-lipe counting
chamber (American Optical Corp. Buffalo, New York). By incorporating
one of the two vital stains into the diluting fluid as described the viability
of cells can be obtained simultaneously during cell enumeration. In the
hemocytometer viable cells will appear as bright circles whereas dead
cells appear as dark circles, often with irregular edges.
B . PHENOTYPIC ANALYSIS
Several cell surface components have been shown to vary in expression during various stages of the cell cycle."-ls Although it is impractical
to maintain cells in suspension culture in synchronous growth, this condition is approachable by close attention to the culture procedures previously described. If the main interest, for example, is cell surface components, it is important to evaluate the particular property of interest during
growth in order to determine the optimal time to harvest cells for maximal
yield. For example, Thy-1 alloantigen expression on murine lymphoblastoid cells is lowest during the lag and stationary phases of growth, T M and
maximal expression occurs when cells are maintained in the exponential
phase of growth. One method to quantitate the amount of cell surface
component on a cell is the absorption of antiserum as detected by the
~aH. J. Phillips, in "Tissue Culture Methods and Application" (P. Krase, Jr. and M. Patterson, Jr., eds.), p. 406. Academic Press, New York, 1973.
~4R. K. Zwerner and R. T. Acton, J. Exp. Med. 142, 378 (1975).
~s M. Cikes, J. Natl. Cancer Inst. 45, 979 (1970).
~6M. Cikes and G. Klein, J. Natl. Cancer Inst. 49, 1599 (1972).
~R. A. Lerner, M. B. S. OIdstone, and N. R. Cooper, Proc. Natl. Acad. Sci. U.S.A. 68,
2584 (1971).
~aM. A. Pellegrino, S. Ferrone, P. G. Natali, A. Pellegrino, and R. A. Reisfeld, J. Immunol.
108, 573 (1972).
[17]
219
cytotoxicity assay. 19--21By comparing the ability of cells to absorb a standard antiserum during various stages of the growth curve, one can determine the exact stage for harvest that yields the highest concentration of a
particular cell surface component. 7.22
As previously indicated, there are a number of factors that affect the
rate of cell growth. In the initial phase of optimizing cell growth one
should determine which culture medium produces the maximum number
of cells per unit volume in the shortest possible time. However, it is also
important to determine if the culture medium chosen also yields the highest quantity of the cell ~omponent under study. The type of serum supplement used in culturing cells can influence the expression of routine
lymphoblastoid cell surface components. For example, the L251A cell
line had a 15-fold greater expression of the routine thymus leukemia antigen (TL) when grown in medium supplemented with 10% fetal calf serum
instead of 10% horse serum.~4 Although it is currently difficult to ascertain
at this point what factors in serum affect the expression of cell surface
components, it is important to evaluate the culture medium selected with
regard to the particular phenotypic property of interest.
4. Quality Control
A . MICROBIAL C O N T A M I N A T I O N
220
BASIC METHODS
[17]
After the optimal growth parameters for a given cell line have been
established, aliquots of the culture should be frozen and cryopreserved.
Aliquots of cells are taken while in exponential growth, centrifuged at
400g for 10 min, and resuspended at 4 in either fetal calf serum or horse
serum containing 5% dimethylsulfoxide (Fisher Scientific Co.) to a concentration of 1-5 10* cells/ml in sterile, 2-ml, prescored cryule ampules
(Wheaton 200 Brand #651486, Wheaton Scientific). The tip of the ampule
is sealed in a hot flame by use of the pull-seal method. The ampules are
frozen at a controlled rate of l/min in a BF-4-1 freezing chamber regulated by a Linde BF-6 controller (Union Carbide Corp., Linde Division).
If a controlled-rate freezing device is not available the ampules can be
placed in a small Styrofoam box lined with cotton. The lid is taped into
place, and the carton is placed in a - 7 0 freezer overnight. After freezing,
the ampules are stored in liquid nitrogen. The next day, an ampule or two
of the frozen cells are placed in culture to evaluate the freezing process by
determining viability, growth properties, and possible microbial contamination. Periodically throughout the maintenance of a given stock of cells,
z5T. C. Hsu, in "Tissue Culture Methods and Application" (P. Krase, Jr. and M. Patterson,
Jr., eds.), p. 764. Academic Press, New York, 1973.
[18]
HARVESTING
PRODUCTS
OF
CELL
GROWTH
221
5. Summary
The general approach discussed here is one that should serve as a
guide for the culture of almost any type of cell in suspension culture.
Following the initiation of cells in culture, it is advised that optimal conditions for growth be determined. In order to maintain the integrity of the
cells and to protect the time invested in their growth it is also mandatory
that one effect adequate quality-control measures to assure that cells are
maintained in a healthy state, free from contamination, and that aliquots
are stored for future reference and use. By adherence to these relatively
simple rules, one will find that cells in culture can be a most valuable tool
in molecular biology.
Acknowledgments
This work was supported by U.S. Public Health Service Grants CA 15338, CA 18609,
and CA 13148 from the NCI, IM33C from the American Cancer Society, GB-43575X from
the Human Cell Biology section of the NSF and the Diabetes Trust Fund. The work of
R. T. A. was done during the tenure of an Established Investigatorship of the American
Heart Association. P. A. B. was supported in part by National Service Award F32-AI 05423
from the NIAID. R. K. Z. was supported in part by National Research Service Award
T32-GM 07561 from the NIGMS.
[18]
H a r v e s t i n g t h e P r o d u c t s o f Cell G r o w t h
222
BASIC METHODS
[18]
CELL SUSPENSION
8~ @ 2.5 x 10~cells/ml
Centrifuge @ 650 1- 20 rain
BeCkman J6, JS-4.2 rotor
CELL SLURRY
CELL SUPERNATANT
SUPERNATANT
CELL DEBRIS
CELLS
Re~uspendto 1 x 10= cells/ml
in Tris-NaCI
SUPERNATANT
Disrupt
I
I
p113-1.18 fractions
pooted and diluted
with STE buffer
POOLED-VIRUS
CONTAINING FRACTIONS
Centrifuge @ 100.(~0 g-1 hr
I Beckman L5-50. SW 27 rotor
PELLET
SUPERNATANT
SUPERNATANT
Cm~trifuge@ 4809-20 mm
Soi~'alIRC-2, SS-34 rotor
NUCLEI AND
NONDISRUPTED
CELLS
I Resuspend Jn Tr~s-NaCI
Dounce homogenize
Centrifuge @ 480 g-20 rain
I
SUPERNATANT
NUCLEAR
POOL
PELLET
i
I
Cenlrifuge @ 4000B-20 mtn
SorvallRC-2, SS-34 rotor
SUPERNATANT
SUPERNATANT
POOL
MITOCHONDRIAL
Resuspend in TrlS-NaCI
Dounce homogenize
Centrifuge (~ 4000 g-20 rain
MITOCHONDRIAL
PELLET
Centrifuge @ 20.0001-30 mm
IISorva#RC-2,
SS-34 rotor
I
I
SUPERNATANT
I
MEMBRANE
PELLET
MuLV
PELLET
[18]
IOC
8C
223
I00
8O
6c
t~
.o
~ 4c
40
2o
2O
,~o
2~o
36o
,~o
may damage the cells due to shearing effects. The cells are resuspended in
one-half the original volume of Tris-NaCl buffer (10 mM Tris chloride and
0.15 M NaCI at pH 7.4) and centrifuged as before to remove residual
culture medium. Following this procedure 95% or more of the cells in the
starting suspension culture can be harvested with a viability greater than
95% as determined by trypan blue exclusion. The cells are resuspended in
Tris-NaCl buffer to a concentration of 1 x 108 cells/ml.
To obtain subcellular organelles, cells must be disrupted in a manner
preserving the integrity of these structures and allowing effective separation into pure fractions. We have found the Stansted Cell Disrupter
equipped with a model AO 612 air-driven hydraulic pump and a model 716
disrupting valve (Energy Service Co., Washington, D.C.) an excellent
instrument for this purpose. This instrument provides a means for the
continuous and controllable disruption of a wide variety of cell types.l'3'4
Disruption is caused by the shearing effect of the fluid as it passes under
pressure through an orifice whose dimensions are controlled by air
pressure on a piston. For each cell line a calibration curve must be determined to ascertain a disrupting pressure that results in the highest yield of
the desired product. Figure 2 represents a typical controlled disruption
profile for the murine lymphoblastoid cell line EL4. Disruption is measured by the severity of total cellular disintegration and cell viability as
a B. M. Wright, A. J. Eduardo, and V. E. Jones, J. lmmunol. Methods 4, 281, (1974).
4 R. T. Acton, P. A. Barstad, R. M. Cox, R. K. Zwerner, K. S. Wise, and J. D. Lynn, in
"Cell Culture and Its Application" (R. T. Acton and J. D. Lynn, eds.), p. 129. Academic
Press, New York, 1977.
224
BASIC METHODS
[18]
[18]
225
TABLE 1
DISTRIBUTION OF THY-I.1 ACTIVITY IN FRACTIONSOF DISRUPTED BW5147 CELLS
Fraction
Disrupted cells
400 g Supernatant
400 g Pellet
4000 g Supernatant
4000 g Pellet
20,000 g Supernatant
20,000 g Pellet
Total
protein
(mg)
Total
activity
(Thy-1 units)
Protein
(%)
Purification
Yield
(%)
2296
1019
1085
800
95
796
105
45.6
38.5
16.3
29.0
7.4
6.7
21.9
-44
47
35
4
35
5
-1.8
0.7
1.7
3.7
0.4
9.9
-79
34
60
15
14
45
226
BASE M E T H O D S
[18]
1.24
,~
,oo I
I '~
,.,,
o~
,~-
U
tw
IM
,.14
z ~ -
9< ~
82
WI.12
a
5W
,.,o
t o oa.~ 34 -~
>-
1.08
2U.I
i oC
LOS, ~
0
~
5
~-..9-~,-~-.~-~,.
10
15
20
25
FRACTION NUMBER
[18]
227
The low-speed cell supernatant fluid obtained from the initial centrifugation step (see Fig. 1) is further centrifuged in 250 ml polypropylene
bottles (Fisher Scientific Co., Cat. No. 5579) at 6000 g for 30 min at 4 in a
Sorvall RC-2 centrifuge with a GSA rotor to remove residual cells and
cellular debris.
A step gradient is formed in a Beckmart CF-32 continuous-flow zonal
rotor by sequential addition of STE buffer (200 ml), 15% (w/w) sucrose
(120 ml), 43% sucrose (110 ml), and 50% sucrose until STE buffer is eluted
from the rotor. Taking care not to disturb the sediment, the clarified
supernatant liquid is removed and continuously pumped at 2.5 liters/hr
into the rotor spinning at 30,000 rpm. After the sample has been applied,
STE buffer is pumped through the rotor for about an hour to allow partial
equilibration of the sedimenting virus. The gradient is then eluted from the
rotor by pumping 55% sucrose into the bottom of the gradient, and 10-ml
fractions are collected. Virus is found distributed in the gradient at densities between 1.13-1.18 g/cm ~. The fractions are pooled, diluted, and
centrifuged in aellulose nitrate tubes (Beckman No. 302237) at 100,000 g
in a Beckman SW27 bucket rotor at 4 for 60 min.
If cultures are contaminated with mycoplasma, this pellet may contain
large quantities of these organisms. Virus can be further purified by
isopycnic centrifugation on linear gradients of potassium tartrate. Prior to
this step, thorough dispersion of the 100,000 g pellet in STE buffer is
necessary and may be carded out either by vigorous homogenization in a
Dounce homogenizer fitted with a Teflon pestle or by gentle sonication.
The latter technique should be carefully controlled and possibly avoided if
structural integrity of the virus is essential. We have observed that envelope components of MuLV may be disrupted by excessive sonication.
This suspension is applied to a linear gradient of 5--40% (w/w) potassium
tartrate in STE buffer, and isopycnic centrifugation is conducted in celM. F. Baffle, in "Cell Culture and Its Application" (R. T. Acton and J. D. Lynn, eds.), p.
291. Academic Press, New York, 1977.
228
BASIC METHODS
[18]
75, 4479
[18]
H A R V E S T I N PRODUCTS
G
OF CELL GROWTH
229
III. Comments
The procedures described were designed to allow the effective purification o f cells, subcellular organelles, and components from suspension
culture medium. By varying this general approach one should be able to
harvest products from the growth of various cell types in suspension
culture in volumes of 1-12 liters. When several products are utilized from
a given suspension culture it b e c o m e s a very economical approach and
enhances the possibility of gaining further information on the molecular
biology o f the cell.
Acknowledgments
This work was supported by U.S. Public Health Service Grants CA 15338, CA 18609,
and CA 13148from the NCI, IM33C from the American Cancer Society, GB-43575Xfrom
the Human Cell Biology section of the NSF, and the Diabetes Trust Fund. R. K. Z. and
K. S. W. were supported in part by an institutional National Research Service Award No. T32
GM07561 from the NIGMS. This work by R. T. A. was done during tenure of an Established
Investigatorship of the American Heart Association.
[19]
233
[19] Cell C y c l e A n a l y s i s b y F l o w C y t o m e t r y 1
By
J. W. GRAY
and E
COFFINO
ISBN 0-t2-181958-2
234
[19]
SPECIALIZED TECHNIQUES
TABLE I
COMMERCIALLY AVAILABLE FLOW CYTOMETERS AND FLOW SORTERS
Instrument
type
Company a
Becton-Dickinson FACS
Ortho Instruments
Coulter Electronics, Inc.
Flow
Flow
Flow
Flow
Flow
Address
sorter 4
cytometer
cytomete~ '7
sorter
sorter 6
Sample(A)
[~
Fluorescence
detector
Sheathwater ~ . 3._-f~ ~
Deflection
pJat~
/:~
~.~ ~'~
" :
~
"~::-..__ Microscopeobjective
""~G.
~ "-'--"....
Sorted
"'-,DLaserbeam
(~~illection tubes
[19]
235
All flow cytometers operate on the principle that stained cells in aqueous suspension will flow with the suspending medium. By channeling this
flow properly, the cells can be forced through a region where their optical
properties can be measured. Usually the medium containing the cells is
surrounded by a sheath of similar fluid.
Figure 2 shows a liquid jet emerging from a nozzle into the air; this
occurs in most flow sorters. The black stream was produced by injecting
ink into the cell sorter instead of a cell suspension. In an instrument such
as the one pictured here, cells are measured immediately after the jet is
formed. In other instruments, cells are injected into glass capillary tubes
or water-filled chambers where they are measured. An intense light
source illuminates the cells in the sample stream. If the diameter of the
sample stream is not sufficiently small, some cells may be illuminated less
intensely than others and thus the fluorescence frequency distributions
will be distorted. The sample stream diameter can be decreased by increasing the pressure on the sheath relative to that on the sample. When
the cell concentration is low, the number of cells analyzed per second
should not be increased by increasing the sample stream diameter, because of the danger of improper illumination. Instead, the cell sample
should be concentrated.
3 M. A. Van Dilla and M. L. Mendelsohn, in "Flow Cytometry and Sorting." Wiley, New
York (in press).
4 L. A. Herzenberg, R. G. Sweet, and L. A. Herzenber~, Sci. Am. 234, 108-117 (1976).
5 L. A. Kamentsky, Adv. Biophys. Med. Phys. 14, 83 (1973).
A. Brunsting, in "Flow Cytometry and Sorting," pp. 79-88. Wiley, New York (in press).
7 This flow cytometer operates according to somewhat different principles than those described in this review. For more information, see W. G6hde, in "Fluorescence Techniques
in Cell Biology," pp. 79-88. Springer-Verlag, Berlin and New York, 1973.
236
SPECIALIZED TECHNIQUES
[19]
FIG. 2. A liquid jet ejected into air by a cell sorter. The sorter nozzle was formed by a
drawn glass capillary with a 50-/.Lminside diameter. The black stream was formed by injecting ink in place of the cell suspension; the confinement of the ink to the center of the jet is
clearly visible. In practice, the sample ~tream should be even smaller than the ink stream
shown in the figure.
F l u o r e s c e n c e Excitation a n d Collection
[19]
/,/
>~ 1.0
f'~
;,\
/
/,
i/11
4OO
\1
ChrornomycinA3-
50O
237
6OO
\
700
Wavelength, nrn
FIG. 3. Excitation and emission spectra ~ for the dyes propidium iodide (A) and
chromomycin A3 (B). Dashed lines represent excitation spectra and solid lines emission
spectra.
238
SPECIALIZED TECHNIQUES
50O0
I
Chromomycin A3
[19]
I
Propidium Iodide
"6
j~ 2S00
=E
Z
....... J
,,.....~/
50
100
50
Relative fluorescence
],~ ..........
100
FIG. 4. Fluorescence distributions of $49 mouse I.ymphoma cells stained with (A)
chromomycin A3 and (B) propidium iodide. The data points are solid circles. The solid lines
are the result of least squares best fits to the data; each solid line is the sum of the dotted
component curves.
to avoid changing the laser power. The G1 peak mode should be adjusted
to the same location at the start of each experiment to standardize data.
Most flow cytometers can measure and record more than one parameter for each cell. For example, both the dye fluorescence and the scattered laser light might be recorded to form a two-parameter distribution,
the dye fluorescence proportional to DNA content and the scattered light
proportional to cell size. The exact relationship between scattered light
and cell size is difficult to quantitate; it depends on the light collection,
angle, and aperture, index of refraction of the suspending fluid, etc. Usually, however, the scattered light increases monotonically with cell volume.
Cell Sorting
The selection of cells on the basis of their DNA content can be coupled
to tracer technology. For example, the cell cycle location of a radioactively labeled cohort of cells can be determined by sorting cells according
to their varying DNA contents (and hence position in the cell cycle) and
assaying the fractions for radioactivity by autoradiography or liquid scintillation counting.
Commercially available cell sorters use the electrostatic deflection
principle illustrated in Fig. 1 to fractionate cells. The cells are ejected into
air and their stain content measured immediately. The cells continue
down the liquid jet until the jet breaks into droplets. When a cell to be
[19]
239
sorted reaches this point, a brief voltage pulse is applied to the jet, and the
droplet containing the cell is charged when it separates from the liquid
column. The distance from the measurement point to the droplet breakoff
must be constant during operation of the sorter. This distance determines
the time delay between cell measurement and the application of the charging pulse and is stabilized by vibrating the flow chamber at high frequency, e.g., 30 kHz. The distance can be disturbed by a change in the
pressure producing the liquid jet, by the occurrence of a bubble within the
flow nozzle, or by a piece of debris lodged in the nozzle. Should one of
these events occur, the wrong droplets will be charged. Since such malfunctions are not uncommon, the droplet breakoff point should be
checked frequently.
Methods
Cell Dispersal
All flow cytometric techniques require analysis of individual cells.
Therefore, prior to analysis, the population must be dispersed into a suspension of single cells.
Cells grown in suspension culture usually require no dispersal, but
those grown in monolayer cultures must be dissociated. To accomplish
this, the medium is decanted and the monolayer gently rinsed with 5 ml
cold PBS (solution 1). 1 The PBS is decanted, 2 ml of trypsin (solution 2)
are added, and the cells incubated for about 5 min at 37. The incubation
should be restricted to the minimum time sufficient to free the cells from
the flask. The cells will leave the monolayer in sheets. This process can be
hastened by striking the culture flask sharply with the heel of the hand.
Dispersal of clumped cells will be aided by gently passing the suspension
in and out of a pipette. After dispersal, 8 ml of culture medium containing
serum are added to inhibit trypsin and the mixture is transferred to a
plastic centrifuge tube. The cells from the monolayer culture now should
be a suspension of single cells. Subsequent steps are identical for cells
grown in monolayer or in suspension. The cells are centrifuged (250 g for 5
min in this and subsequent steps), the supernatant liquid aspirated, and
the cells resuspended vigorously in 10 ml cold PBS with a vortex mixer.
Centrifugation and aspiration are repeated and the cell pellet is vigorously
resuspended in the residual fluid and fixed by adding 5 ml of cold 70%
ethanol dropwise during continuous agitation with a Vortex mixer. After
fixation and cell suspension can be stored almost indefinitely at 4 .
~0 All working solutions are described in the Apperidix.
240
SPECIALIZED TECHNIQUES
[19]
Staining
In our experience, the simplest and most reliable DNA stain is the dye
chromomycin A3 (Calbiochem). The staining procedure is adapted from
that of Crissman and Tobey who used the dye mithramycin, n Mithramycin and chromomycin A3 have similar properties, and chromomycin A3
was chosen because of its availability and high purity.
Cells should be fixed in 70% ethanol for at least 30 rain before staining.
A suspension containing approximately 5 106 fixed cells is centrifuged
and the supernatant liquid removed by aspiration. The cells are resuspended by a Vortex mixer in the CA3 staining solution (solution 3). After
30 rain of incubation in the dark at room temperature, the cells are ready
for flow cytometric analysis. This procedure is elegant in its simplicity;
unfortunately CA3 fluorescence cannot be excited by some flow cytometers (see discussion of flow cytometry above).
When CA3 cannot be used, propidium iodide (PI) can be substituted
with a slight modification of the staining procedure. TM Cells must be
treated initially with RNase because the dye binds to all nucleic acids. To
accomplish this, approximately 5 x 106 cells from the fixative are re~
moved by centrifugation and resuspended with a Vortex mixer in
RNase-buffer solution (solution 4) and incubated for 15 rain at 37. The
cells are centrifuged and the supernatant solution aspirated. Then the cells
are resuspended by vigorous agitation by a vortex mixer in the PI staining
solution (solution 5). The stained cells should remain in the dye solution
for 30 min at room temperature before analysis.
[19]
241
242
[19]
SPECIALIZED TECHNIQUES
TABLE II
ESTIMATED FRACTIONS OF CELL CYCLE PHASES IN A CULTURE OF
ASYNCHRONOUS 849 MOUSE LYMPHOMA CELLS
Propidium iodide
Chromomycin
0.20
0.68
0.12
0.27
0.63
0.10
G1
S
GeM
of Chinese hamster ovary (CHO) cells on the growth rate and the distribution of cells in the cycle. Figure 5A presents the growth curves for two
exponentially growing cultures. One culture was maintained as a control;
the second was established by a 4-fold dilution of one-fourth of the control
culture at time zero. The control culture was started 48 hr, i.e., almost
four population-doubting times, prior to dilution to ensure the decay of
any synchrony that might have been induced initially. The growth curves
of the two cultures, based on frequent sampling, indicated that both grew
exponentially for at least 24 hr after the dilution step. At later times the
cultures entered stationary phase with saturation densities greater than
1 106 cells/ml.
Because of the apparent continued exponential growth after dilution,
the noticeable perturbation in the DNA distribution of the diluted culture
(Fig. 5B) was unexpected. The control culture contained a relatively constant proportion of cells in the G1 phase, both before and after the time
,oo1,,,
,_
_../B'
b0L
'
'
'
~8o
1
-40
-20
20
40
60
201
I
,
I
80
-10 0
20
Time after dilution (hours)
I
40
l
60
i
80
FIG. 5. The response of Chinese hamster ovary cells to dilution of the culture. At - 4 4 hr
a spinner flask was established with 1100 ml culture medium and allowed to reach asynchronous, exponential growth (O--O). At time zero, 275 ml of this control culture was
poured into a second spinner flask containing 825 ml of fresh 37 medium (D--D), giving a
1 : 4 dilution. Cell counts (A) and estimates of the fraction in the G1 phase (B) were made
periodically for both diluted and undiluted cultures.
[19]
243
244
SPECIALIZED TECHNIQUES
Experimental
[19]
Simulation
10 Hours
i Pl-'l/
FIG. 6. Light-scatter and D N A - c o n t e n t distributions o f S49 m o u s e l y m p h o m a cells responding to the administration of d b c A M P at zero time. Experimental distributions are
s h o w n on the left and distributions simulated according to the p a r a m e t e r s in Table III are
s h o w n on t h e right.
[19]
245
TABLE III
KINETIC PARAMETERS ASSOCIATED WITH THE RESPONSE OF $49
MOUSE LYMPHOMACELLS TO dbcAMP a
Average
phase duration
(hours)
Coefficient
of variation
2.1
12.1
2.5
0.25
0.25
0.25
G1
S
G= + M
a The intensity of 90 light scatter was assumed to increase linearly with cell volume; the
light scatter from mitotic cells is twice that from G1 cells. The effect of the dbc AMP
in the model was assumed to be an immediate and complete block in early G1 phase.
The rate of DNA synthesis as a function of DNA content was described by a normal
distribution with a coefficient of variation of 0.4 and the mean centered in mid-S phase.
(This choice in the model reflects the data in Fig. 7.)
1500
~o
o
100
2O0
Relative
DNA
content
246
SPECIALIZED TECHNIQUES
[19]
volume [20]). Such studies were limited by the synchrony techniques and
could not be applied in vivo. The use of flow sorting to select prelabeled
cells according to size and/or DNA content eliminates the need for
synchronization.
An example of such tracer techniques is illustrated by the technique
used in the estimation of the rate of DNA synthesis as a function of DNA
content for CHO cells. 1~ Prior to sorting, cells were pulse labeled for 30
min with 0.5/xCi/ml tritiated thymidine ([ZH]TdR) (specific activity - 2 0
Ci/mM). The cells were then fixed, stained with CA3, and processed
through a cell sorter where 104 cells were sorted from each of ten DNAcontent regions into which the S phase was divided. The sorted cells from
each region were collected on glass filters and the radioactivity measured
by liquid scintillation counting. Figure 7 shows the results of these measurements. The rate of [ZH]TdR uptake is maximal in mid-S phase, decreasing to minimal values during early and late S phase.
Application of the methods described here and related techniques provides a rapid and convenient means for answering questions related to cell
cycle kinetics that would be otherwise difficult or impossible to study. The
rapidity of the fixation and staining methods makes it possible to monitor
results while experiments are in progress. The recent development of
fluorescent DNA-staining techniques that do not affect cell viabilityTM contributes further to the utility of these methods. Somewhat balancing these
advantages, however, are the instrumental complexity and the considerable hardware cost of this technology, which may limit its accessibility for
the occasional user.
KHsPO4
H20
Amount
800 mg
200 mg
1150 mg
200 mg
1000 ml
[19]
247
Solution 2--Trypsin
Component
NaEDTA
Trypsin (Difco
Bacto-trypsin
#0153-60)
PBS
Amount
4 ml (stock solution:
1 g NaEDTA/100 ml H20)
1 vial reconstituted
in 10 ml PBS
186 ml
Amount
10 mg
1.5 g
500 ml
Solution 4---RNase
Component
RNase (Sigma;
71 Kunitz
units/rag)
Na2HPO4.7 H20
Na~HPO4
H20
Amount
10 mg
55.6 mg
168.9 mg
10 ml
Solution 5---PI
Component
Propidium iodide
H20
Amount
10 mg
1000 ml
248
SPECIALIZED TECHNIQUES
[20]
Acknowledgments
The authors gratefully acknowledge helpful technical comments from Y. S. George. This
work was performed under the auspices of the Department of Energy, Contract #W-7405ENG-48 with support from USPHS grant 5R0114533, NIH grant GM16496, and NSF grant
PCM 75-06764. P. C. is the recipient of a Research Career Development Award from the
NIH, Institute of General Medical Sciences.
[20] C e l l S y n c h r o n i z a t i o n
[20]
CELL SYNCHRONIZATION
249
250
[20]
SPECIALIZED TECHNIQUES
100
x
X/x
/
35o
_?
"E
X/"
,t
o
ib
~o
Jo
FIG. 1. Cumulative percentage of cells in DNA synthesis, after quiescent K12 cells were
stimulated to proliferate by serum. K12 cells, a ts mutant that arrests in G1 at the nonpermissive temperature, were continuously exposed to [aH]thymidine. The ordinate gives the
cumulative percentage of cells labeled by [aH]thymidine, and the abscissa gives the time
after stimulation, at the permissive (X---X) or at the nonpermissive (O--O) temperatures.
scribed in detail in a monograph by Baserga and Malamud 4 and is presented in this volume [22]. Briefly, to determine the degree of synchrony
the cells can be continuously exposed to [3H]thymidine or, alternatively,
pulse labeled with [3H]thymidine for a brief period of time, 30 rain or so.
In the former case the concentration of [3H]thymidine (6.7 Ci/mM) in the
medium should not exceed 0.1 /~Ci/ml. We use, satisfactorily, a concentration of 0.05 t~Ci/ml. In the latter case a concentration of 0.5/~Ci/ml is
permissible. The cells are fixed at the desired intervals after the synchronization procedure, usually in Carnoy, and then autoradiographed
with Eastman Kodak NTB nuclear emulsion (at the concentrations mentioned above, an exposure time of 7-10 days is usually sufficient), developed, fixed stained with hematoxylin and eosin, and scored at the light
microscope.
A typical result with this procedure is illustrated in Fig. 1, which
shows the cumulative labeling index of K12 cells synchronized by serum
deprivation. Serum-deprived cells (48 hr in 0.5% serum in coverslip cultures) were stimulated by addition of fresh medium plus 10% serum and
immediately exposed to [3H]thymidine, 0.1 /zCi/ml. Duplicate coverslips
were then fixed at the intervals indicated on the abscissa, autoradiographed, and analyzed. As one can see from Fig. 1, the entry of serumstimulated K12 cells into S is not sharply synchronized. At the time of
serum stimulation most of the cells are in G0--G1, and about 20 hr after
stimulation, 80% or more are in S phase. However, they are not synchronized at the same point. In other words, if one wishes to obtain cells
synchronized in the samephase of the cell cycle (G1 orS), then the method
4 R. Baserga and D. Malamud, "Autoradiography. Techniques and Application." Harper
(Hoeber), New York, 1969.
[20]
CELL SYNCHRONIZATION
251
252
SPECIALIZED TECHNIQUES
[20]
[20]
CELL SYNCHRONIZATION
253
10o
50-
FIG. 2. Cumulative percentage ofceUs in DNA synthesis in K12 cells collected by mitotic
detachment and replated at either the permissive (--) or the nonpermissive (X--X) temperatures. Cells were exposed to [SH]thymidine from the time of replating.
tially growing populations are detached from the plate with 0.25% trypsin
in Hanks's solution for 2 min. They are plated in 100-mm dishes (surface
of 78.5 cm2), at a concentration of 1.5 10~ cells per dish. By microscopic observation of the dishes with an inverted microscope, the time at
which mitotic figures are most frequent can be determined. For K12 cells,
this time interval is about 18 hr after plating. Mitotic cells are collected by
shaking the plates gently. The yield is about 5-8% of the total number of
cells, and the percentage of mitotic cells in the collected fraction is 90% or
more. The mitotic cells are then plated in 35-mm dishes, at a density of
2 x 104 cells/cm 2. The degree of synchronization can be established by
continuous labeling with [3H]thymidine at a concentration of 0.1 /zCi/ml,
added at the time of plating, and by counting the number of labeled cells at
various intervals after plating (see above). The results of such an experiment are shown in Fig. 2.
Degree of Synchrony. The degree of synchrony (Fig. 2) is satisfactory
and such cell populations can be used for a study of G1 events and events
in the S phase. The labeling index at the beginning is less than 5%. At the
permissive temperature, 80% of the cells at 34 go from G1 into S in a
period of only 2 hr. It is possible to follow such cell populations and to
determine how this degree of synchrony is maintained by observing the
wave of mitoses that follows the period of DNA synthesis. With separate
cultures it also can be shown that the number of cells doubles in a rather
brief interval, corresponding to the wave of mitoses. A note of caution is
in order: When mitotic cells are plated, the number of cells plated is
calculated from the number of mitotic cells that one actually seeds in
different dishes. Within 15 min after plating most of the mitotic cells have
completed mitosis, so that the number is already doubled. This should be
kept in mind if one wishes to determine the increase in cell number at a
later period.
254
SPECIALIZED TECHNIQUES
[20]
lOft
8
|
[20]
CELL SYNCHRONIZATION
255
the plated dishes for the appearance of mitoses every time that he deals
with a different cell line.
The yield of mitotic cells can be increased in a variety of ways. Increase in the yield is often desirable because, as mentioned above, a dish
will yield only about l0 n cells. This is sufficient for experiments in cell
biology, but biochemical studies demand larger numbers of cells; one can
easily calculate that 100 100-mm dishes would be necessary to obtain 107
mitotic cells. To increase the yield of mitotic cells several techniques are
available. The most popular one is the use of Colcemid. This technique
was first devised by Stubblefield and Klevecz TM who found that Chinese
hamster cells could be treated for 2 hr with Colcemid, 0.06/~g/ml; the
effect was to arrest cells in metaphase in a reversible manner. These investigators also used a brief period of trypsinization, 45 sec at 4, resulting in
an increased yield of about 8-12% of the total population. In general, the
use of Colcemid for brief periods (2-3 hr) has no remarkable effect on the
biochemical events in mitotic cells or in synchronized G1 and S phase
cells. However, we occasionally have observed that cells treated with
Colcemid had different kinetics of entry into S phase from cells that had
not been so treated. Again, a simple rule cannot be given, and the investigator will have to determine for each cell line whether the increased
yield of mitotic cells that can be obtained with Colcemid is not counterbalanced by possible toxic effects to the mitotic cells. Nias and Fox, 17 for
instance, found that Colcemid treatment beyond 2 hr leads to an increased
number of aberrant mitoses; we have similar experience with several cell
lines.
Another way of increasing the yield of mitotic cells is tO use repeated
harvesting, a procedure first introduced by Petersen e t a l . 2 With this
procedure the monolayers were shaken at 10-min intervals, and each harvest was rapidly cooled to 4. Again, previous experiments had shown
that cells stored at 0 for a period of 4 hr are still capable of completing
mitosis in a manner identical to that of control cultures when plated in
warm medium. Repeated shaking at 10- or 30-min intervals can therefore
yield a larger number of mitotic cells. These may be stored at 4 without
loss of viability for a period of up to 8 hr, and the collected mitotic cells
can then be plated. Undoubtedly a combination of Colcemid and repeated
harvesting leads to an increased yield of mitotic cells. Repeated shaking,
however, may decrease the purity of the preparation, i.e., the percentage
of mitotic cells in the mitotically detached population. Provided one moni~a E. Stubblefield and R. Klevecz, Exp. Cell Res. 40, 660 (1965).
20 D. F. Petersen, R. A. Tobey, and E. C. Anderson, Fed. Proc., Fed. Am. Soc. Exp. Biol.
28, 1771 (1969).
256
SPECIALIZED TECHNIQUES
[20]
[20]
CELL SYNCHRONIZATION
257
ble to select cells from different points of the cycle on the basis of volume,
and thereby produce a synchronized population. In practice, pure populations of G 1, S, or G2 cells have not been obtained, although some of the
physical methods used have produced enriched populations. Linear sucrose gradients, Ficoll gradients, and gradients in fetal calf serum in
phosphate-buffered saline have been used. The several methods have
been discussed at length in the review by Nias and Fox 17 who concluded
that with all the physical methods " a high degree (more than 90%) of
synchronization has been demonstrated only in cells harvested from
monolayer cultures by the mitotic selection technique." Two recently
introduced methods, one by Everson et al. 23 and the centrifugal elutriation
method of Mitchell and Tupper, 24 are described in more detail.
C. FICOLL GRADIENT ACCORDING TO EVERSON et al. 23
This has been used to separate human lymphoblastoid cells growing in
culture. Since these cells are of interest because they are the only human
diploid cells that grow indefinitely, the technique for separation on a
FicoU gradient is given below, as described by Everson et al. 23
A 5-20% (wt/w) Ficoll linear continuous gradient is generated in a density gradient generator, resulting in a total volume of 80 cm 3 contained in
cylindrical polycarbonate tubes 3 cm in diameter and 10.5 cm in length.
On top of this gradient 10 cm 3 of 5% (w/w) Ficoll are layered to modify the
initial slope of the gradient. A suspension of cells in Hanks's balanced salt
solution is then carefully layered on top of this 5% Ficoll buffer zone. The
cell load can reach approximately 5 107 cells, suspended in a final concentration of 1 107/cm3.
Centrifugation is carded out at 4 in a PR.2 centrifuge using a
swing-out rotor at 80 g for 20-25 rain. Fractions are collected by placing a
stainless-steel tube through the center of the gradient to the bottom and
sampling via a polyethylene tubing with a polystatic pump.
The Ficoll is prepared to a starting concentration of 40% (w/w) with
water. It is then subsequently adjusted to a final concentration of 20%,
with an equal volume of double-strength Hanks's balanced salt solution.
This 20% Ficoll solution is then diluted 1 : 4 with isosmolar Hanks's balanced salt solution to obtain a 5% (w/w) FicoU solution. The two concentrations are used to generate the necessary gradient.
D e g r e e o f S y n c h r o n y . The degree of synchrony is modest, as can be
seen from the original figures in the paper by Everson et al. 2~ However,
reasonably good populations of G 1 and S can be obtained.
z3L. K. Everson, D. N. Buell, and G. N. Rogentine, Jr., J. Exp. Med. 137, 343 (1973).
Z4 B. F. Mitchell and J. T. Tupper, Exp. Cell Res. 106, 351 (1977).
258
SPECIALIZED TECHNIQUES
[20]
2. Chemical Methods
A. SYNCHRONIZATION BY ISOLEUCINE DEPRIVATION
[20]
CELL SYNCHRONIZATION
259
Procedure. The detailed technique for synchronization by serum restriction and hydroxyurea is as follows. Cells are plated at a density of
5 105 cells per 100-mm dish for 3 days. The medium is removed, the
plates are washed thoroughly, and the cells are exposed to their usual
medium containing 1% (or 0.5%) serum for 48 hr. After 48 hr, the serumdeficient medium is replaced by a medium containing 10% donor calf
serum. Six hours later, concentrated hydroxyurea stock solution is added
to each dish to reach a final concentration of 1.5 mM hydroxyurea. After
the cells are blocked with hydroxyurea for 14 hr the medium is removed,
the plates are washed, and new medium without hydroxyurea is added.
During the entire process, prewarmed washing solutions and medium are
always used. These precautions are taken to avoid the effect of
temperature-change shock on the synchronization of cells.
Degree of Synchrony. With BHK cells and derivatives of BHK cells,
the above-mentioned procedure gives a good synchrony and little toxicity
(Fig. 4). ~6 Upon removal of hydroxyurea, the cells quickly enter S phase
in a synchronized fashion. The procedure is therefore satisfactory if cells
synchronized in S are desired. The amount of DNA per dish increases by
about 80%, which is about as much as one can get with hydroxyurea using
concentrations that will synchronize the cells at the G1/S boundary.
Lower concentrations will let cells go through the S phase, although
slowly; higher concentrations are more toxic.
26 H. Chang and R. Baserga, J. Cell. Physiol. 92, 333 (1977).
260
SPECIALIZED TECHNIQUES
[20]
12-
100
IG
lo
90
!o
I-3
FIG. 4. Synchronization of ALl06 by a combination of serum restriction and hydroxyurea (HU) treatment. ALl06 cells were plated on 100-mmpetri dishes for 3 days. Cells
were then cultured in medium containing 1% serum for 2 days. Fresh medium containing
10% serum was then given and HU (1.5 mM) was added 6 hr later. HU was removed at zero
time on the abscissa. Cells were pulsed with [aH]dT for 3 hr at the times indicated for
determination of specific activity. Cells were continuously labeled for autoradiography. Bar,
specific activity of DNA; OpO, /.,g DNA/dish; X--X, % labeled cells. (Reprinted, with
permission, from Chang and Baserga.~e)
Variations. The time periods given do not apply to all cells, as one can
easily understand. Some cells may require more than 48 hr of serum
restriction to achieve quiescence. Other cells may have a longer prereplicative phase and, therefore, h y d r o x y u r e a must be added at a later time.
Generally speaking, one should try out the optimal conditions for each cell
line. Our suggestion is that one should not use more than 14 hr of exposure
to h y d r o x y u r e a ; otherwise toxicity can be very high. Similarly, it should
be emphasized that the final h y d r o x y u r e a concentration of 1.5 m M can
apply to certain cell lines and not to others. For instance, AF8 do better at
1.25 m M whereas a hybrid between AF8 and L N S V , called A L l 0 6 , does
better at 1.5 mM. Again, the investigator is advised to test the optimal
concentration of h y d r o x y u r e a which will induce a G ~/S b o u n d a r y block
with a minimal amount of toxicity.
Mironescu and Ellem ~r have suggested that pretreatment o f serumrestricted cells with either h y d r o x y u r e a or cytosine arabinoside, before
they are stimulated to proliferate, increases the degree of synchrony of
entry into S phase. The effect has no relationship to the role of hyd r o x y u r e a or cytosine arabinoside as specific inhibitors o f D N A synthesis,
[20]
CELL SYNCHRONIZATION
261
since the serum-restricted cultures were mitotically quiescent; and a similar enhanced response can be induced in density-inhibited cultures by
only 2 hr of prestimulation exposure to cycloheximide, an inhibitor of
protein synthesis. Since we do not have any direct experience of the
ability of this treatment to increase the synchrony of density-inhibited
cells, the reader is referred to the original paper 27 for details.
Advantages and Disadvantages. This is an excellent method for obtaining cells synchronized at the G1/S boundary and the subsequent S phase.
With an appropriate choice of manipulations (length of serum deprivation,
percentage of serum during the restriction period, amount of and length of
exposure to hydroxyurea), toxicity can be low.
C. SYNCHRONIZATION BY DOUBLE-THYMIDINE BLOCK
262
SPECIALIZED TECHNIQUES
[20]
et al. 2a have shown that, when the concentration of calcium in the ex-
tracellular medium is reduced to 10/~M, WI-38 go into a state of quiescence and do not enter DNA synthesis. It may take more than 48 hr to
bring most of the WI-38 cells out of the cycle. Increasing the calcium
concentration to 1.35 mM at this point immediately produces a burst of
DNA synthesis which is highly synchronous. The rapidity with which this
burst occurs suggests that a decreased amount of calcium may synchronize the cells very closely to the onset of DNA synthesis. It should be
noted that as usual transformed cells cannot be synchronized by this
method but continue to grow even in very low concentrations of calcium. ~9
E. OTHER CHEMICAL METHODS
[21]
RECONSTITUTED
BASEMENT MEMBRANE RAFTS
263
[21] N e w T e c h n i q u e s f o r C u l t u r i n g D i f f e r e n t i a t e d Cells:
Reconstituted Basement Membrane Rafts
By LOLA M. REID and MARCOS ROJKIND
264
SPECIALIZED TECHNIQUES
[21]
[21]
265
16 I. Fentiman, J. Taylor Papadimitriou, and M. Stoker, Nature (London) 264, 760 (1976).
lr W. R. Loewenstein, Proc. Can. Cancer Res. Conf. 8, 162 (1969).
~8 N. S. McNutt, R. A. Hershberg, and R. S. Weinstein, J. Cell Biol. 51, 805 (1971).
~a J. D. Pitts and R. R. Burk, Nature (London) 264, 762 (1976).
20 G. R. Cunha, Int. Rev. Cytol. 47, 137 (1976).
21 p. Davies, A. C. Allison, and C. J. Cardella, Philos. Trans. R. Soc. London 271, 363
(1975).
22 R. Fleischmajer and R. Billingham, eds., "Epithelial-Mesenchymal Interactions." Williams & Wilkins, Baltimore, Maryland, 1968.
23 C. Grobstein, Science 11, 52 (1953).
24 R. L. Pictet, and W. L. Rutter, in "Cell Interactions in Differentiation" (M. KarkinenJaafkelainen, L. Fafen, and L. Weiss, eds.), p. 339. Academic Press, New York, 1977.
25 j. W. Saunders, Jr., M. T. Gasseling, and M. D. Gfeller, J. Exp. Zool. 317, 39 (1958).
2~ F. L. Vaughan and I. A. Bernstein, Mol. Cell. Biochem. 12, 171-179 (1976).
2r H. Green, J. G. Rheinwald, and T. Sun, in" Shape and Surface Architecture," p.493. Allen
R. Liss, Inc., New York, 1977.
266
SPECIALIZED TECHNIQUES
[21]
rated from the feeder cells. 2ra The type of stromal cell associated with the
epithelium is also important as shown by Sakakura and her associates who
demonstrated that the differentiation of mouse mammary gland depends
on association with mammary stroma but not other types of stroma. 2s
Conclusive evidence for epithelial-mesenchymal interactions in adult
tissues is not yet available. However, such a relationship is suggested by
the histopathological observation that the anatomical relationship between the epithelium and the mesenchyme exists in normal and benign
tumor tissue but is missing in malignant tissues. Malignancy is, in fact,
defined in solid tissues when the basement membrane is disrupted 29.3and
when either epithelial cells alone (carcinoma) or mesenchymal cells alone
(sarcoma) proliferate with concomitant loss of the epithelialmesenchymal association. This observation may explain, in part, why
malignant ceils establish in cell culture more easily than do normal cells;
normal cells may be interdependent with other cells in the tissue matrix,
whereas malignant cells may be qualitatively or quantitatively independent of these interactions.
Some of the secretions of the epithelium and the mesenchyme found in
the basement membranes are known to be important for the maintenance
of cultured epithelial cells. Best studied of the basement membrane components are the collagenous proteins, adhesion proteins, and
glycosaminoglycans.
Collagen has been used frequently for establishing primary cultures
since most normal epithelial cells will attach more efficiently to collagen
than to other cell culture substrates. 4 This is true despite the fact that
basement membrane collagen (Type IV collagen) is genetically distinct
from the collagen typically used in cell culture (Type I). More detail on
collagen types is given in subsequent sections. Floating collagen substrates have been used by Pitot's group for normal liver cells 31 and by
Pitelka's group az for normal mammary epithelium to extend the survival
of tissue-specific functions of those epithelial cells for up to a month. The
advantages of floating collagen substrates over collagen plates (in which
collagen is present at the bottom of the plate) are thought to be increased
oxygen tension at the air-media interface and the ability of epithelial cells
to contract the raft and, thereby, to undergo a change in cell shape apparently essential for differentiated cellular functions, al-aa
27a G. Martin, personal communication.
T. Sakakura, Y. Nishizuka, and C. J. Dawe, Science 194, 1439 (1976).
29 L. Liotta, L Leinerman, P. Catanzaro, and D. Ryrnbrandt, J. Natl. Cancer Inst. 58, 1427
(1977).
a0 S. Read, Guy's Hosp. Rep. 123, 53 (1974).
3~ G. Michalopoulos and H. C. Pitot, Exp. Cell Res. 94, 70 (1975).
3~ j. T. Emermanand D. R. Pitelka, In Vitro 13, 346 (1977).
83 T. Allen and C. S. Potten, Nature (London) 264, 545 (1976).
es
[21]
267
268
SPECIALIZED TECHNIQUES
[21]
of sulfated or acetylated disaccharides that are often associated with proteins forming complexes referred to as proteoglycans. The glycosaminoglycans and proteoglycans are present in all connective tissues where they
form a gelatinous charged matrix. The best known and most studied of the
glycosaminoglycans and proteolgycans include hyaluronate (skin, aorta),
dermatan sulfate (skin, tendon, aorta), chondroitin sulfate (cartilage,
bone, cornea, notochord, skin), heparan sulfate (aorta, lung, liver), and
heparin (lung, liver, skin). Their chemical structures, biosynthesis,
specific tissue localization, variations among species, and evolution are
reviewed elsewhere. 49
In dovelopmental systems it has been shown that glycosaminoglycans
and proteoglycans together with collagenous proteins can have a significant influence on cellular differentiation, 5'51 proliferation, ~2aggregation, 53
and migration. 54 Hyaluronate stimulates aggregation of chick embryo
ceUs. 54 In the development of the cornea and the notochord, hyaluronate is
thought to play a role in collagen fibril formation that in turn induces
cellular migration. Further development of these tissues involves replacement of hyaluronate with sulfated glycosaminoglycans that are the
typical polyanionic glycans of the adult tissues. Development of the
mouse salivary gland requires the presence of chondroitin sulfate at the
cell surface. Specific enzymes that eliminate the chondroitin 6-sulfate
from the developing tissues arrest their development; morphogenesis can
be reactivated by renewed biosynthesis of the proteoglycan. 51'55-57
In adult tissues the functions of the proteolgycans and glycosaminoglycans in cellular differentiation and proliferation are uncertain.
Kraemer's studies 5s-6 indicate that mammalian cells in cell culture are
coated with a variety of glycoproteins and polyanionic glycans among
which the most common is heparan sulfate. Kraemer hypothesizes that
49 M. B. Mathews, "Connective Tissue: Marcromolecular Structure and Evolution."
Springer-Verlag, Berlin and New York, 1975.
50 E. A. Balazs and B. Jacobson, in "The Amino Sugars" (E. A. Balazs and R. W. Jeanloz,
eds.), Vol. 2A, pp. 281-309. Academic Press, New York, 1966.
51 M. R. Bernfield, S. D. Banerjee, and R. H. Cohen, J. Cell Biol. 52, 674 (1972).
52 M. Lippman, in "Epithelial-Mesenchymal Interactions" (R. Fleischmajer and R. E. Billingham, eds.), p. 208. Williams & Wilkins, Baltimore, Maryland, 1968.
B. Pessac and V. Defendi, Science 175, 898 (1972).
54 B. P. Toole and R. L. Treistad, Dev. Biol. 26, 28 (1971).
55 M. R. Bernfield and S. D. Banerjee, J. Cell Biol. 52, 664 (1972).
58 R. H. Cohn, S. D. Banerjee, and M. R. Bernfield, J. Cell Biol. 73,464 (1977).
57 R. H. Cohn, J.-J. Cassiman, and M. H. Bernfield, J. Cell Biol. 71,280 (1976).
58 p. M. Kraemer, in "Biomembranes" (L. A. Manson, ed.), Vol. 1, p. 67. Plenum, New
York, 1971.
59 p. M. Kraemer, Biochemistry 10, 1437 (1971).
a0 p. M. Kraemer, Biochemistry 10, 1445 (1971).
[21]
269
270
SPECIALIZED TECHNIQUES
[21]
[21]
271
gens in skin, liver, heart, and kidney, but is absent from normal hyaline
and elastic cartilages. Type II collagen has a chain composition of
[al(II)]a; it is the only collagenous component of hyaline and elastic cartilages. Type III collagen has a chain composition of [al(III)]a; it is present
in most tissues containing Type I collagen except for bone, tendon, and
tooth. 62-~ Collagen also has been isolated from structures identified morphologically as basement membranes. Despite suggestions that these collagens are homogeneous with a 'chain composition of [al(IV)]a, the evidence is more convincing that basement membranes constitute another
class of proteins containing collagenous sequences, ranging in molecular
weights from 25,000--100,000. 65-67 Collagens isolated from basement
membranes differ in amino acid composition from interstitial collagens in
that they contain 3-hydroxyproline, relatively more hydroxylysine, and
relatively less alanine. Recently tissues containing Type I and Type III
collagens have been found also to contain collagenous components similar
in amino acid composition to those found in basement membrane collagens. 68"6a Since these collagens are extracted from tissues and not from
just the basement membranes alone, they are referred to as basement
membrane-like collagens. The basement membrane-like collagens as well
as Type III collagen are solubilized mainly by limited proteolysis with
pepsin.
Collagen used as a cell culture substrate 21'7.71 or as a raft 31,32 is obtained from skin or tendon by extraction with neutral salt solutions or
dilute acid. These extracts contain primarily Type I collagen that has been
shown to substantially lengthen the survival in culture of liver cells al and
mammary epithelium, az Since other types of collagen are part of the in
vivo substrate for epithelial cells (e.g., in normal liver, Type III collagen 7z'ra and basement membrane-like collagens TM rather than Type I collagen are in close association with the hepatocytes), these collagens also
62 p. M. Gallop and M. A. Pax, Physiol. Rev. 55, 418 (1975).
K. I. Kivirikko and L. Risteli, Med. Biol. 54, 159 (1976).
64 E. J. Miller, Mol. Cell. Biochem. 13, 165 (1976).
6.~ B. G. Hudson and R. G. Spiro, J. Biol. Chem. 247, 4229 (1972).
66 B. G. Hudson and R. G. Spiro, J. Biol. Chem. 247, 4239 (1972).
67 T. Sato and R. G. Spiro, J. Biol. Chem. 251, 4062 (1976). See also, this volume [6].
68 E. Chung, K. Rhodes and E. J. Miller, Biochem. Biophys. Res. Commun. 71, 1167 (1976).
69 R. L. Trelstad and K. R. Lawley, Biochem. Biophys. Res. Commun. 76, 376 (1977).
70 M. B. Bornstin, Lab. Invest. 7, 134 (1958).
71 W. D. Hillis and F. B. Bang, Exp. Cell Res. 17, 557 (1959).
72 L. Biempica, R. Morecki, C. H. Wu, M. A. Giambrone, and M. Rojkind, Gastroenterology 73, 1213 (1977).
73 S. Gay, P. P. Fietzek, K. Remberger, M. Eder, and K. Kuhn, Klin. Wochenshr. 53, 205
(1975).
272
SPECIALIZED TECHNIQUES
[2 l ]
0951).
[21]
273
274
SPECIALIZED TECHNIQUES
[21]
[21]
275
276
SPECIALIZED TECHNIQUES
[21]
sinization; as soon as the cells begin to detach from the plates, medium
containing serum is squirted onto the plates. All serum (chicken serum is
an exception) contains a trypsin inhibitor. Thus, squirting the cells with
the serum-supplemented medium will complete the detachment from the
plate and inactivate trypsin. The cell suspension is centrifuged in a desktop centrifuge at 900 rpm at 4, the supernatant liquid is removed, and the
pellet of cells resuspended in cold (4) medium containing serum. The cell
suspension should be kept on ice until ready for addition to the collagen or
RBM substrates.
2. TRANSPLANTABLE TUMORS
[21]
277
centrifuged at 1000 rpm for 10 min, and the serum is decanted into a fresh
sterile Falcon conical tube. Varying dilutions from 1 : 4 to 1 : 500 are made
o f antiserum in serum-free media.
b. Test for Antibody Titer. The antiserum at the varying dilutions is
added as a 1% solution in serum-free medium along with 1% unimmunized
rabbit serum as a source of complement; it is tested on both the host cells
and the tumor cells. A dilution is selected that gives a high titer of activity
against host cells and a minimal one against tumor cells. To test the titer, a
cell suspension or plate of cells attached to a substrate is rinsed with
phosphate-buffered saline, treated with the appropriate dilution of antiserum + complement, and the cells incubated at 37 for 1 hr after which
they are rinsed with phosphate-buffered saline and incubated in fresh
medium containing serum. Within an hour, the host cells will lyse and
only t u m o r cells remain.
3. PRIMARY TUMORS AND NORMAL TISSUE
278
SPECIALIZED TECHNIQUES
[21]
CO~ in air. Once the cells attach to substrates, the substrates can be
released to form rafts by rimming the gelatinous layer with a sterile
spatula. The substrates will float and can be transferred from one plate to
another by gentle pipetting with a large-mouth pipette. In culture, the rafts
with attached epithelial cells will contract to as little as one-half their
original size.
[22]
AUTORADIOGRAPHY
279
[22] Autoradiography
By
GRETCHEN
H.
STEIN
and
ROSALIND
YANISHEVSKY
280
SPECIALIZED TECHNIQUES
[22]
lest (approximately 2300 /~ diameter) of the three, z NTE, which is designed for electron microscope autoradiography, produces very small
grains (approximately 500/~ diameter). 3 NTB-3 is approximately twice as
sensitive as NTB-2, which is twice as sensitive as NTB or NTE. 4,s Because increased sensitivity means that less energy is needed to make an
activated crystal develop into a silver grain, the background increases
with the sensitivity.
Ilford Ltd. (Ilford, Essex, Great Britain) also manufactures nuclear
emulsions that cover a range of sensitivities and grain sizes. Ilford L4 is
frequently used for electron microscope autoradiography because it has a
small grain size (1200 A) and is much more sensitive than NTE. Recently,
Kodak has produced an improved emulsion for electron microscope autoradiography, which is also more sensitive than NTE. It is called Kodak
Special Product Type 129-01.
Dipping Vessels. We use Lab-Tek No. 4310 Cyto-Mailers as dipping
vessels (see Fig. 1). These vessels are thrown away when the emulsion is
discarded.
Slide Boxes. Small black Bakelite slide boxes (Scientific Products),
which hold 25 slides, are used for storing the slides during the exposure
period. Some investigators use this type of slide box as a light-tight box,
provided that it is sealed with tape. However, we prefer to store the taped
slide boxes inside a second light-tight box.
Desiccant. We include small packets of Drierite in the slide boxes
during the exposure period because nuclear emulsion is most sensitive
when it is dry and because moisture can cause latent image fading. The
packets of Drierite, including some blue indicator Drierite, are wrapped in
several layers of cheesecloth and secured with tape.
Autoradiography Darkroom
An autoradiography darkroom should exclude all light. This demand
exceeds the normal requirements of a photographic darkroom. (A model
darkroom for autoradiography is described by Kopriwa.6) The darkroom
should be inspected for light leaks when the eyes have adapted to darkness (a minimum of 15 min). Sources of light entry may be window and
door frames, door steps, ventilation shafts, or where pipes pass through
floors. Light may also come from electrical contacts that spark on switch2 L. G. Caro, Methods Cell Physiol. I, 327-363 (1964).
a M. M. Salpeter, Methods Cell Physiol. 2, 229-253 (1966).
4 A. Ron and D. M. Prescott, Methods Cell Biol. 4, 231-240 (1970).
5 A. W. Rogers, "Techniques of Autoradiography." Am. Elsevier, New York, 1973.
6 B. M. Kopriwa, J. Histochem. Cytochem. 11,553 (1963).
[2 ]2]
AUTORADIOGRAPHY
281
FIc. 1. A Lab-Tek Cyto-Mailer, which is used as a dipping vessel, is shown on the left.
The position of a slide rack used for drying emulsion-coated slides is shown on the right.
ing, equipment containing vacuum tubes that emit light from the rear,
clocks and watches that have luminous dials, and indicator lights such as
those on waterbaths. These objects should be removed or taped over to
shut out the light. Fluorescent lights continue to glow for an appreciable
time after being turned off and, if possible, should not be used in the
darkroom. The darkroom should have an entrance corridor with double
doors so that one may enter or leave without letting light into the darkroom.
A safelight may be used during autoradiography according to the
emulsion manufacturer's recommendations. For example, a dark red
Wratten No. 2 filter can be used for Kodak NTB, NTB-2, and NTB-3
emulsions. The power of the light bulb should be no greater than 15 W.
The safelight housing must be light tight so that light escapes only through
the filter. Safelights and filters may be purchased from Kodak. The
safelight should be mounted about 1 m above the working space. Turn off
the safelight when it is not needed, especially when the slides are drying,
because dry emulsion is the most sensitive.
Basic Procedure for Autoradiography of Tissue Culture Cells
1. Cell Culture
282
SPECIALIZED TECHNIQUES
[22]
The coverslips are cleaned before use by: (1) 5 min in 95% ethanol to
remove grease; (2) 5 min in 1 N HCI; (3) 5 min in deionized water, followed by four short rinses in deionized water; (4) two rinses in 95%
ethanol; and (5) two rinses in 100% ethanol. They are sterilized in a 250
oven for 7 hr.
Cells seeded in small petri dishes and in four-well multiplates tend to
settle more heavily in the middle of these vessels. This creates different
growth conditions at the center and periphery. In addition, the cells may
be too crowded in the center for single cell analysis by autoradiography.
We have found that filling the vessels very full when the cells are seeded
reduces this problem. After the cells have attached, the volume of medium may be reduced. We use 5 ml medium to seed cells in 35-mm petri
dishes and 12 ml medium per well in four-well multiplates.
Tissue sections fixed to slides, or cell suspensions smeared on slides,
may be processed for autoradiography in the same way as cells grown on
coverslips. 1The use of a cytocentrifuge to flatten cells in suspension onto
slides is discussed in the section on self-absorption.
2. Fixation
We fix the cells by adding to the growth medium an equal volume of
3 : 1 methanol : acetic acid fixative that has been freshly prepared. After 10
min, the half-strength fixative is removed by aspiration and replaced with
an equal volume of undiluted methanol: acetic acid fixative. After 10 rain,
the coverslips are removed and allowed to air dry. Methanol : acetic acid
fixative can cause some bursting of cell plasma membranes; however, it
flattens the cells well. By adding the fixative to the medium first, we have
eliminated this problem for our experiments with human cells.
[22]
AUTORADIOGRAPHY
283
FIG. 2. T98G human glioblastoma multiforma cells were labeled with 4 /~Ci/ml
[aH]thymidine(specific activity 50 Ci/mM) for 10 min. The cells were fixed in methanol : acetic acid (3 : 1) and extracted with 5% TCA. The slides were processed according to our basic
procedure, using an exposure time of 3 days. The cells were stained with Giemsa stain.
Silver grains are concentrated over the nuclei that synthesized DNA during the labeling
period.
4. Coverslip Mounting
The coverslips are mounted cell side up on clean microscope slides
using a drop or two o f P e r m o u n t (Fisher) or Euparol (Carolina Biological).
Generally we let the mounting medium dry for several hours at 60 , or
overnight at r o o m temperature, before the slides are dipped.
5. Preparation of Emulsion
We routinely use K o d a k NTB-2 for analysis of [aH]thymidine incorporation into tissue culture cells because it has low background, produces
grains large enough to count at x400 magnification, and needs only 3-4
days exposure to develop 25-50 grains/labeled nucleus under our experimental conditions (0.01 /~Ci/ml [aH]thymidine for 24 hr).
The emulsion is a gel at room temperature and is melted in a waterbath
at 39-43 . Although NTB-2 can be handled under a K o d a k Wratten No. 2
284
SPECIALIZED TECHNIQUES
[22]
safelight filter, we suggest keeping the light off whenever possible, e.g.,
when the emulsion is melting. Gently stir the emulsion with a clean glass
rod to check its fluidity after approximately 30 min, being careful not to
create air bubbles.
It is useful to test the background of a new bottle of emulsion by
dipping a clean slide into the bottle, letting the slide dry for 2-3 hr in a
light-tight box, and then developing it. One bottle of emulsion (118 ml) is
enough to coat 1000-3000 slides. 7
Because repeated heating of the emulsion to melt it causes an increase
in the background, we dispense new emulsion into Lab-Tek Cyto-Mailers
and store them at 4. For subsequent experiments, a single Cyto-Mailer of
emulsion is melted for 15 min and used as a dipping vessel. In general, 10
ml of NTB-2 and 10 ml of distilled water are added to each vessel and
mixed gently. For relatively flat samples, diluted emulsion forms an adequate layer of emulsion. The major advantage of using diluted emulsion is
that subsequent staining is easier. For preparations that have an uneven
surface, undiluted emulsion covers better.
[22]
AUTORADIOGRAPHY,
285
7. Exposure
We expose autoradiograms at 4 to minimize latent image fading. 5
However, for short-term exposures room temperature is also satisfactory.
The duration of the exposure depends on the experimental procedure
used and the grain dengity desired. The way to determine an exposure
time is to include several test slides in the experiment. These can be
developed at various times and used to estimate the correct exposure time
for the experiment.
8. Development
We use Kodak D19 developer and Kodak Fixer 197-1746, a generalpurpose hardening fixer. These solutions are stored in brown bottles and
are discarded after 2 months. If the D19 turns yellow before then, it is
discarded. To develop slides, unused developer and fixer are poured into
staining dishes and are used at 18-20 maximum. The slides are allowed to
warm to room temperature and are placed in staining racks. Slides coated
with either NTB, NTB-2, or NTB-3 are developed for 2 min in D19, rinsed
10-30 sec in 1% acetic acid stop bath, and fixed for 5 min in Fixer. After
fixing, the slides are rinsed gently in running tap water for 20 min, given a
final rinse in distilled water, and air dried. Ideally, the running tap water
should be at the same temperature as the other solutions used to avoid
stressing the gelatin by temperature changes. A thermostatic regulator,
which may be purchased from a photographic supply store, can be used to
maintain the appropriate temperature. However, a regulator is a convenience rather than a necessity.
9. Staining
We use Giemsa Blood Staining Stock Solution (J. T. Baker Chemical
Co.) diluted 1:25 in 0.01 M sodium phosphate at pH 7.1. Developed
autoradiographs are stained for 50 min and destained for 5 min in the same
buffer. Thereafter, the film of emulsion on the back of the slide is wiped
off with a damp Kimwipe. This procedure gives good definition of the
nucleus and cytoplasm without staining the nucleus so darkly that grains
over it are difficult to count. Much shorter staining and destaining periods
286
SPECIALIZED TECHNIQUES
[22]
may also be used successfully. The choice of a staining procedure depends on the nature of the experiment. Other stains are discussed below.
Other Techniques
Prestaining
Prestaining a specimen before applying nuclear emulsion generally
yields a clearer image than poststaining the developed autoradiogram.
This is so because, in prestaining, the gelatin of the emulsion layer is not
stained and the specimen stains more vividly and precisely. However,
only a few stains may be used for prestaining because most stains cause
increased background (positive chemography) even if they are removed
before the emulsion is applied. Prestains are useful for observing morphological details that may be obscured by overlying silver grains after
autoradiography. For example, chromosomes can be more easily identified before autoradiography. Stains used to detect enzymic reactions are
often used as prestains because the autoradiographic process alters or
destroys the enzymic reaction of interest)
Basic fuchsin, used in the Feulgen procedure, and aceto-orcein are the
most commonly used prestains. Even these acceptable prestains can
cause a high background if they are of low purity or are old. Loss of
radioactive material during staining is also a potential problem in using
prestains. The hydrolysis step of the Feulgen procedure may remove
some radioactivity from the specimen; however, for most qualitative studies, the loss of radioactivity is insufficient to alter the interpretation of the
results. ~,~0Aceto-orcein, used as a prestain, fades during the photographic
processing. Furthermore, aceto-orcein cannot be used as a poststain because it removes developed silver grains. The Feulgen procedure also
cannot be used for poststaining because grains are removed during the
hydrolysis step.
Poststaining
An effective poststain must overcome the presence of a layer of emulsion over the specimen and the chemical changes brought about by the
photographic processing. If the gelatin takes up too much stain, there will
be little contrast between the specimen and the stained gelatin. If the
gelatin affects the staining reagents, or provides an environment of inapR. Baserga and K. Nemeroff, Stain Technol. 37, 21 (1962).
10 W. Lang and W. Maurer, Exp. Cell Res. 39, 1 (1965).
[22]
AUTORADIOGRAPHY
287
288
SPECIALIZED TECHNIQUES
[22]
[22]
AUTORADIOGRAPHY
289
oxazolyl)benzene] dissolved in dioxane. The emulsion becomes impregnated with scintillator molecules. Photons are released as /3 particles
pass through the scintillator. These photons activate more silver crystals
in the emulsion than would have been activated by the/3 particles alone.
Theoretically, the efficiency of the high-speed method of tritiumlabeled molecules could be 30-fold that of conventional autoradiography. 26 However, different investigators have had variable success with
this technique; in some cases only a small increase in efficiency was
produced and was accompanied by an increase in the background.
Self-Absorption
Increasing the thickness of a radioactive specimen beyond a certain
point does not increase the number of/3 particles entering the emulsion.
This phenomenon is due to self-absorption of the/3 particles by the specimen. The distance that/3 particles can penetrate depends on their initial
energies and on the density of the specimen. Because most of tritium' s /3
particles penetrate less than 3 /zm through cellular material, 3 selfabsorption is an important factor in autoradiography of tritium-labeled
whole cells. Nevertheless, tritium is useful for autoradiography because it
has a short pathlength and, therefore, can be localized better than more
energetic isotopes.
Tissue culture cells are sufficiently large that they must be in a flat
configuration for tritium autoradiography in order to minimize self~70. L. Miller, Jr., G. E. Stone, and D. M. Prescott, Methods Cell Physiol. 1, 371-379
(1964).
zs S. B. Horowitz, Methods Cell Biol. 8, 249-275 (1974).
2a W. E. Stumpf, Methods Cell Biol. 13, 171-193 (1976).
30 W. Maurer and E. Primbsch, Exp. Cell Res. 33, 8 (1964).
290
SPECIALIZED TECHNIQUES
[22]
absorption. Many types of cells flatten out on the solid substrate on which
they are grown. Other types of cells are round, e.g., cells that are
grown in suspension. In addition, flat cells become round when they are in
mitosis and when they are removed from the substrate, e.g., with trypsin.
Spreading may take from 2-12 hr, depending on the cell type, the substrate, and the culture conditions. Generally, cells spread more rapidly on
tissue culture plasticware than on glassware. Coating glass with a layer of
evaporated carbon enhances spreading, al When round cells are to be used
for autoradiography, they can be flattened onto microscope slides with a
cytocentrifuge (Shandon Southern Instruments, Sewickley, Pennsylvania). The chambers of a cytocentrifuge are designed so that the centrifugal force drives the cells against a slide at the back of the chamber.
The cells are flattened on the slide and are then fixed as usual.
The choice of fixative affects the degree of flattening of the cells. Even
cells that are well spread in culture are several micrometers thick. For
example, Chinese hamster ovary cells in monolayer culture are about 7
/~m thick and can benefit from additional flattening. We have found that
3 : 1 methanol : acetic acid flattens cells much more effectively than neutral formalin fixative. The ratio of methanol : acetic acid may have to be
modulated to obtain an intact, well-flattened specimen; increasing the
concentration of acetic acid will flatten cells but may disrupt cellular
integrity.
Uniform self-absorption in flattened cells simply reduces the efficiency
at which/3 particles are detected by the emulsion, whereas nonuniform
self-absorption can create artifacts. For example, cells increase in size
during the cell cycle, and large G2 cells may not be flattened to the same
thickness as small G1 cells. Consequently, the efficiency could be less in
the G2 cells. Nonuniform self-absorption is also inherent within individual
cells because the density of various organelles differs: the nucleus is less
dense than the cytoplasm, which is less dense than the nucleolus, z
Autoradiographic Background
The most common causes of background are listed and discussed
briefly. More detailed discussions are given by Boyd, n2 Baserga, a3 and
Rogers. 5
1. Photographic Processing. Excessive background will occur if the
temperature of the developing solutions is high, or if the duration of
development is long. This problem can usually be recognized because
31G. A. Meek, "Practical Electron Microscopyfor Biologists." Wiley,New York, 1970.
32G. A. Boyd, "Autoradiographyin Biologyand Medicine." Academic Press, New York,
1955.
[22]
AUTORADIOGRAPHY
291
292
SPECIALIZED TECHNIQUES
[23]
[23] I n d u c t i o n a n d P r o d u c t i o n o f I n t e r f e r o n
By ROBERT M. FRIEDMAN
Many cells in culture can be stimulated to produce at least some
interferon. This is a convenient property because it permits the study of a
useful cell product with a specific biological activity, inhibition of virus
growth. Stimulation of interferon by cells in culture appears to be due to
induction of a protein that is not ordinarily made. Recent studies have
suggested that interferon production is controlled by a repressor ~ and is
determined by specific loci on chromosomes that have in some cases been
identified. 2
I R. L. Cavalieri, E. A. Havell, J. Vil~ek, and S. Pestka, Proc. Natl. Acad. Sci. U.S.A. 74,
4415 (1977).
2 p. p. Creagan, Y. H. Tan, S. Chen, and F. H. Ruddle, Fed. Proc., Fed. Am. Soc. Exp. Biol.
34, 2222 (1975).
[23]
INDUCTION
A N D P R O D U C T I O N OF I N T E R F E R O N
293
294
SPECIALIZED TECHNIQUES
[23]
[23]
295
Cell line
(reference)
RSTC-2 (4)
RSTC-2
FS-4 (3)
FS-4
Treatment
Time
(hours)
Interferon
yield
0-1
0-1
0-4
1-4
500 a
10,000 a
poly IC (100/~g/ml)
poly IC
+ cycloheximide (50 tag/ml)
+ actinomycin D (1 ttg/ml)
0-2
0-- 2
2-6
5.5-6
100b
10,000 b
ducer for only an hour and then to wash the cells 3-5 times with saline or
medium. In addition, DEAE-dextran (10-100 ~g/ml) may be used to increase poly IC adsorption. The yield of interferon in human FS-4 cells
induced with poly IC alone (20/~g/ml) is usually less than 1000 units/ml. 3,4
Several steps may be taken with poly IC induction to increase interferon yields, but the single most effective way to do this is a superinduction
procedure that employs actinomycin D and cycloheximide or puromycin. 3,4 The timing of the addition of antimetabolites is critical. Monolayers
of cells confluent for at least 6 days are usually used, especially if interferon is being induced in diploid cells. After the cultures are washed with
buffered saline, Eagle's Medium (MEM, serum-free) containing poly IC
(5-20/zg/ml) and cycloheximide is added. After 5 hr, actinomycin D (final
concentration 1-5/zg/ml) is added to the medium for 1 hr and, 6 hr after
induction, the medium is removed and discarded. The cells are washed 4
times with saline and MEM with 0.2% human plasma protein (U.S.P.) or
2% fetal calf serum is added. The medium containing interferon is collected 30 hr after the onset of the production run. The yields of interferon
in FS-4 or RSTC-2 human diploid cells with this procedure are at least
10,000-20,000 reference units/ml so that a very significant potentiation of
interferon production may be obtained. However, it is important to note
that superinduction with antimetabolites does not work in all systems.
Mouse L cells, for instance, are not superinductable for interferon production. Table I reviews the results obtained with superinduction of interferon in two systems.
296
SPECIALIZED TECHNIQUES
[24]
[24] E v a l u a t i o n o f C h e m i c a l C a r c i n o g e n i c i t y b y in Vitro
Neoplastic Transformation
METHODS
IN ENZYMOLOGY,VOL. LVIII
[24]
CHEMICAL CARCINOGENITY
297
cell type and assay procedure that have been recorded, one of the more
rapid and reliable is described in this article; a it uses a fibroblast line of
baby hamster kidney cells, BHK 21/cl 13, 4 and assays for its transformation after a brief treatment by suspected carcinogen by testing the ability
of the treated cells to form colonies in soft agar. 5
This established BHK cell line offers advantages over cells newly
placed in culture in that cells grow rapidly (12-hr doubling time at 37),
transform with high frequency in response to a variety of carcinogens, 3,6-8,9'1'11 clone readily to give homogeneous populations, and are
amenable to culture for periods of time sufficient to carry untreated controls to the end of an experiment. The technique is sometimes criticized
because the cells may have already undergone one or more steps in the
process leading to malignant transformation, being "immortal" whereas
normal diploid cells are not. However, in defense of the method it should
be noted that recently cloned BHK cells are not tumorigenic in vivo at
moderate doses, TM whereas their virally and chemicallys transformed derivatives are. Thus the transformation induced in vitro clearly represents a
very crucial step in any series of changes that may be required for
tumorigenesis.
The soft agar assay for in vitro transformation, although slightly more
cumbersome than some others, has the advantage of selecting in a single
step for cells capable of anchorage-independent growth. This particular
characteristic of the transformed phenotype of a fibroblast cell is the only
selective one that is consistently correlated with in vivo tumorigenicity. 13
Cells and Culture Conditions
A culture of BHK 21/cl 13 should be obtained, preferably from someone currently working with it, at as low a passage level as possible. It
a N. Bouck and G. di Mayorca, Nature (London) 264, 722 (1976).
4 M. Stoker and I. Macpherson, Nature (London) 203, 1355 (1964).
5 I. Macpherson and L. Montagnier, Virology 23, 291 (1964).
6 y. Ishii, J. A. Elliott, N. K. Mishra, and M. W. Lieberman, Cancer Res. 37, 2023 (1977).
7 R. F. Newbold, C. B. Wigley, M. H. Thompson, and P. Brookes, Murat. Res. 43, 101
(1977).
s G. di Mayorca, M. Greenblatt, T. Trauthen, A. Soller, and R. Giordano, Proc. Natl.
Acad. Sci. U.S.A. 70, 46 (1973).
9 I. F. H. Purchase, E. Longstaff, J. Ashby, J. A. Styles, D. Anderson, P. A. Lefevre, and
F. R. Westwood, Br. J. Cancer 37, 873 (1978).
10 j. A. Styles, Br. J. Cancer 36, 558 (1977).
1i j. Ashby, J. A. Styles and D. Anderson, Br. J. Cancer 36, 564 (1977).
12 O. Jarrett and I. Macpherson, Int. J. Cancer 3, 654 (1968).
13 S. Shin, V. H. Freedman, R. Risser, and R. Pollack, Proc. Natl. Acad. Sci. U.S.A. 72,
4435 (1975).
298
SPECIALIZED TECHNIQUES
[24]
[24]
CHEMICAL CARCINOGENITY
299
lute the stock solution in TD buffer 14 (0.8% NaC1, 0.038% KCI, 0.01%
Na~HPO4, 0.3% Trizma base, and HCI to pH 7.0 to give a range of concentrations that are double the final desired doses. The final concentration
of dimethylsulfoxide in contact with the cells should be less than 2%.
2. Harvest by trypsinization cells that have been grown from the selected clone, wash them by centrifugation in TD buffer containing 2% calf
serum, and resuspend them at 2 106 cells/ml in TD buffer without
serum.
3. Mix equal volumes of cell suspension and diluted carcinogen, usually 1-3 ml each, in sterile tubes containing small magnetic stirring bars
and stir very gently for 1 hr at 37. Include a zero-dose control tube
containing solvent and cells but no carcinogen.
4. At the end of the hour, dilute the cell-carcinogen mixture with 5-10
volumes of complete medium containing 20% serum, recover the cells by
centrifugation, and wash and resuspend them in fresh medium with 20%
serum.
5. To determine the amount of cell killing due to the carcinogen treatment, dilute an aliquot of the treated cells and plate in quandruplicate in
complete medium with 20% calf serum at densities of 200, 2000, or 20,000
cells/60-mm dish. The number of cells added per dish depends on the
expected killing. It is safest to plate at several cell densities from a given
dose. Count cplonies arising on these plates after incubation for 7 days at
37 .
6. Plate another aliquot of the treated and washed cells at a concentration of about 2 105 cells/100-mm dish in complete medium with 20%
serum and grow at 37 for 4 days. If, during this time, any dishes become
more than 70% confluent, the cells should be subcultured. At the end of
this expression period, cultured cells from each dose should be assayed in
soft agar as described below.
300
SPECIALIZED TECHNIQUES
[24]
[24]
CHEMICAL CARCINOGENITY
301
Variations
If the action of a particular carcinogen requires that the cells be actively growing during exposure, they can be treated on plates instead of in
suspension. The problem with treating growing cells is that it provides an
opportunity for the carcinogen to act as a selective agent and enrich the
proportion of preexisting transformants.
~6R. J. Munsonand D. T. Goodhead,Mutat. Res. 42, 145 (1977).
302
SPECIALIZED TECHNIQUES
[25]
B H K can activate effectively a variety of carcinogens, and its hydrolytic enzymes are present at a good constitutive level. However, if there is
any doubt about the capacity of the cells to activate a given carcinogen, a
liver extract 17 may be added before and/or during the treatment with the
carcinogen. 10
A period of growth is usually required between carcinogen treatment
and challenge with soft agar to attain the maximum expression of transformation by newly induced transformants. Recently, however, a B H K
subclone has been reported 8 that apparently is able to carry out the divisions necessary for maximum expression while suspended in soft agar,
thereby eliminating the growth period in liquid culture. Substituting
Noble agar 6,1 or agarosC a for Difco Bacto agar may also allow one to
skip the growth period in liquid. This is useful for large screening programs, although the number of background colonies is high and maximum
transformation frequencies may not be achieved.
17 B. N. Ames, W. E. Durston, E. Yamasaki, and F. D. Lee, Proc. Natl. Acad. Sci. U.S.A.
70, 2281 (1973).
is I. Macpherson, in "Tissue Culture Methods and Applications" (P. F. Kruse and M. K,
Patterson, eds.) p. 277. Academic Press, New York (1973).
[25] I n d e p e n d e n t
By
ROBERT
B.
CAMPENOT
[25]
303
FIG. 1. A petri dish (35 mm, Falcon, viewed from above) divided into three chambers by
a Teflon divider (7.5 mm high) sealed to the floor of the dish (a collagen-coated coverslip)
with silicone grease. The floor of the narrow (1 x 5 mm) central chamber (a), in which the
neurons are plated, is transected by 20 parallel scratches that serve to guide the growing
neurites into the left (b) and fight (c) side chambers.
petri dish. The grease is applied to the entire face of the divider that is to
mate with the coverslip, and the two are pressed together. Before assembling the system, 20 parallel scratches are made in the collagen-coated
coverslip (Fig. 1); since the growing neurites tend not to cross these
scratches and are thus confined to the collagen-coated channels between
them, the scratches guide the growing neurites into the side chambers (see
below). Just before assemblying the system, a drop of L-15 COs medium
containing Methocel (composition given in this volume [53]) is placed on
the scratched region of the coverslip; this prevents the silicone grease on
the divider from adhering to the coverslip in this region. After applying
the drop of medium, the Teflon divider and coverslip are gently pressed
together so that the scratched, medium-coated region spans the narrow
central chamber and extends into the side chambers to the left and right
(Fig. 1). Although the silicone grease does not adhere to the coverslip in
this region, it prevents significant bulk flow of medium between the central and side chambers; fluid-level differences of about 5 mm between the
304
SPECIALIZED TECHNIQUES
[25]
central and side chambers are supported for 4 days without appreciable
equalization.
The somas of principal neurons dissociated from superior cervical
g a n g l i a o f n e w b o r n r a t s ( p r o c e d u r e s a r e gPcen in this v o l u m e [53]) a r e t h e n
p l a t e d i n t o t h e n a r r o w c e n t r a l c h a m b e r . T h e n e u r i t e s g r o w p a r a l l e l to t h e
s c r a t c h e s a n d t r a v e r s e t h e b a r r i e r s , p r e s u m a b l y in a t h i n film o f m e d i u m
b e t w e e n t h e g r e a s e a n d t h e c o v e r s l i p . T h e n e u r i t e s e m e r g e i n t o t h e side
c h a m b e r s a f t e r a b o u t 3 o r 4 d a y s in c u l t u r e ( s e e Fig. 2).
Construction a n d U s e of t h e System
[25]
305
petri dish, and the coverslip is bonded over the hole on the outside suface
of the dish with Sylgard 184 encapsulating resin (Dow Coming). A day or
two before plating the neurons, the coverslip is coated with a dried collagen film (see this volume [53]). Twenty parallel scratches about 200/zm
apart are made in the collagen-coated coverslip with a rake made by
cementing together 20 insect pins; a rubber mat under the coverslip helps
to make the depth of the scratches uniform. The dishes are then sterilized
with ultraviolet light.
Teflon Dividers
Teflon dividers are machined from I-in. rod stock. They are 7.5 mm in
height; the central chamber is about 1 mm wide and 5 mm long. The side
chambers hold about 0.5 ml of medium; the central chamber communicates with the rest of the dish that holds 1-2 ml. The slotted shape of the
central chamber protects the neuronal somas from disturbance when the
medium sloshes during handling of the dish.
The first time the dividers are autoclaved, they are distorted and their
faces must be sanded flat with fine sandpaper; subsequent autoclaving
does not distort them further. Before each use, the dividers are wiped
clean of silicone grease and sequentially washed briefly in Nochromix
(Godax Laboratories Incorporated), in 95% ethanol for 24 hr, and in
flowing, deionized water for several hours; they are then autoclaved in
glass-distilled water, rinsed several times in glass-distilled water, and finally placed in a glass petri dish and sterilized by autoclaving. They must
be completely dry before use.
306
SPECIALIZED TECHNIQUES
[25]
[25]
307
NGF withdrawal from a chamber into which the neurites have grown
stops the growth of the neurites; it also appears to result in slow degeneration of the neurites, even though the somas and proximal portions of the
neurites continue to have access to NGF. (c) When NGF is withdrawn
from the chamber containing the somas, the neurons appear to survive
only if their neurites have crossed into chambers where NGF is continuously available. In these experiments, the fluid level in the NGF-free
chambers was always kept at a higher level than that in adjacent chambers
that contained NGF, thereby eliminating transfer of NGF by bulk flow.
Acknowledgments
This work was performed in the laboratoryof Edwin J. Furshpan and David D. Potter.
Support was provided by National Institutesof Health Research Grants NS-02253, NS03273, NS-11576, and Training Grants NS-07009 and NS-07112.
2 p. A. Walicke, R. B. Campenot, and P. H. Patterson, Proc. Natl. Acad. Sci. U.S.A. 74,
5767-5771 (1977).
308
SPECIALIZED TECHNIQUES
[26]
[26] M u t a n t I s o l a t i o n
B y LARRY H. THOMPSON
During the last decade somatic cell genetics has developed into a
recognized discipline--largely because of the development of techniques
for somatic cell hybridization and methods for the isolation of a variety of
mutant phenotypes. Already the spectrum of mutant cell lines available is
sufficient to have a substantial effect on approaches to studying metabolic
regulation and growth control in animal cells. In this article the term
" m u t a n t " will be used to refer to heritable phenotypic changes that appear to arise from changes in DNA structure, as evidenced by stability,
response to mutagens, the presence of altered gene product, and other
criteria. Despite some earlier concerns, an epigenetic basis for variant
phenotypes has received little documentation and does not appear to be a
major complicating factor in the isolation and use of mutations. Most
mutant lines that have been isolated behave in a reproducible way and are
suitably stable in phenotype, provided a minimal number of technical
precautions are taken.
The diploid nature of somatic cells undoubtedly remains a major barrier to isolating mutants. Since the great majority of mutations are recessive when tested in somatic cell hybrids, such mutations will only be
expressed in autosomal genes when both alleles are altered. There is some
evidence suggesting that certain established cell lines, particularly the
well-studied Chinese hamster ovary (CHO) lines, may contain significant
hetero- or hemizygosity. 1'2 The precise extent of this functional haploidy,
however, is still unclear since other evidence indicates that the cells behave essentially as diploid. 3 In general, when planning a mutant isolation,
one should assume that the gene coding for the protein (or RNA) molecule
of interest is probably present in two copies. This constraint does not
imply that mutants for such loci cannot be isolated, but it does suggest
that a m u c h greater effort will be required than in the case of an X-linked
function such as the commonly studied hypoxanthine phosphoribosyltransferase (HPRT).
Because many mutants are found at very low frequency, even after
potent mutagenesis, the probability of a successful isolation may be directly proportional to the magnitude of the experiment, i.e., to the number
1L. Siminovitch,Cell 7, 1 (1976).
L. Siminovitchand L. H. Thompson,J. Cell. Physiol. 95, 361 (1978).
3 M. J. Siciliano, J. Siciliano, and R. M. Humphrey,Proc. Natl. Acad. Sci. U.S.A. 75,
1919 (1978).
M E T H O D S IN ENZYMOLOGY, VOL. LVIII
ISBN 0-12-181958-2
[26]
MUTANT ISOLATION
309
of cells screened. Herein lies a major difficulty. At best, one can handle
about l0 s cells per liter of medium under selective conditions; in
monolayer culture this number of cells would typically be distributed
among 50 or more 100 mm-petri dishes. These plating requirements set a
practical lower limit on detectable mutant frequencies at = 10-a, i.e., one
mutant from 500-1000 dishes (5-10 liters of medium). Thus, the isolation
of mutations, especially of the temperature-sensitive conditional class, at
loci that require simultaneous alteration in both alleles will often be very
difficult. However, for certain "recessive" mutations it is possible to
obtain the homozygous ( - / - ) state by a two-step process because the
heterozygous ( + / - ) state is selectable on the basis of a slight difference in
phenotype (such as degree of drug resistance, discussed subsequently for
the aprt locus) from the wild type. Only a few mutations having dominant
(or incompletely dominant expression) have been identified; these include
resistance to a-amanitin, ouabain, Methotrexate, ricin, and colchicine.
Choice of Cell Line and Culture Requirements
310
SPECIALIZED TECHNIQUES
[26]
tially associated with chromosomal change. Mutant lines that have been
carefully examined were found to resemble closely the wild type. 9
Another attractive feature of CHO cells is that they are not unreasonably fastidious in terms of culture requirements, and they can be
grown under a variety of conditions. A high plating efficiency, an important property for quantitative mutant isolation, is routinely achievable.
Unlike most other hamster lines, CHO cells can be grown in liquid suspension culture, in soft agar, on top of soft agar, 1 as well as in monolayer.
In ot-MEM medium lacking nucleosides (K. C. Biological, Inc., Cat. No.
DM-325) but containing 10% fetal bovine serum CHO cells have a doubling time of about 12 hr both in monolayer and suspension culture.
Not surprisingly, a number of different wild-type CHO strains exist,
having different karyotypes, growth rates, morphology, ease of growth in
suspension, and other properties. This variety should be kept in mind
when choosing a strain for mutant isolation. There is as yet no evidence
that any particular strain is generally more mutable than another, except
in some specific instances where certain clones behave as though they are
hemizygous at a specific autosomal locus that was not intentionally sub:
jected to selection pressure. 2
Suspension vs. Monolayer Culture
[26]
MUTANT ISOLATION
311
TABLE I
GENERALIZEDOUTLINEFOR MUTANTISOLATION
Approximate
number of cell
generations
1
6
12
12
2
24
57 generations
(43 days a)
312
SPECIALIZED TECHNIQUES
[26]
Induction of Mutants
[26]
MUTANT ISOLATION
313
3. After incubation for 16 hr the mutagen is removed. This is accomplished by centrifugation and resuspension in buffered saline to rinse the
cells, and then suspension in fresh medium at a reduced cell concentration
to allow for growth of the culture.
Independent Cultures
It is often important to know that individual mutants represent independent mutational events and not siblings. In order to achieve this condition, the experiment should be designed so that separate cultures are
mutagenized or that a large culture is subdivided immediately following
mutagen treatment before there has been opportunity for mutations to
become replicated as siblings within the population.
Direct Selection of Drug-Resistance Mutants
As a general class of mutants, drug-resistance mutations are among the
easiest to isolate because the selective situation favors the direct outgrowth of the mutant clones. This approach can be used in any situation in
314
SPECIALIZED TECHNIQUES
[26]
[26]
MUTANT ISOLATION
315
tion. In this example o f selecting a presumptive heterozygote, it is essential that the drug concentration be carefully adjusted for the desired
survival value. It is also necessary to demonstrate explicitly an increased
drug resistance and reduced e n z y m e activity for the presumptive mutant
colonies after they have been isolated since some colonies may not be
mutant for statistical reasons, 1~ and resistance may involve other loci.
Often, more than one gene locus is involved in the mutation to resistant phenotypes, as in the case of Methotrexate, TM diptheria toxin, 13 or
colchicine; TM with each agent, isolation of mutations in the intracellular
target protein is complicated by the presence of permeability variants.
Since different genetic classes of resistance may occur at different drug
concentrations, the drug dose must be chosen accordingly.
Details o f Plate Inoculation and Incubation
316
SPECIALIZED TECHNIQUES
[26]
were loosely attached or floating over the colony but then reattached
elsewhere on the dish. The dishes should be moved only if it is absolutely
necessary to replenish the medium because of drug instability or other
problem. The presence of large numbers of dead cells on the dish is not
esthetically pleasing but generally does not interfere with the selective
conditions and can be ignored.
Mutant colonies that are to be isolated for characterization should be
taken while fairly small, approximately 1 mm in diameter, to minimize the
risk of cross-contamination associated with large colonies on the dish. At
a normal growth rate, CHO cells produce this size colony in about 6-7
days, but mutants under drug selection may require incubation of 2-3
weeks. In any mutant selection, control dishes with cells in drug-free
medium should be set up with 200-300 cells per dish in order to determine
the plating efficiency. This control is important for calculating the mutant
frequency, and it gives an indication that the incubation conditions were
valid. Control dishes are usually stained sooner than selection dishes and
should, therefore, be placed on separate incubator trays to avoid the
problem of colony dispersal mentioned above. Procedures for isolating
the colonies are discussed in the final section.
Multistep Selections
When either more than one gene or more than one allele can be altered
to effect resistance, it may be desirable to produce highly resistant cells
by performing additional selections at increasing drug concentration on
clonal isolates. For example, it might be necessary to obtain a permeability mutant initially, before selecting for an alteration in an intracellular
molecule. Mutagenesis can be carded out as needed at each round of
selection.
[26]
MUTANT ISOLATION
317
318
SPECIALIZED TECHNIQUES
[26]
[26]
MUTANT ISOLATION
319
medium by pipetting directly 1.0 ml of cell suspension into each dish and
dispersing it.
8. The dishes are incubated at 34 for 16-18 days before isolating the
colonies. (See section below on clone isolation.) Other versions of the
above procedure can presumably be designed to obtain a specific synthetase mutation by conducting the experiment at a single temperature and
altering a particular amino acid concentration to a low level in the selective medium, and to a high level during mutagenesis expression and colony formation. Such a procedure should select for specific amino acid
hyperauxotrophs--mutants that require an abnormally high concentration
of an amino acid for growth. These will likely prove to be temperature
sensitive also in many cases.
320
SPECIALIZED TECHNIQUES
[26]
[26]
MUTANT ISOLATION
321
because it is more rigid and less water absorbing. With this procedure it
seems possible to screen the colonies from 104-105 mutagenized cells
when as many as 500-1000 colonies per dish are replicated. While it is not
clear that more than one useful replicate can be made, even one would
facilitate the isolation of mutants such as the UV-sensitive clone of
CHO-K1 cells that has been reported. 24
Mutant Colony Isolation and Cloning
Isolating Colonies from Petris Dishes
As indicated earlier, when colonies are being taken from petri dishes
as presumptive or prospective mutants, it is preferable to do this when
they are about 1 mm in diameter; larger colonies are more prone to shed
loose cells. Our standard procedure involves removing the colonies with a
Pipetman autopipette, using the 0.2 ml plastic disposable tips that are
autoclavable. The colonies should be rinsed twice with serum-free medium to r e m o v e loose cells as soon as they are taken from the incubator,
and then circled with a marking pen. Each colony is removed simply by
scraping the circled area with the pipette tip while applying simultaneous
suction by slow release of the plunger. This effectively removes the cells
with little damage. A dish should be rinsed once or twice again before
each additional colony is taken to minimize the chance of crosscontamination. The purity of colonies isolated in this manner is sufficiently high for effective mutant screening when done with the precautions indicated. The method is very simple and amenable to the isolation
of large numbers of colonies using standard laboratory equipment. Another, perhaps more rigorous, method involves isolating each colony with
a stainless-steel cylinder by using autoclaved vacuum grease, and then
trypsinizing the cells.17,25
Cloning in Multiwell Plastic Trays
To ensure genetic homogeneity, it is desirable to clone rigorously cultures that have been identified as mutant. This is done conveniently with
96-well cluster trays having good optical quality, e.g., the CoStar ( # 3 5 % )
or N u n c (#N-1480) trays. Wells are inoculated with 0.2 ml of a cell suspension in medium such that about one well in every 3 to 4 receives a
viable cell. The low concentration minimizes the probability of getting
two cells in one well. When the colonies are recognizable with an inverted
24T. D. Stamato and C. A. Waldren, Somat. Cell Genet. 3, 431 (1977).
~5T. T. Puck, P. I. Marcus, and S. L. Cieciura, J. Exp. Med. 103, 653 (1956).
322
SPECIALIZED TECHNIQUES
[?7]
[27] Karyotyping
By
Introduction
K a r y o t y p i n g should not be an e n d e a v o r restricted to the cytogeneticist. Rather, it is an e x t r e m e l y valuable r e s e a r c h tool for anyone who
works with cell cultures, regardless o f the p r i m a r y field of interest.
M o s t biologists k n o w that the g e n o m e o f higher plants and animals is
organized into discrete c h r o m o s o m e s , that the c h r o m o s o m e s can be set
out in groups to f o r m a k a r y o t y p e , and that the k a r y o t y p e is characteristic
o f the species. M a n y will be less familiar with the fact that in p e r m a n e n t l y
established cell lines there can be extensive k a r y o t y p i c variation f r o m line
to line derived f r o m the s a m e species, or f r o m cell to cell within a line.
The biochemist, therefore, should b e aware of this genetic heterogeneity
in order to properly interpret cell culture data, and m a y on occasion use
this heterogeneity to a d v a n t a g e in designing n e w experiments.
When tissues are explanted and put in culture, the cells that grow
generally display the k a r y o t y p e of the individual as, for example, in direct
bone m a r r o w cultures, short-term l y m p h o c y t e cultures, or p r i m a r y fibroblast cultures. H o w e v e r , w h e n cell cultures b e c o m e established the rigid
control o v e r the k a r y o t y p e is generally lost, and c h r o m o s o m a l variation
[9. 7]
KARYOTYPING
323
Uses of Karyotyping
1. Gene Dosage. In many types of biochemical research, including, for
example, studies on regulation of enzyme synthesis, it may be desirable to
know whether the cell line is diploid, pseudodiploid, or heteroploid.
Clearly, such studies will be more valid when carried out with diploid
cells rather than pseudodiploid or heteroploid cells. Furthermore, information is rapidly accumulating on the human chromosomal map location
for many enzymes so that in the future the biochemist may want to know
the details of which chromosomes or chromosomal parts are duplicated or
deleted in order to estimate the role that gene dosage may play in the
regulatory phenomenon under investigation.
2. Identification of Cell Lines. Marker chromosomes generated spontaneously by structural rearrangement in cell cultures are usually characteristic of the line, and three or four such unique markers can provide an
unequivocal identification test for a cell line. That such an identification
test is important was realized a few years ago when it was found that a
number of presumably different cell lines, thought to have been derived
from a variety of different human tissues, were not different lines at all,
but were in fact sublines of the heteroploid HeLa cell line derived many
years ago from a human cervical carcinoma. Much of the proof for this
startling discovery consisted of the finding of several marker chromosomes, characteristic of HeLa cells, in the other cell lines. 1Contamination
of one cell line by another is a potentially enormous problem for the
investigator who studies a tissue-specific enzyme or tissue-specific celluW. A. Nelson-Rees and R. R. Flandermeyer, Science 191, 96 (1976).
324
SPECIALIZED TECHNIQUES
[27]
lar function. Since most established lines do carry one or more characteristic marker chromosomes, karyotyping provides a handy method for
verifying that one is working with the correct cell line. Indeed, in many
instances the karyotype may be the only means available to check the
identity of a cell line.
3. Monitoring of Cell Lines. In many experiments, the cell line being
studied comes in deliberate contact with other cell lines, with subcellular
preparations derived from other cell lines, or with viruses grown in other
cell lines. Examples of direct cell contact would include enzyme crossfeeding experiments or the passage of tumor cells through animals. Experiments involving contact with subcellular preparations such as chromosome transfer experiments carry the danger of cross-contamination by
live cells from the donor line. In all these cases karyotyping provides a
method for monitoring cell lines to confirm that cross-contamination has
not taken place in the course of an experiment.
4. Verification of Hybrid Formation. Cell hybrids (described in this volume [28]) are extensively used for biochemical studies. In inter-species
hybrids the chromosomes provide a definitive set of markers to confirm
that the putative hybrid is in fact a hybrid of the desired type. In intraspecies hybrids, the use of parental cell lines with different characteristic
marker chromosomes can often provide a means of verifying that the
hybrid is the desired one.
5. Cytogenetic Research. In the four examples cited above karyotyping
is used as an aid in a research project that is not cytogenetic in nature.
Other work is concerned with gene mapping or karyotype evolution in
which karyotyping plays a major role. These projects often involve the
routine procedures described in detail below. They may also make use of
more highly specialized procedures outlined briefly below but the details
for which are beyond the scope of this article.
[:27]
KARYOTYPING
325
II
FIG. 1. A human metaphase spread stained first by solid staining (A), then destained by
placing the slide in 95% ethanol for 10 min, and G-banded (B).
bands (Fig. 1B) that correspond to the bright quinacrine bands; (3)
R-banding or reverse banding, a technique involving heat treatment of the
slides and Giemsa staining that results in banding patterns in which the
alternating light and dark regions are the reverse of those seen with
G-banding.
In this article we will deal with solid staining and G-banding in considerable detail, and the reader is referred to the original literature or to
review articles z-5 for details of Q- and R-banding.
In addition to the three major banding techniques there are several
other specialized procedures that will not be detailed in this article although they are mentioned briefly here to provide an idea of the available
technology. These include: (1) C-banding, a procedure for specifically
staining the centromere regions of chromosomes as well as regions of
constitutive heterochromatin; 6 (2) a technique in which a fluorescent dye
is used to stain centromeres of mouse but not human chromosomes, useful for identifying the species of origin of chromosomes in human-mouse
2 T. C. Hsu, Annu. Rev. Genet. 7, 153 (1973).
3 p. Pearson, J. Med. Genet. 9, 264 (1972).
4 0 . J. Miller, D. A. Miller, and D. Warburton, Prog. Med. Genet. 9, 1 (1973).
5 B. Dutrillaux and J. LeJeune, Adv. Hum. Genet. 5, 119 (1975).
6 F. E. Arrighi and T. C. Hsu, Cytogenetics 10, 81 (1971).
326
SPECIALIZED TECHNIQUES
[27]
Detailed Methods
In this section we provide details for preparing microscope slides of
air-dried metaphase chromosome spreads, and the detailed techniques for
solid staining and for G-banding. The solid staining requires less expertise
than G-banding and is adequate for counting chromosomes or for identifying uniquely shaped marker chromosomes. The G-banding procedure is
more demanding and requires higher-quality metaphase spreads, but it
gives considerably more karyotypic information than does solid staining
(see Fig. 1A and B)
Preparation of high-quality chromosome spreads suitable for banding
contains an eleliaent o f " a r t , " and because of this the preparations usually
become better with practice. As is the case for the typical enzyme purification procedure, quality varies slightly from day to day, and the optimal
procedures vary somewhat from one cell type to another.
Slide Preparation
[2 7]
KARYOTYPING
327
achieve a high mitotic index, (2) treatment with a mitotic inhibitor to allow
cells to accumulate at mitosis, (3) swelling of the cells in a hypotonic
environment, (4) fixation of the cells, and (5) spreading and drying of the
mitotic cells on a slide. Some general considerations with regard to each
step are described below.
1. Optimization of Growth. For cultured cells, a high mitotic index is
usually easy to achieve. For some cell lines, changing the medium the day
before making chromosome preparations may be helpful. Rapidly growing
lines are usually subcultured 1-2 days before and slower growing lines 3-4
days before "harvesting" the cultures for chromosome studies.
2. Metaphase Arrest. Mitotic spindle inhibitors such as colchicine or
deacetylmethyl colchicine (Colcemid) are normally used for blocking cells
in metaphase. Exposure time depends on the length of the cell cycle
(longer treatments for slower growing cells), but 0.5-2 hr is usually sufficient. The concentration of Colcemid is not critical: we have found 0.11.0/zg/ml adequate for many cell lines. Higher concentrations can cause
excessive contraction of the chromosomes reducing the clarity of banding
patterns.
3. Hypotonic Swelling. Swelling of the cells is a critical step and is
essential to achieve well-spread chromosomes. A variety of hypotonic
solutions are in common use, but 0.075 M potassium chloride is the most
popular and is a good choice for the initial attempt. Treatment time in
" h y p o " varies from one cell line to another, but 15-45 min is standard
and, if periods greater than 45 min are required, the hypotonic solution
might be diluted to increase its effectiveness.
4. Fixation. This is another critical step, and it always follows the
hypotonic treatment. The near universal fixative is 3 parts methanol to 1
part glacial acetic acid. It is usually made fresh each day and kept cold
until use. It is always added slowly to prevent cells from clumping. Some
protocols suggest removal of the hypotonic solution from the cells and
subsequent addition of fixative, while others call for slow addition of the
fixative to the hypotonic solution containing the cells. Both methods
work, but a given cell line may respond better to one or the other method.
To insure complete fixation the fixative is changed 3-4 times before drying
the cells onto a slide.
5. Air Drying. Spreading of the cells on a slide is also a crucial step. If
not correctly performed either the mitotic cells will fail to spread and the
chromosomes will remain in a heap in the cell or they will spread too
much with rupture of the cell membrane and release of the chromosomes
from the cell. Cells are spread by drying them onto a slide. Optimum
spreading of the cells seems to be related to the drying of the fixative,
which in turn depends on the temperature and relative humidity in the
328
SPECIALIZED TECHNIQUES
[27]
[2 7]
KARYOTYPING
329
Solutions
Colcemid (Grand Island Biological Co., #521L), 10/zg/ml, stored at
4
Hypotonic: 75 mM KC1 warmed to 37 (5 ml per culture)
Fixative: 3 parts methanol to 1 part glacial acetic acid, made fresh on
day of harvest and kept on ice in a tightly stoppered vessel (20 ml
per culture)
Steps
1. OPTIMIZATION OF GROWTH. TWO days before harvesting the culture for chromosome spreads, subculture so that cells will be in mid to late
exponential growth phase on the day of harvest.
2. METAPHASE ARREST. TO 3 ml of lymphoblast culture add 2 ml of
fresh medium and 0.1 ml Colcemid. Incubate at 37 for 30 min.
3. HYPOTONIC SWELLING. Centrifuge cells at approx. 1000 rpm (200
g) for 8 min. Remove all but 0.2 ml of supernatant with suction apparatus.
Resuspend cells in this small volume, add 5 ml of warm hypotonic solution, and invert to mix. (Do not pipette cells because swollen cells are
fragile.) Incubate at 37 for 15 min.
4. FIXATION. Centrifuge cells at 1000 rpm for 8 min. Remove all but
about 0.2 ml of supernatant, and resuspend cell pellet in this small volume
by gentle agitation (no pipetting). Add first 1 ml of cold fixative slowly,
drop by drop with constant agitation. Add another 4 ml of fixative and
leave on ice for 15 min. Repeat fixation twice as above, but the fixative
can be added more quickly on the 2nd and 3rd fixation. Allow 15 min
between successive fixations. At this point cells can be stored overnight at
4 or slides can be made immediately.
5. AIR DRYING. Centrifuge cells at 1000 rpm for 8 min. Remove
supernatant and suspend cells in about 0.5 ml of cold fresh fixative. (Make
fresh fixative if cells have been stored overnight). Shake excess water and
ice from a microscope slide and, using a Pasteur pipette, drop 3-4 drops
of cell suspension onto the slide. Blow gently over the surface of the slide
to spread the cell suspension thinly, wipe excess liquid from the underside
of slide, and place on hot plate. Leave 5 min to dry thoroughly. Store
slides at room temperature in slotted trays or boxes so that surfaces do
not touch. (Note: It is our practice to prepare one slide and check it in the
microscope under phase contrast so that adjustments can be made on
subsequent slides if cells are too crowded or if spreading is poor--see
Table I.)
330
SPECIALIZED TECHNIQUES
[27]
TABLE I
COMMON PROBLEMS IN SLIDE PREPARATION AND SOME SUGGESTED SOLUTIONS
Problem
Suggested solutions
B. T H E
In
Situ
M E T H O D ( A s APPLIED TO H U M A N FIBROBLASTS)
Equipment
Lab-Tek chambers (single chamber, #4801, Lab Tek Products, Miles
Laboratories)
Pasteur pipettes and 22-gauge needle
Suction apparatus with collecting flask
Hot plate (e.g., Corning, model PC 351) covered with paper towel
and set to maintain 100 ml of water in a 100 ml beaker at 60-70 )
[27]
KARYOTYPING
331
Solutions
Colcemid (Grand Island Biological Co., #521L) 10/~g/ml, stored at 4
Hypotonic: 0.075 M KC1, warmed to 37 (2 ml per culture)
Fixative: 3 parts methanol to 1 part glacial acetic acid, made fresh on
day of harvest and kept on ice in a tightly stoppered vessel (10 ml
per culture)
Steps
1. OPTIMIZATION OF GROWTH. T w o days before harvesting, subcul-
ture cells from the flasks or plates in which they normally grow into two or
more Lab-Tek chambers. The cells should cover no more than 5-10% of
the growth surface after they are attached, and the final volume of medium is 4 ml. In order to prevent cells from settling primarily around the
edges, first add medium to the chamber and then pipette the concentrated
ceils into the center of the chamber and rock gently to distribute outwards. Incubate for about 2 days at 37. (Slow-growing strains may require 3 or more days.)
2. METAPHASE ARREST. On the day of harvest, cultures should be
nonconfluent (about 10-20% of surface covered by cells---see Fig. 2A)
and actively growing (several rounded-up mitotic cells or doublets should
be seen). Add 0.2 ml Colcemid and incubate for 90 min.
3. HVPOTONICSWELLING. With as little agitation as possible so as not
to detach loosely adhering mitotic cells, carry chamber slides from incubator. Very slowly, aspirate all of the culture medium by placing a
22-gauge needle connected to a suction flask into one corner of the
chamber. Add 2.0 ml of warm hypotonic solution drop by drop over the
surface of the chamber. Incubate for 30 min at 37.
4. FIXATION. Remove chamber slides very carefully from incubator
and, without removing any of the hypotonic solution, add 2.0 ml of cold
fixative drop by drop over the surface of the chamber. Let sit on the bench
for 20 min. Repeat fixation 3 times more at 15-min intervals, each time
aspirating all the fixative and replacing it with 2-3 ml fresh fixative. The
15-min interval is a minimum time, and longer times can be used if more
convenient. Slides can be dried immediately or stored at 4 overnight.
5. AIR DRYING. Aspirate all the fixative, remove chamber walls from
slide, blow gently over the surface of the slide to spread the cells, wipe
excess fixative from the underside of the slide, and place slide on hot plate
to dry. Leave at least 10 rain to dry thoroughly. Remove any rubber
cement remaining after removal of chamber walls and store slides at room
temperature in slotted trays or boxes so that slide surfaces do not touch.
332
SPECIALIZED TECHNIQUES
[27]
[27]
KARYOTYPING
333
Since different cell lines can differ considerably in their nuclear size,
amount of cytoplasm, number and size of chromosomes, and response to
a hypotonic environment, any single protocol will not work well for all cell
lines. We have had experience with a number of cell lines of human,
hamster, and mouse origin and summarize below some general guidelines
for them.
Most primary cultures of fibroblast-like cells, regardless of the species
or tissue of origin, can be harvested with the in situ procedure described
above for human skin fibroblasts.
Human amniotic fluid cells are primarily of two types, fibroblast-like
and epithelial-like, and these cultures respond well to the same in situ
procedure. In fact, for amniotic fluid cultures set up for prenatal diagnosis
it is common to initiate the cultures in Lab-Tek chamber slides and to
examine the chromosomes of individual colonies of cells in these primary
cultures. This has two advantages. It allows diagnosis to be made in the
shortest possible time (no subculturing is required), and it provides information regarding the colony of origin of any cell with an unusual
karyotype.
The CHO cell line, a permanent line of Chinese hamster ovary origin,
grows in suspension culture and can be harvested with the suspension
method described above but with a longer period in Colcemid (60-90
min).
CHO cells also grow in monolayer but are less well attached than
primary fibroblasts; at mitosis the cells round up and are easily dislodged
from the surface (Fig. 2B and C). Thus, while CHO can be harvested by
the in situ method (at the cell density shown in Fig. 2B), great care has to
be taken to avoid washing away all the mitotic cells during the hypotonic
and fixation steps. An alternative procedure, starting from a monolayer
(at the cell density shown in Fig. 2C), is to treat for 90 min with Colcemid,
then tap or shake the flasks (or plates) to detach many of the mitotic cells,
removing the medium with detached cells to a centrifuge tube. Any remaining mitotic cells can be recovered by lightly trypsinizing: the trypsin
solution used for subculture is added for 30-60 sec. After pooling the
trypsinized cells with the mitotic cells, we centrifuge, resuspend in
hypotonic solution, incubate at 37 for 30 min, and add an equal volume of
fixative to the hypotonic solution before the next centrifugation. The cells
are fixed twice more, and slides are prepared according to the suspension
protocol.
A number of other commonly used cell lines of Chinese hamster origin
(V-79, GM-7, M3-1, 18-1, DON), mouse origin (3T3, L, LMTK), or
334
SPECIALIZED TECHNIQUES
[27]
Staining Techniques
A. SOLID STAINING
While many different stains have been used to give uniform staining of
the chromosomes, Giemsa stain seems to be the most common and is the
one we use routinely. We use commercially prepared concentrated
Giemsa (Giemsa stain, Original Azure Blend Type, Harleco #620). Like
many cytogenetic labs we dilute into Gurr's buffer at pH 6.8 (Gurr's buffer
tablets, Searle Diagnostic, England, 1 tablet per liter of distilled water) but
10 mM potassium phosphate at pH 6.8 works equally well.
For solid staining, set up 3 coplin jars (glass jars with flat sides and
ridges that hold 5 slides) or 50-ml beakers as follows:
1. 50 ml of Gurr's buffer (or 10 mM phosphate at pH 6.8) containing
1.5 ml of concentrated Giemsa;
2. 50 ml of water;
3. 50 ml of water.
Place slides into the stain for 3 min and rinse by dipping 8-10 times in
each beaker of water. Shake off excess water and stand on end on a paper
towel to drain and dry.
If chromosomes are not sufficiently dark the slide can be returned to
the stain for a few minutes. The diluted Giemsa can be used to stain up to
about 50 slides and is discarded at the end of the day.
I . GIEMSA (G) BANDING
Giemsa banding or G-banding was so named because it was the first
banding procedure described to utilize this stain. However, it is
clearly a misnomer because the Giemsa stain is not the critical ingredient
in the procedure and also because Giemsa can be used for solid staining
(above) as well as in other banding procedures. In the original G-banding
technique the critical step was incubation of the slide at high temperature.
[27]
KARYOTYPING
335
This has largely been replaced by incubation of the slide in a crude trypsin
solution, and that has been our standard banding method for several
years.
The trypsin is a crude preparation (Bacto trypsin, #0153, Difco Laboratories, Detroit, Michigan) dissolved in 10 ml of distilled water and
stored frozen in 1-ml portions. The Giemsa stain is the same as that used
for solid staining (Original Azure Blend Type, Hareleco #620). The
Giemsa has a shelf life of only about 3 months at room temperature;
thereafter it may be used for solid staining but not for G-banding. As for
solid staining, the Gurr's buffer in which the stain is diluted can be replaced with 10 mM phosphate at pH 6.8.
A prerequisite for good G-banding is a high-quality slide. Chromosomes should be straight and slender with sister chromatids lying close
together. Cells should be well spread with few overlapped chromosomes,
but the cells should not be broken. Even on a good slide, only one
metaphase spread out of 10 or 20 will fit these criteria. Therefore, many
mitotic cells should be provided on the slide. These properties can be
checked by phase-contrast microscopy before proceeding with banding.
Our technique, modified only slightly from the original trypsinGiemsa technique of Seabright, 13is done at room temperature by passing
the slide through a series of 7 coplin jars or 50-ml beakers as follows:
1. 50 ml of 0.15 M NaCl--brief rinse
2. 50 ml of 0.15 M NaCI containing 1 ml of trypsin stock solution-30-120 sec
3. 50 ml of 0.15 M NaCl--brief rinse (dip slide 8-10 times)
4. 50 ml of 0.15 M NaCl--brief rinse
5. 50 ml of Gurr's buffer (or 0.01 M phosphate buffer, pH 6.8) containing 1.5 ml of concentrated Giemsa--l-2 min
6. 60 ml of distilled water brief rinse (dip slide 8-10 times)
7. 50 ml of distilled water--brief rinse
Slides are placed on end on a paper towel to drain and dry.
Since the time required in trypsin depends to some extent on how the
slides were prepared, especially the temperature of the hot plate, a few
trial slides may be necessary.
Following either solid staining or G-banding, slides are ready for microscopic study. We do not bother to mount a coverslip on the slides since
the presence of a coverslip does not seem to significantly affect the quality
of the microscopic images. When using oil-immersion lenses we place the
immersion oil on the surface of the slide and then remove it immediately
after use by dipping the slide into fresh xylene (xylol) several times. The
,3 M. Seabright, Lancet 2, 971 (1971).
336
SPECIALIZED TECHNIQUES
[27]
new imersion oils, if left on the slide, will extract the stain resulting in
fading of the image.
If a coverslip is to be mounted, add a drop of Permount (Fisher Scientific Co., #SO-P-15), or a 50:50 mixture of Permount and xylene. Coverslips can be removed if necessary by soaking the slide overnight in
xylene.
[27]
KARYOTYPING
337
TABLE II
COMMON PROBLEMSIN BANDINGAND SOME SUGGESTEDSOLUTIONS
Problem
Suggested solution
338
SPECIALIZED TECHNIQUES
[27]
FIG. 3. (A) A portion of a poorly spread cell, trypsin banded. The thick layer of cytoplasm over the chromosomes causes background staining (blue through the microscope, grey
on the print) and prevents the chromosomes from becoming clearly banded. It also refracts
the light making it impossible to focus clearly on the chromosomes. (B) A portion of a
well-spread, clearly banded cell for comparison with (A), (C), and (D). (C) A portion of an
over-trypsinized cell. (D) A portion of an under-trypsinized cell.
on the top at a reasonable cost. Automatic film a d v a n c e is an expensive
luxury and not necessary in m o s t cases. A u t o m a t i c e x p o s u r e control is
also u n n e c e s s a r y since the a m o u n t o f light reaching the film is relatively
constant f r o m one m e t a p h a s e spread to another, and f r o m slide to slide.
Thus, once e x p o s u r e time has been determined for a particular combination of m i c r o s c o p e , c a m e r a , and film, it need not be altered. A green filter
is c o m m o n l y used and gives sharper images on black and white film.
The choice o f film is not critical and any m e d i u m - c o n t r a s t black and
white film should provide adequate pictures. I f y o u develop y o u r o w n
films a local photographic supplier should be able to provide advice on the
choice of film and developer. One combination that we have used successfully is Tri-X (Kodak) film d e v e l o p e d in Microdol X (Kodak) d e v e l o p e r
according to the m a n u f a c t u r e r ' s instructions. Fixation in Rapid Fix
(Kodak) and w a s h p r o c e d u r e s are standard.
[27]
KARYOTYPING
339
340
SPECIALIZED TECHNIQUES
[27]
II If
tm
Ill
FIG. 4. ,The human male karyotype, G-banding. Top row: Groups A (chromosomes 1-3)
and B (4 and 5). Second row; Group C (chromosomes 6-12). Third row: Groups D (chromosomes 13-15) and E (16-18). Fourth row: Groups F (chromosomes 19 and 20) and G (21 and
22) and the sex chromosomes X and Y.
FIG. 5. The Chinese hamster (Cricetulus griseus) male karyotype, G-banding. Top row:
Chromosomes 1-5. Bottom row: Chromosomes 6--10 plus X and Y.
[27]
KARYOTYPING
341
ii
342
[27]
SPECIALIZED TECHNIQUES
T A B L E III
KARYOTYPE CHARACTERISTICS OF A FEW CELL STRAINS AND ESTABLISHED CELL LINES
Species
Human
(2n = 46)
Chinese
hamster
(2n = 22)
Syrian
hamster
(2n = 44)
Mouse
(2n = 40)
Rat
(2n = 42)
Modal no. o f
chromosomes
No. o f
marker
chromosomes
Reference to
karyotype
46
63-78
0
16--25
~.b
b-~
(HeLa
contaminants)
46
b-e
f
diploid
CHO
CHW
22
20-21
22
0
10-13
4
a.t
J
diploid
BHK
44
44
0
0
k
t
diploid
CAK
RAG
A9
LMTK-
40
41
approx. 60
approx. 60
52
0
1-3
approx 15
several
approx. 90
m
n
P
q
r
diploid
42
Name of
line
WI-38
(diploid
flbroblast)
HeLa
KB,Hep-2,
D98,HEK,
etc.
HT-1080
Drosophila
melanogaster
(2n = 10)
Variable
with time
[27]
KARYOTYPING
343
karyotyped. Although it is impractical to list all of the lines that have been
karyotyped, a few of the more commonly used lines are listed in Table III
with references to the detailed karyotypes.
In attempting to karyotype one's own cell line for the first time, there
are a few facts to keep in mind. First, chromosomes are continually contracting as the cell proceeds into mitosis and in so doing several fine bands
present on a chromosome of a prophase cell or early metaphase cell will
coalesce into a smaller number of thicker bands by mid-metaphase. Thus,
cells with long thin chromosomes are generally in an early stage of mitosis
and have more total bands (and hence different patterns) than cells with
shorter thicker chromosomes. With long Colcemid treatment, all cells
tend to be at the same stage, that represented in Figs. 4, 5, and 6, thereby
considerably reducing this source of variation.
Second, there is subtle variation in chromosomes between the individual members of a species. In humans, for example, this variation can be in
the width of certain bands that stain darkly by C-banding, or in the
fluorescent intensity of certain bands after staining with quinacrine. However, such inherited chromosomal variants or polymorphisms are not
readily apparent by solid staining or G-banding and should not present
major difficulties in karyotyping by these techniques,
Third, in comparing the karyotype of one's cell line with published
karyotypes, keep in mind that karyotypes evolve with continued culture.
rS. Rasheed, W. A. Nelson-Rees, E. M. Toth, P. Arnstein, and M. B. Gardner, Cancer
33, 1027 (1974).
~M. Ray and T. Mohandas, Cytogenet. Cell Genet. 16, 83 (1976).
hL. L. Deaven and D. F. Petersen, Chromosoma 41, 129 (1973).
~R. G. Worton, C. C. Ho, and C. Duff, Somatic Cell Genet 3, 27 (1977).
J P. A. Gee, M. Ray, T. Mohandas, G. R. Douglas, H. R. Palser, B. J. Richardson, and
J. L. Hamerton, Cytogenet. Cell Genet. 13, 437 (1974).
kN. C. Popescu and J. A. Di Paoio, Cytogenetics 11,500 (1972).
i R. Marshall, Chromosoma 37, 395 (1972).
m "Standard Karyotype of the Mouse," J. Hered. 63, 69 (1972).
hR. A. Farber and R. M. Liskay, Cytogenet. Cell Genet. 13, 384 (1974).
o The number of marker chromosomes is enhanced in mouse cell lines because the
normally telocentric (centromere at one end) mouse chromosomes tend tp fuse
together at the centromere creating biarmed markers that are not as unique as those
created by random breakage and reunion.
P E S. Hashmi, E W. Allderdice, G. Klein, and O. J. Miller, Cancer Res. 34, 79 (1974).
qE W. Allderdice, O. J. Miller, D. A. Miller, D. Warburton, P. L. Pearson, and
H. Harris, J. Cell Sci. 12, 263 (1973).
rA. R. Rushton, Can. J. Genet. Cytol. 15, 791 (1973).
' "Standard Karyotype of the Norway Rat," Cytogenet. Cell Genet. 12, 199 (1973).
t S. F. Dolfini, Chromosoma 58, 73 (1976).
344
SPECIALIZED TECHNIQUES
[27]
For example, a CHO line may have several normal Chinese hamster
chromosomes, .several marker chromosomes characteristic of CHO lines
already described, and additional new marker chromosomes not previously described that have evolved by structural rearrangements during
continuous cell culture. Because of this evolution, it is not valid to assume
that one's own CHO line will have the same karyotype as CHO lines in
other laboratories. If it is important for the interpretation of biochemical
or other data, then the karyotype of the specific subline under study must
be determined.
Comments
Karyotyping can be an extremely valuable research tool for anyone
working with cultured cells. In some instances, a quick chromosome
count may be all that is required. For this, solid staining would be sufficient, and photographs of chromosomes would not likely be required. The
solid-staining technique is easily learned, and for many cell lines one can
expect adequate chromosome spreads on the first attempt (also see this
volume [27]). Thus, it should take no more than a few hours to obtain a
chromosome count on 20-25 cells.
If more detailed information is necessary, then chromosome banding
may be required. The G-banding technique presented above will be adequate for most applications, although some situations may require one of
the special techniques that have been noted. Since the requirement for
well-spread chromosomes is greater for banding than for solid staining, a
few days may have to be invested in optimizing the techniques for one's
own cell line. Banded chromosomes are also difficult to analyze in the
microscope without considerable experience, thus making it necessary to
photograph, print, cut out, and arrange the chromosomes in a karyotype.
However, once the technique is developed, even a detailed banded analysis can be performed in a day or two.
While several days or even a few weeks invested in developing
karyotyping expertise may seem excessive, it is completely inconsequential compared to the many years that already have been spent on HeLa cell
research by scientists who thought they were studying something else. It
is similarly inconsequential when compared to the time that may be involved in studying gene regulation in heteroploid cell lines where abnormal gene dosage relationships may preclude the operation of normal regulatory mechanisms.
Put in these terms we suggest that karyotyping expertise is not only a
valuable tool, but often an essential one for the biochemist who works
with cultured cells.
[28]
CELL FUSION
345
[28] Cell F u s i o n
By ROGER H. KENNETT
Cell fusion is a biological phenomenon that has been studied and developed from two distinct points of view. Biochemists and cell biologists
have been interested in the role that the fusion of biological membranes
may play in normal cellular processes such as plasma membrane formation and in the movement of vesicles in the processes of endocytosis and
exocytosis.l On the other hand much progress in the areas of eukaryotic
genetics, 2 virology, 3 and the analysis of malignancy4 has been made possible by application of cell fusion techniques to the production of heterokaryons and hybrid cells. Heterokaryons result from the fusion of plasma
membranes of two different cell types so that one or more nuclei from
each type are included within a common cytoplasm. A hybrid cell is
formed when growth and division processes take place in a heterokaryon
and the genetic materials from the two types of nuclei are included in the
single nucleus of a proliferating cell. Such hybrid cells can then be propagated in culture with the growth characteristics usually depending on the
cell types fused. Generally, the hybrids can be maintained in culture
under the same conditions and for as long as the most actively growing of
the two parental cells. Several selective systems have been devised that
allow preferential growth of hybrid cells and eventual elimination of the
two parental types. These methods will be described below.
There has been interaction among those approaching cell fusion from
these two points of view at the level of the various agents used to promote
the fusion of cellular membranes. These promoters of membrane fusion
fall into two general categories: enveloped viruses and lipophilic or lipolytic reagents such as lysolecithin and polyethyleneglycols.5
I will concentrate here on those methods for cell fusion that allow
production of a high proportion of viable heterokaryons or hybrids and
will discuss the numerous ways in which such fused cells are useful to
investigators involved in analysis of biochemical and genetic properties of
cultured cells. For those who would like a more detailed account of the
i G. Poste, Int. Rev. Cytol. 32, 157 (1972).
2 V. A. McKusick and F. H. Ruddle, Science 196, 390 (1977).
3 H. Koprowski, Fed. Proc., Fed. Am. Soc. Exp. Biol. 30, 914 (1971).
4 G. Barski, in " T i s s u e Culture: Methods and Applications" (P. F. Kruse, Jr. and M. K.
Patterson, eds.), p. 469. Academic Press, N e w York, 1973.
5 G. Poste and A. C. Allison, Biochim. Biophys. Acta 300, 421 (1973).
346
SPECIALIZED TECHNIQUES
[28]
Expression
The ability to include genetic material from two cell types within the
same cytoplasm makes it possible to pose questions concerning the interactions of two sets of genes. The two types of questions that have been
asked are:
1. What positive or negative regulatory effects can be observed when
genetic material from two cells in different states of differentiation are
included in the same cytoplasm? Do elements from one genome turn on or
off genes in the other genome?
2. Is genetic complementation observed when nuclei from cells, each
deficient in some function, are included in the same cytoplasm?
In general, it can be concluded that when two cells expressing different
states of differentiation are fused, there is usually suppression of differentiated functions. 8 For example, fusion of melanomas with fibroblasts
produces hybrids that do not produce melanin as do the melanoma parental cells.a Similarly, fusion of a Friend erythroleukemia mouse cell line,
which can be induced to synthesize hemoglobin, with a human fibroblast
suppresses globin message formation when the mouse globin loci are present as shown by nucleic acid hybridization studies. TM
A notable exception to this rule is that fusion of a rat liver tumor cell
line synthesizing several liver-specific proteins with other types of cells
sometimes l'esults in hybrids that synthesize some of these proteins. 1~
There.are even reports that, when a rat liver tumor cell is hybridized with
some types of cells, e.g., human fibroblast, not only are the rat liver
proteins synthesized but, in some clones, liver-specific proteins are synthesized from the genes of the other species included in the hybrid.12 In
hybrids in which these liver-specific functions are suppressed, one often
finds reexpression of one or more of the liver-specific functions after the
6 B. Ephrussi, "Hybridization of Somatic Cells," Princeton Univ. Press, Princeton, New
Jersey, 1972.
7 H. Harris, "Cell Fusion," Harvard Univ. Press, Cambridge, Massachusetts, 1970.
s R. C. Davidson, Syrup. Soc. Study Dev. Biol. 31, 295 (1973).
9 R. L. Davidson, B. Ephrussi, and K. Yamamoto, J. Cell. Physiol. 72, 115 (1968).
10 A. Deisseroth, R. Velez, R. D. Burk, J. Minna, W. French Anderson, and A. Nienhuis,
Somatic Cell Genet. 2, 373 (1976).
~1 G. J. Darlington and F. H. Ruddle, Mod. Trends Hum. Genet. 4, 111 (1976).
12 G. J. Darlington, H. P. Bernhard, and F. H. Ruddle, Science 185, 859 (1974).
[28]
CELL FUSION
347
cells are grown in culture for several passages.13 This may result from loss
of chromosomes and a resulting change in relative gene dosage favoring
reexpression of one or more of the liver-specific proteins. These observations are consistent with the finding that when a near tetraploid derivative
of the liver cell line is fused with a diploid cell, the liver-specific functions
are expressed in most of the hybrids.14
The suppression and reexpression of differentiated functions in hybrids, and the possibility that this results from chromosome loss, point out
a significant difference between hybrids and heterokaryons. Hybrids start
out after the fusion event with at least one full set of chromosomes from
each parental cell. As the hybrids are continued in culture, variants arise
that have lost chromosomes derived from one or both of the parental cell
types. 15 This presumably is a consequence of unequal distribution of
chromosomes during mitosis and cell division and the growth advantage
that some of the resulting variants have over the original hybrid clone.
How rapidly a variant with a reduced chromosome number comes to
predominate in the hybrid culture depends on the types of cells fused and
the species from which they each originate. An extreme case of chromosome loss exists in the formation of most rodent cell human cell hybrids. In most cases, the human chromosomes are rapidly lost leaving only
a few human chromosomes in each hybrid clone.16 Since each clone has a
different set of a few human chromosomes, mouse human hybrids have
in fact been very useful tools for human gene mapping as indicated below. lr
One confirmed exception to the rule of extinction in heterokaryons
occurs when fibroblasts fused with chick erythroblast nuclei are reactivated and begin to synthesize globin. This also happens when the erythroblasts are fused with cells from which the nuclei have been removed. TM
In regard to the second question above, genetic complementation has
been demonstrated in several systems either by assaying the individual
heterokaryons or the mass culture after the fusion for the particular factor(s) missing from the unfused cells. 19,2 Another method is to detect
13 M. C. Weiss, R. S. Sparkes, and R. Bertolotti, Somatic Cell Genet. 1, 27 (1975).
14 j. A. Peterson and M. C. Weiss, Proc. Natl. Acad. Sci. U.S.A. 69, 571 (1972).
15 E. Engel, J. Empson, and H. Harris, Exp. Res. 68, 231 (1971).
~e M. Weiss and H. Green, Proc. Natl. Acad. Sci. U.S.A. 58, 1104 (1967).
17 D. Bergsma, ed., " H u m a n Gene Mapping III," Birth Defects: Orig. Art. Ser. XII(7).
Karger, Basel, 1976.
is T. Ege, J. Zerthen, and N. R. Ringertz, Somatic Cell Genet. 1, 65 (1975).
19 Reviewed in N. R. Ringertz and R. E. Savage, "Cell Hybrids." Academic Press, New
York, 1976.
20 E. Weed-Kastelein, W. Keijzer, and D. Bootsma, Nature (London), New Biol. 238, 80
(1972).
348
SPECIALIZED TECHNIQUES
[28]
[28]
CELL FUSION
349
350
SPECIALIZED TECHNIQUES
[28]
ecules produced by the antibody genes introduced into the hybrid by the
spleen cell nucleus, zs The culture medium from the hybrids contains microgram per milliliter amounts of the specific antibody. If the "hybrid
tumor" is injected into mice that have been pristane primed 2a to encourage ascites fluid formation, the tumor will grow and produce the antibody
in milligram per milliliter amounts in the ascites fluid.
Such hybrids will provide a constant supply of monoclonal antibody
against antigens of choice. These antibodies can be used as reagents for
any procedures for which antibodies were previously used, but with the
added advantages of higher levels of discriminatiori, lower background,
and a continuously available supply. The other obvious application of the
procedure is that large amounts of homogeneous antibody against specific
antigens will be available for sequence analysis; using current techniques,
the nucleic acid for these specific antibody genes can be isolated from the
hybrid cell lines and characterized, s It will soon be possible to compare
the genes and gene products of several cell lines that produce antibodies
against the same antigenic determinant. One could even choose lines
making such antibodies on the basis of those types of antibodies that
appear early in mouse neonatal development and those that appear later.31
The analysis of antibody and gene structure could have significant implications in regard to the genetic mechanism involved in the generation of
antibody diversity.
The above system may be but a prototype for others in which a chosen
cell line is hybridized with a specific cell type that produces a desired
product; the "immortalized" primary cell type thereby provides a continuous hybrid line producing enough of the chosen product to allow its
use as a reagent or to make it available for biochemical analysis. Systems
of this sort may be applicable to the production of cell lines expressing
specific immunological T cell receptors, a2 hormone receptors, or such
other cell surface molecules as differentiation antigens or tumor antigens.
It may also provide the means of producing significant quantities of hormones and other cellular products.
Some general principles that may be applicable to other possible systems can be derived from the work with hybrid plasmacytomas. It is
important that the cell line used has the capacity to produce the desired
28 D. Margulies, N. M. Keuhl, and M. D. Scharff, Cell 8, 405 (1976).
29 M. Potter, Physiol Rev. 52, 631 (1972).
a0 S. Tonegawa, C. Brack, N. Hozumi, and R. Schuller, Proc. Natl. Acad. Sci. U.S.A. 74,
3518 (1977).
31 N. Klinman and J. L. Press, Fed. Proc., Fed. Am. Soc. Exp. Biol. 34, 47 (1975).
az R. Goldsby, B. Osborne, E. Simpson, and L. A. Herzenberg, Nature (London) 267, 709
(1977).
[28]
CELL FUSION
351
Selection Procedures
352
SPECIALIZED TECHNIQUES
[28]
39
[28]
CELL FUSION
353
Another system of selection uses the drug ouabain 43as a specific inhibitor of the N a - K activated ATPase of the plasma membrane, the enzyme responsible for the active transport of K into the cell and the
extrusion of Na . Fusion of a cell that can be selected by HAT medium or
by its growth characteristics but that is not sensitive to a chosen concentration of ouabain, with another cell type that is sensitive to the same
concentration, allows selection of hybrid cells. Since human cells are
killed by low concentrations (10 -7 M) and mouse cell by higher concentrations (10 -a M) of ouabain this agent has become useful for selecting
mouse human hybrids.
One recently described selective medium, glucose-flee medium, results in selective growth of hepatoma cell lines producing gluconeogenic
enzymes.44 Variations of the medium select for hybrid clones reexpressing
one or more of these enzymes from hybrid lines in which liver-specific
functions were extinguished in the initial hybridization.
It should be clear that many selective systems are potentially available. One is limited only by the ability to derive cell lines that are differentially sensitive to metabolic inhibitors or differing in metabolic substrate
requirements.
Selection procedures are usually applied to hybrid cells 24-48 hr after
fusion to assure expression of the functions necessary for survival although it is not evident from our experience that this delay is really necessary when HAT medium is used. It is important in selective media such as
HAT, media in which cells must grow to be selected against, that the
fused cells should have enough surface area to divide 2-3 times after the
selective medium is added. Plating of the cells at too high a density will
slow the growth and increase the time that the cultures must be handled
before hybrid clones are isolated.
Fusion Protocols
Fusion with Sendai virus or polyethylene glycol can be done with cells
in mixed monolayers or with cells in suspension. In both cases it is essential that healthy cells with.high viability be used for fusion. When cells are
removed from a plastic or glass surface in order to fuse in suspension the
high viability should be established by phase microscopy or dye exclusion
after the cells are in suspension. During the washing and fusion procedures, care should be taken to avoid extremes in pH (maintain 7.2-7.6).
This is particularly important with Sendai virus fusions that involve long
43L. H. Thompsonand R. M. Baker, Methods Cell Biol. 6, 209 (1973).
44R. Bertolotti,Somatic Cell Genet. 3, 579 (1977).
354
SPECIALIZED TECHNIQUES
[9-8]
[28]
CELL FUSION
355
52
356
SPECIALIZED TECHNIQUES
[9-8]
[28]
CELL FUSION
357
fusion efficiency.6a With increased PEG concentration, the time of exposure will probably have to be decreased to avoid toxicity. It is our experience that the centrifugation step does increase the frequency although we
have fused several combinations of cells simply by exposing the loosened
pellet of cells to 1 ml of 50% PEG 1000 or PEG 6000, mixing the pellet
with PEG solution by gently rotating the tube, and, after 1-2 min of
exposure, adding 10 ml of medium without serum taking care that the
PEG is mixed and diluted with the medium.
The ratio of cells fused may vary from 1 : 1 for two cell lines to 5-10 : 1
when spleen cells or other primary cells that do not divide in culture are
fused with a cell line. In the cases in which 5-10 : 1 spleen cells : cell line
are used, it is clear that the unfused spleen cells act as a feeder layer and
increase the cloning efficiency of the hybrid cells. When we fused human
peripheral blood cells with mouse plasmacytoma cell lines the number of
hybrid clones derived was increased significantly by adding 1000 irradiated (4500 R) human diploid fibroblasts per microwell (Linbro FB
96TC) as a feeder layer.
The procedure below is used for fusion of plasmacytoma lines, cells
that are killed by HAT medium, with spleen cells from immunized
mice. 2~'65 We have also used it for making mouse x human hybrids with
several combinations of cell types. Spleen cells for the fusion are removed
3-4 days after the mouse is immunized. The spleen is placed in a 60-mm
petri dish in tissue culture medium with serum and is perfused with medium by injecting with a 26-gauge needle at several sites thereby forcing
medium into the spleen. This is continued until most of the cells are
removed; the number of spleen lymphocytes recovered is usually 5-10
107 per organ. Care must be taken not to repeatedly draw the suspended
cells up into the syringe and force them through the needle. Remove the
cells to a centrifuge tube, pellet them (1000 rpm, IEC MS), remove the
supernatant liquid, and suspend them in 5 ml of cold 0.17M NIL C1. Keep
the tube on ice for 10 min to lyse erythrocytes and then add 10 ml of cold
medium with 20% serum. Pellet the cells, count and check viability, and
mix with 107 of the plasmacytoma cell line.
1. Wash the cells in medium free of serum by centrifugation in a
round-bottomed tube (Falcon 2001).
2. Remove all the supernatant liquid by suction and loosen the pellet
by tapping the tube.
3. Add 0.2 ml of 30% PEG in medium without serum. The 30% PEG is
made by adding 3 ml of PEG 1000 at 41 to 7 ml of medium without serum
at 41 . PEG tends to lower the pH of the medium which should be ad65 R. H. Kennett, K. A. Denis, A. S. Tung, and N. R. Klinman, Current Topics
in Microbiol. and Immunol. 81, 77 (1978).
358
SPECIALIZED TECHNIQUES
[28]
justed to approximately 7.6 with NaOH. After mixing the PEG solution it
is kept at 37 until used.
4. The cells are maintained in the 30% PEG for 8 min. During the
8-rain period, cells are pelleted in the 30% PEG (1000 rpm, IEC MS,
radius 35 cm). The centrifugation time should be 3-6 min. It is our impression that the longer the centrifugation, the more hybrids. The limitation,
as indicated above, is the toxic effect on the particular cells used. This
may be monitored by phase microscopy or trypan blue exclusion. At the
end of 8 min the PEG is diluted with 5 ml of medium without serum and
then 5 ml of medium with 20% serum is added.
5. The cells, in diluted PEG, are pelleted and resuspended in 30 ml of
culture medium with serum. We have found the following medium optimal
for cloning and growth of hybrid plasmacytoma lines:
Dulbecco's MEM with high glucose (4.5 g/liter) to which are added
10% NCTC 109 medium,2S 20% fetal calf serum, 0.150 mg/ml oxaloacetate, 0.050 mg/ml pyruvate, 0.200 units/ml bovine insulin (Sigma),
and antibiotic of choice.
In addition to the purine and pyrimidine bases present in the NCTC 109
medium, thymidine (16 ~M) and hypoxanthine (0.1 mM) are added to the
medium in which the fused cells are plated.
6. The 30 ml of cells are evenly suspended and gently distributed into
6 microplates (Linbro FB96TC), 1 drop (about 50/zl) per well. The next
day, an additional drop of the above medium with aminopterin (0.8/zM,
twice Littlefield's concentration) is added to make HAT selective
medium.
7. The wells are fed two additional drops of medium 6-7 days later;
clones appear macroscopically within 2 weeks.
8. If necessary, the clones may be fed weekly by removing most of the
medium in the well and replacing it with fresh medium.
9. In most fusions, clones arise in nearly all the wells of the six microplates. The valuable wells are identified by screening the supernatant
liquid for production of the desired antibody, and these clones are transferred to larger wells (Linbro FB16-24-TC) with not more than 0.5 ml of
medium.
10. At early stages, the clones must be watched carefully so that they
are not allowed to grow to too high a density nor diluted too sparsely.
11. When the cells have been transferred into four of the larger
wells, they may be transferred to flasks, grown, and stocks prepared and
frozen.
12. The frequency of hybrids usually requires that the cells be cloned
in agarose over a feeder layer of human fibroblasts to be sure that the cells
[29]
359
are truly clonally derived. We have often cloned the cells from the original
well into two cloning plates successfully. -66
13. If the reversion rate of the parental lines is not known, control
cells should be put under selective conditions. For those cells selected
against a HAT medium it is wise to grow the parental cells in the appropriate selective drug, 5-bromodeoxyuridine or 8-azaguanine, before the fusion. The hybrid nature of the clones can of course be assessed by testing
for expression of parental enzymes, antigens, or chromosomes? 1
In addition to being used to fuse cells to form heterokaryons and
hybrids, the fusion methods with Sendai virus and PEG also have been
used recently to incorporate nuclei into enucleated cells, TM to transfer a
few chromosomes that are enclosed in microceUs into other cells, s7,ss and
to transfer strbstances enclosed in lipid vesicles e9 or erythrocyte ghosts TM
into cells. These techniques are likely to become important and useful
applications of membrane fusion techniques in the future.
Acknowledgments
The author is supported by NIH Grant CA 18930, NSF Grant PCM 76-82997, and the
Cystic Fibrosis Foundation. I thank Kathleen Denis for her discussions a n d work on hybridizations of mouse plasmacytomas with mouse neonatal cells and spleen fragments.
K. Sato, R. Slesinski, and J. Littlefield, Proc. Natl. Acad. Sci. U.S.A. 69, 1244 (1972).
67 R. E. K. Fournier and F. H. Ruddle, Proc. Natl. Acad. Sci. U.S.A. 74, 3937 (1977).
6s R. E. K. Fournier and F. H. Ruddle, Proc. Natl. Acad. Sci. U.S.A. 74, 319 (1977).
69 G. Poste, D. Papahadjopoulos, and W. J. Vail, Methods Cell Biol. 14, 34 (1976).
70 M. Furusawa, M. Yamaizumi, T. Nishimura, T. Uchida, and Y. Okoda, Methods Cell
Biol. 14, 73 (1976).
[29] F u s i o n o f H i g h e r P l a n t P r o t o p l a s t s
B y A L B E R T W . RUESINK
For many years the higher plant cell wall prevented any significant
research utilizing the fusion of higher plant cells, but the development of
techniques for removing the cell walls to leave behind li~,ing protoplasts 1
has recently provided exciting opportunities for research on genetics, development, and crop modification. In contrast to animal cells, plant cells
are often totipotent, and whole plants can be regenerated from individual
cells, from isolated protoplasts, or even from the product of the fusion of
two protoplasts, 2 which is called a somatic hybrid. Though the route to
1 A. W. Ruesink, this series, in press, (1979).
2 p. S. Carlson, H. H. Smith, and R. D. Dearing, Proc. Natl. Acad. Sci. U.S.A. 69, 2292
(1972).
360
SPECIALIZED TECHNIQUES
[29]
[29]
361
Three main methods for promoting protoplast fusion have been reported, but the most recent one is so superior to the earlier ones that the
first two will be mentioned only briefly.
The first successful method used NaNO3 as the fusing agent. 14 ProtolO A. W. Ruesink and K. V. Thimann, Proc. Natl. Acad. Sci. U.S.A. 54, 56 (1965).
11 j. M. Widholm, Stain Technol. 47, 189 (1972).
1~ G. Weber, F. Constabel, F. Williamson, L. Fowke, and O. L. Gamborg, Z. Pflanzenphysiol. 79, 459 (1976).
1~ R. K. Horine and A. W. Ruesink, Plant Physiol. 50, 438 (1972).
14 j. B. Power, S. E. Cummins, and E. C. Cocking, Nature (London) 225, 1016 (1970).
362
SPECIALIZED TECHNIQUES
[29]
[29]
363
3. Add 0.6 ml of PEG solution drop by drop. (Make the PEG solution
by dissolving 1 g of polyethylene glycol (mol wt 1500) in 2 ml of 0.1 M
glucose, 10 mM CaCI2, and 0.7 mM KH~PO,.) PEG sources: PEG 1540,
Polysciences, Inc., Warrington, Pennsylvania; MODO-PEG 1500: Modokemi, Stenungssund, Sweden.
4. Incubate at room temperature for 40 min. Occasional rocking of the
petri dish should prove helpful if the protoplasts are not touching each
other.
5. Gently add 0.5 ml of protoplast culture medium that should contain
about 5 mM Ca. A pH of 5.7 is usually used, but a higher pH, up to 10,
will enhance the number of protoplasts fusing.
6. After 10 min, add another 1.0 ml of medium.
7. Wash the preparation several times with 2-ml aliquots of culture
medium, using a Pasteur pipette to remove the old medium each time. Be
sure to leave a thin layer of medium over the protoplasts at all times.
8. Culture the protoplasts or study them microscopically while they
are still on the coverslip. Alternatively, gently wash them free from the
glass and embed them in soft agar (0.6%, procool/ed before adding protoplasts). TM An optimal arrangement is to embed the protoplasts in a 1-mm
thick layer on top of a previously solidified 5-mm layer.
Occasionally, protoplasts will fuse spontaneously as they are being
released from tissue, apparently by expansion of the plasmodesmata linking cells together in the tissue. 2 Such spontaneous fusions are rare and
are of little concern to workers interested in interspecific fusions. The
method described above will produce up to 35% interspecific hybrids in a
preparation. The rest are unfused cells and intraspecific fusion products.
Although fusion products resulting from the fusion of just two protoplasts
predominate, the products of triple, quadruple, and multiple fusions are
also present. Very large cells resulting from the fusion of many protoplasts
tend to be unstable and hence are typically present only in small numbers.
Both the molecular weight and the concentration of PEG are critical to
inducing successful fusions. Polyethylene glycols of less than 1000 mol wt
are not able to produce tight adhesion, and those of up to 6000 are more
and more active per mole in inducing fusions but produce too viscous a
solution for easy handling. The choice of 1500 is a good compromise. The
concentration used above (about 0.33M) provides maximal fusions, with
significantly less fusion at both lower and higher concentrationsY 1
A distinction must be made between aggregation, where two or more
protoplasts just stick to each other, and true fusion, where mixing of
lg I. Takebe, G. Labib, and G. Melchers, Naturwissenschaften 58, 318 (1971).
20 L. A. Withers and E. C. Cocking, J. Cell Sci. 11, 59 (1972).
zl K. N. Kao, F. Constable, M. R. Michayluk, and O. L. Gamborg,Planta 120, 215 (1974).
364
SPECIALIZED TECHNIQUES
[29]
Hybrid Fusions
M u c h of the interest in plant cellfusion resultsfrom its abilityto bring
together new combinations of genetic information. The following tabulation liststhe cases where hybrid fusions have occurred and indicates that
usually P E G was the fusion-inducing agent. Often, some cell divisions
subsequently occurred. In only two cases, however, has it been possible
to regenerate a hybrid plant.
Considerable attention has been devoted to means by which hybrid
fusions can be detected. One important way is the use of selectivegrowth
media as will be described below, but microscopic methods are also used.
Workers have looked for: nucleus size differences,3 nucleus staining
22 T. Kameya, Planta 115, 77 (1973).
2a j. Burgess and E. N. Fleming, Planta 118, 183 (1974).
24 K. Glimelius, A. Wallin, and T. Eriksson, Physiol. Plant 31, 225 (1974).
25 A. W. Ruesink, Plant Physiol. 47, 192 (1971).
26 B. W. W. Grout and R. H. A. Coutts, Plant Sci. Lett. 2, 397 (1974).
2r R. Kanai and G. E~ Edwards, Plant Physiol. 52, 484 (1973).
2s M. Ito, Plant Cell Physiol. 14, 865 (1973).
29 H. Binding, Z. Pflanzenphysiol. 72, ,122 (1974).
3o C. W. Jones, I. A. Mastrangelo, H. H. Smith, H. Z. Liu, and R. A. Meek, Science 193, 401
(1976).
Fusion method
NaNO3
NaNO3
Unreported
Unreported
NaNOa
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
& Ca
PEG
PEG
PEG
Hybrid plant
Some cell divisions
Fate
Ref.
a
b
Colonies up to 10cells
Some cell divisions
Some cell divisions
Some cell divisions
Nuclei cohabit cells
up to 6 days
Hybrid plant
Small colonies
Small colonies
Mixed cytoplasm;
intermediate cell
surface structure
J
k
t
366
SPECIALIZED TECHNIQUES
[29]
[29]
367
genome from each parent plant must be selected and maintained in the
culture. (4) An intact plant must be regenerated. Significant progress has
been made on all problems but number 3.
To select a desired fusion product, the genetic complementation of
auxotrophic mutants has been used with such systems as Neurospora 3s
and liverworts. 39 So far, it has not been successful with higher plant cell
fusions, apparently because of cross-feeding between cells that allows
parent cells to grow up. 2 Differential drug sensitivity of parent species has
recently shown promise as a selection device in a petunia system, 4 and
related work indicates that complementation, bringing green coloration
into albino tissue culture cells, may prove useful. 4~An alternative method
has been to use semilethal, recessive chlorophyll-deficient mutants that
complement in diploids. 42
To procure some cell divisions from protoplast fusion products is not a
major problem as can be seen from the table. However, the appropriate
culture conditions for maintainance of continued proliferation of the cells
have not been worked out. An equally difficult problem has been the
regeneration of whole plants from protoplasts. Whole plants can be regenerated from tobacco protoplasts more readily than from those of most
species, although there is no intrinsic reason against regeneration of plants
from the protoplasts of many other species. Only six plants have yielded
to efforts to obtain whole plants from protoplasts: tobacco, TM c a r r o t , 4a
petunia,44 asparagus,45Atropa, 46and rapeseed. 47 A number of laboratories
that want to perform genetic engineering work are now focusing their
primary attentions on the elucidation of basic information needed to convert tissue-cultured cells, particularly of crop plants, into differentiated
plants; these researchers realize that a much better understanding of that
preliminary process is essential before protoplast fusions can be used in
genetic engineering.
3a L. Ferenczy, F. Kevei, and M. Szegedi, Experientia 31, 50 (1975).
39 O. Schieder, Z. Pflanzenphysiol. 74, 357 (1974).
40 j. B. Power, S. F. Berry, E. M. Frearson, and E. C. Cocking,Plant Sci. Lett. 10, 1 (1977).
4~ E. C. Cocking, D. George, M. J. Price-Jones, and J. B. Power, Plant Sci. Lett. 10, 7
(1977).
42 G. Melchers and G. Labib, Mol. Gen. Genet. 135, 277 (1974).
43 H. J. Grambow, K. N. Kao, R. A. Miller, and O. L. Gamborg, Planta 103, 348 (1972).
44 E. M. Frearson, J. B. Power, and E. C. Cocking, Dev. Biol. 33, 130 (1973).
45 D. Bui-Dang-Ha and I. A. Mackenzie, Protoplasma 78, 215 (1973).
46 G. Gosch, Y. P. S. Bajaj, and J. Reinert, Protoplasma 86, 405 (1975).
4r K. K. Kartha, M. R. Michayluk, K. N. Kao, O. L. Gamborg, and F. Constabel,PlantSci.
Lett. 3, 265 (1974).
368
SPECIALIZED TECHNIQUES
[30]
[30] Cell T r a n s f o r m a t i o n
By IRA PASTAN
Cultured fibroblastic cells are frequently used in studies of "malignant
transformation"; the term "transformation" is also often used by itself.
"Malignant transformation" was coined to distinguish it from the "transformation" process by which DNA is introduced into bacteria. A "transformed" cell is one that has undergone a stable heriditable change that
enables it to grow into a tumor in an appropriate recipient animal. Since
the animal from which a cell is derived may not be available for testing for
tumorgenicity in vivo, other tests have been developed. Nude mice have
an immunological deficiency that interferes with their ability to reject
foreign cells, thereby allowing their use for tumorgenicity testing (see this
volume [31]).
Since it is cumbersome to test whether a cell has become transformed
by injecting it into an animal, other simpler tests are widely used. One that
strongly correlates with results of animal tests is the ability of transformed
cells to grow in the absence of substratum (agar; methylcellulose; suspension culture). 1 Another prominent metabolic feature of transformed cells
that has long been recognized is their rapid metabolism of glucose; 2 cells
develop this feature shortly after transformation)
Frequently, transformed cells have a different morphologic appearance than normal cells in culture. Normal fibroblasts are elongated and
flattened. Transformed cells often are more rounded. This change in
shape is accompanied by changes in the cell surface. When viewed by
scanning electron microscopy, cells are usually covered with many microvilli and lamelopodia; at times the surfaces are covered with blebs. 4
Another property of transformed fibroblasts is their increased ability to be
agglutinated by plant lectins) ,6 Several theories have been advanced to
explain their enhanced agglutinability. The simplest of these is that
agglutinability is enhanced because the microvilli and other irregularities
i V. H. Freedman and S. Shin, in "The Nude Mouse in Experimental and Clinical Research" (J. Fogh and B. Giovanella, eds.). Academic Press, New York, 1978 (in press).
2 B. Warburg, " T h e Metabolism of Tumors." R. N. Smith, New York, 1931.
a S. G. Martin, S. Venuta, M. Weber, and H. Rubin, Proc. Natl. Acad. Sci. U.S.A. 68,
2739-2741 (1971).
4 K. Porter, D. Prescott, and J. Frye, J. Cell Biol. 57, 815-836 (1973).
5 M. M. Burger, Biochemistry 62, 994 (1969).
6 B. Inbar, and L. Sachs, Proc. Natl. Acad. Sci. U.S.A. 63, 1418 (1969).
[30]
CELL TRArqSVORMATIOrq
369
on the transformed cells' surface provide large surfaces that are available
for the lectins to "glue" the cells together. 7
A very important factor contributing to the abnormal shape of transformed cells is their decreased adhesion to substratum.S Mutant cells have
been obtained that have the morphologic phenotype of transformed cells:
rounded shape, decreased adhesion to substratum, increase in surface
microvilli, and increased agglutinability by lectins. In such cells the primary defect is a decrease in cell adhesion2 Apparently decreased adhesion to substratum causes the cell to assume a rounded shape with microvilli or blebs on the cell surface. Although such adhesion-defective
mutants have the morphologic phenotype of transformed cells, they are
not, in fact, transformed since they neither form tumors in animals nor
grow in agar2 Thus, in the absence of other information, it is not safe to
use morphologic or adhesive features for establishing that the phenomenon of transformation has occurred.
Selection of Transformed Cell Lines for Study
Many transformed cell lines are available for study and can be conveniently obtained from the American Type Culture Collection or other
sources (see this volume [37]). Some of these lines have been in culture
for many generations and may have developed characteristics that are not
related to the original transformation event. Therefore, it is generally best
to use recently transformed cells in which the normal parent is available
for comparative purposes.
High and Low Transformation Systems
It is possible to transform almost all the cells in a culture into cancer
cells within a few days with RNA tumor viruses (high-frequency transformation). A widely used system is that of chick embryo fibroblasts
(CEF) transformed by various strains of Rous sarcoma virus (RSV).
Methods for preparing pure clones of RSV and obtaining RSVtransformed chick embryo cells are described in this Volume [32] and [33].
Similarly, murine sarcoma virus (MSV) when combined with a helper
virus murine leukemia virus (MuLV) will rapidly transform various mouse
and rat cell lines. 1 An advantage of the Rous sarcoma system is the
r M. Willingham, and I. Pastan, Proc. Natl. Acad. Sci. U.S.A. 72, 1263-1267 (1975).
8 K. K. Sanford, B. E. Barker, M. W. Woods, R. Parshad, and L. W. Law, J. Natl. Cancer
Inst. 39, 705-718 (1967).
9 j. Pouyssegur, M. Willing,ham, and I. Pastan, Proc. Natl. Acad. Sci. U.S.A. 74, 243-247
(1977).
10 j. W. Hartley and W. P. Rowe, Proc. Natl. Acad. Sci. U.S.A. SS, 780-786 (1966).
370
SPECIALIZED
TECHNIQUES
[31]
[31 ] U s e o f N u d e M i c e f o r T u m o r i g e n i c i t y T e s t i n g a n d M a s s
Propagation
By
SEUNG-IL
SHIN
[31]
371
372
SPECIALIZED TECHNIQUES
[31]
nude mice (for a review, see Freedman and ShinS). Some recent experiments suggest that nude mice contain elevated levels of certain nonTcell-mediated immunity, and that they are in fact capable of inhibiting the
growth of some tumorigenic cells. 9 It may thus become necessary to
employ additional immunosuppressive techniques on nude mice to assess
the tumorigenicity of selected cell types. However, for the general purpose of cellular tumorigenicity assay of animal cells in culture, the nude
mouse offers the best system presently known. The association between
cellular tumorigenicity in nude mice and various in vitro parameters of
cellular transformation has been reviewed recently. 8 The parameter best
correlated with tumor formation in vivo is the "anchorage-independent"
phenotype,-as measured by colony-forming efficiency in soft agar or
methylcellulose.
Source, Maintenance, and Breeding of Nude Mice
Due to their severe immune deficiency, homozygous mutant (nu/nu)
mice are highly susceptible to infectious diseases. During the early days
after the original discovery of this mutation, when the nature of the genetic defects was unidentified and therefore conventional animal husbandry techniques were used, nude mice rarely survived beyond the first few
weeks. However, if relatively simple precautions are taken it is now possible to maintain nude mice f o r a t least several months in most research
laboratory environments.
For intermittent short-term uses of nude mice, the most practical arrangement would be to obtain young adult nude mice from commercial
breeders. Once received, the animals should be strictly segregated from
other experimental animals, especially rodents; this is a most important
factor in maintaining good viability. Nude mice in different inbred backgrounds can now be purchased from commercial suppliers in the United
States and several other countries. Preferably, the mouse cages should be
placed inside a laminar-flow air filtration system. Conventional laminarflow hoods designed for cell culture can be used for this purpose. For
short-term experiments, the use of clean filterpaper covers that fit over
the cage tops has also proven to be adequate. Cages, bedding, cage tops,
and water bottles should be autoclaved before use if possible. Drinking
water should be boiled beforehand, but normal animal feed can be used
without further modification. Access to the animals should be limited to
8 V. H. Freedman and S. Shin, in " T h e Nude Mouse in Experimental and Clinical Research" (J. Fogh and B. Giovanella, eds.), p. 353. Academic Press, New York, 1978.
L. M. Reid, J. Holland, and C. Jones, submitted for publication.
[31]
373
374
SPECIALIZED TECHNIQUES
[31]
head behind the ears, and keeps the mouse stretched out. The person who
holds the syringe can then lift the fold of skin on the flank of the mouse
midway between the front and the hind legs, and use the other hand for
injection. It is neither necessary nor desirable to anesthetize the mouse
for this purpose. The cells should be deposited subcutaneously at a single
point, as far away as possible from the point of penetration in order to
prevent the cells from oozing out when the needle is withdrawn. Immediately after removal of the needle, hold the needle opening briefly between
two fingers. The fact that the cells are injected subcutaneously, and not
into the abdominal cavity or the rib cage, can be seen easily during the
injection process because the needle is visible throughout due to the translucency of the skin of the hairless mouse.
In order to increase the assay points per nude mouse, injections can be
made bilaterally. For the determination of.the threshold number of cells
required to initiate the tumor growth, etc., the number of cells injected
accordingly can be decreased or increased. Up to 108 cells have b e e n
injected at a single site.
Check the injected mice at least every 2 days thereafter, by both visual
inspection and manual palpation. The latency period between the injection of cells and the first appearance of a palpable nodule at the site of
injection varies greatly, depending upon the malignancy of the cells and
the inoculum used. 8 A typical tumorigenic cell line, such as H e L a or
mouse A9 cells, generates palpable nodules 1-2 weeks following injection
of 2 x l0 s viable cells. The minimum threshold number of cells required
to initiate tumor growth also varies widely. With the most tumorigenic
cells, such as the mouse melanoma cell line PG19, less than 10 cells can
initiate tumor growth, while near-diploid Chinese hamster cell lines such
FIG. 1. A phenotypically normal, heterozygous (+/nu) BALB/c mouse (left), and its hairless, thymus-deficient homozygous mutant nude littermate (right).
[31]
375
as CHO and WOR-6 form growing tumors only if 106 cells or more are
injected. Nontumorigenic cells, such as human diploid cell strains, will fail
to form tumors even with l0 s cells. A tumorigenic cell line should result in
the formation of a progressively growing tumor that, if left alone, would
eventually kill the host. Positive results are rarely ambiguous (see Fig. 1).
376
SPECIALIZED TECHNIQUES
[31]
TABLE I
REPRESENTATIVE
Cell line
TUMORIGENIC
CELL
LINES
DERIVED
FROM HUMAN
Tissue of origin
TISSUES
References
HeLa
D98/AH2
HS 0643
RPMI 2650
KE- I
SH- 1
PA- 1
NB20
3
3
3
14
1~
15
~5
S. Shin,
unpublished
WI-L2
A5/FG/HEK
LN-SV
14
16
17
TABLE II
REPRESENTATIVE TUMORIGENIC CELL LINES DERIVED FROM MOUSE TISSUES
Cell line
L929
A9
CI-I-D
Ehdich/Lettr~
RAG
B-16
Y-1
LLC
PCC4.azal
MPC 45-6
MOPC 315
SV101
Py3T3
KA31
EL4
References
3
3
5
3
3
15
18
15
S. Shin, unpublished
S. Shin, unpublished
S. Shin and H. Eisen,
unpublished
3
18
S. Shin, unpublished
[31]
377
TABLE III
TUMORIGENICCELL LINES FROM OTHER SPECIES
Cell line
Species
GH3
Rat
LG
Rat
C6
F17
Rat
Rat
SVRE-9
Rat
(M-MuSV)NRK
Rat
(KSV)NRK
Rat
CHO
Chinese
hamster
Chinese
hamster
WOR-6
PSN4
BHK 21
BHK-T6a
DMN6A
Syrian
hamster
Syrian
hamster
Syrian
hamster
Syrian
hamster
References
19
PySplA
Syrian
hamster
B(a)P104C1
Guinea pig
DENA HM2C1
Guinea pig
RKT-TG1
Rabbit
RSV-CE-Pr
Chick
S. Shin and
B. Nadal-Ginard,
unpublished
18
16
20
H. Klinger,
unpublished
3
P. Canary
and N. Bouck,
unpublished
P.. Canary
and N. Bouck,
unpublished
21
S. Shin and
R. Junghans,
unpublished
378
[31]
SPECIALIZED TECHNIQUES
TABLE IV
REPRESENTATIVE ESTABLISHED CELL LINES THAT ARE
NONTUMORIGENIC IN NUDE MICE
Cell line
Species
References
CV-1
MDCK
Balb/3T3
3T3 (Swiss)
SVR95
Monkey
Dog
Mouse
Mouse
Mouse
4
is
3
3
4
F1-SV
M22
Mouse
Mouse
NRK C13
BRL
F2408
Rat
Rat
Rat
SVRE 12
Rat
4
4
20
18
S. Shin,
unpublished
4
14 C. D. Stiles, W. Desmond, G. Sato, and M. H. Saier, Jr., Proc. Natl. Acad. Sci. U.S.A.
72, 4971 (1975).
15 B. C. Giovanella, J. S. Stehlin, and L. J. Williams, J. Natl. Cancerlnst. 52, 921 (1974).
16 L. B. Chen, P. H. Gallimore, and J. D. McDougall, Proc. Natl. Acad. Sci. U.S.A. 73, 3570
(1976).
17 C. M. Croce, D. Aden, and H. Koprowski, Proc. Natl. Acad. Sci. U.S.A. 72, 1397 (1975).
Is C. D. Stiles, W. Desmond, L. M. Chuman, G. Sato, and M. H. Saier, Jr., Cancer Res. 36,
1353 (1976).
lg S. Shin, A. L. Brown, and F. C. Bancroft, Endocrinology 103, 223 (1978).
20 j. A. Bilello, V. H. Freedman, and S. Shin, J. Natl. Cancer Inst. 58, 1691 (1977).
21 C. H. Evans and J. A. DiPaolo, Cancer Res. 36, 128 (1976).
32 C. D. Stiles and A. A. Kawahara, in " T h e Nude Mouse in Experimental and Clinical
Research" (J. Fogh and B. Giovanella, eds.), p. 385. Academic Press, New York, 1978.
[32]
379
subsequent operations. After spraying the area around the tumor with
95% ethanol, open the tumor with scissors and remove a section of tumor
about 5 5 x 5 mm from an internal area free of necrotic tissue. Place
the tumor tissue in a sterile petri dish and remove the mouse from the
hood. Rinse the tumor tissue 3 times with at least 5 ml of phosphatebuffered saline or culture medium, placing the tissue into a new petri dish
each time. Mince the tissue into very small fragments of 1 mm s or less and
remove all obvious fatty material. Many tumors can be reduced to mostly
single cells and cell clumps in this manner. Tumor tissues that are not
easily dissociated into single cells can at this point be passed through a
sterilized stainless-steel wire mesh to further disaggregate the cells by
rubbing the fragments against the mesh with blunt-ended scissors. Alternatively, stepwise, gentle trypsinization can be used to dissociate single
cells from tumor fragments. In any case, it is not necessary to obtain pure
single-cell suspensions in order to start a new culture from the tumor.
Suspend the cell or minced tissue preparation in 10-25 ml culture medium
containing antibiotics, and distribute the contents into a set of 60-mm
culture dishes. Change the medium after overnight incubation; most of
the unattached lymphocytes and cell debris can be washed off at this time.
Red cells, loosely attached to the dish, may last for many days, but will
not prevent the growth of the cells. Tumors originally produced by established cell lines almost always give rise to vigorous cultures in a few days.
[32] B i o l o g i c a l T e c h n i q u e s f o r A v i a n S a r c o m a V i r u s e s
By ERIC HUNTER
Rous sarcoma virus (RSV), an avian retrovirus, is capable of inducing
rapidly growing solid tumors of the connective tissue in fowl, and morphological transformation of cells in culture. This virus group is therefore
of particular interest as a model system for studies on viral oncogenesis.
Moreover, it has become clear that the avian sarcoma viruses provide an
excellent system for studies of the control of eukaryotic gene expression
and of the mechanism of integration of viral nucleic acids into a host cell
genome.
Experiments of this nature require methods for obtaining a biologically
and biochemicaUy homogenous virus population and quantitating the biological activity of such a population. This article describes a quantitative
and reproducible assay of RSV, the focus assay, l'z and methods for obR. A. Manaker and V. Group6 Virology 2, 838 (1956).
z H. M. Temin and H. Rubin, Virology 6, 669 (1958).
380
SPECIALIZED TECHNIQUES
[32]
taining cloned preparations of RSV, through both the focus and softagar-colony assays. Large-scale growth of RSV is considered in this volume [33].
Infection of chick embryo fibroblasts by RSV in vitro results in a
gradual change in the fibroblastic morphology of the infected cells to that
of a rounded, refractile transformed cell. Division of the initially transformed cell, in addition to infection and transformation of adjacent cells,
results in the formation of an island or "focus" of morphologically transformed cells within the uninfected monolayer of fibroblasts. A single particle of RSV is sufficient to produce a focus since the relationship between
virus qoncentration and focus count is strictly linear. The shape of the
transformed cell as well a s the morphology of the focus are largely controlled by the viral genome, 3 so that a focus induced b y the SchmidtRuppin strain of RSV is normally diffuse with interdispersed cells of
normal morphology while those induced by the Bryan high-titer strains of
RSV are generally compact and contain only transformed cells. To a
lesser extent focus characteristics are also influenced by the physiological
state of the cells in culture. For instance, cells that have been transferred
several times may tend to respond with the production of transformed
cells with a fusiform morphology.
Most strains of RSV are nondefective for replication; that is, infection
of a cell with a single virion results in both infectious-virus replication and
cell transformation. Viruses of the Schmidt-Ruppin strain of RSV, the
Prague strain of RSV, and the B77 strain of avian sarcoma virus (ASV) are
of this type. The most notable exception is the Bryan high-titer (BH)
strain of RSV, a replication-defective virus that requires a coinfecting
helper virus in order to produce infectious progeny. BH-RSV is a deletion
mutant, lacking most of the envelope glycoprotein gene. 4 It can be complemented by an accompanying helper virus that provides the missing
envelope glycoproteins that are necessary for infectivity. ~ BH-RSV
strains exist primarily, therefore, as "pseudotypes" in conjunction with a
nontransforming helper virus. Such a mixture may be unsuitable for some
biochemical experiments where a single homogenous population of virus
is required.
Viruses of the Schmidt-Ruppin and Prague strains of RSV and the B77
strain of ASV are most frequently used for biochemical studies, since they
can be readily cloned and yield good titers of virus. The initial isolates of
the Schmidt-Ruppin and Prague strains of RSV were mixtures of viruses
3 HI M. Temin, Virology 10, 182 (1960).
4 p. H. Duesberg, S. Kawai, L.-H. Wang, P. K. Vogt, H. M. Murphy, and H. Hanafusa,
Proc. Natl. Acad. Sci. U.S.A. 72, 1569 (1975).
5 H. Hanafusa, Natl. Cancer Inst., Monogr. 17, 543 (1964).
[32]
381
382
SPECIALIZED TECHNIQUES
[3 9-]
both chf and gs expression can be obtained from Spafas Inc. and H & N
Inc. For crucial experiments, quantification of chf expression, as described later in this article, is recommended.
Fertile eggs of other avian species generally can be obtained from local
vendors. Cells from the Japanese Quail and Peking Duck are particularly
useful in some experiments since they appear to lack any endogenous
viral sequences related to RSV.
2. Sera
a. Calf Serum. Not all serum lots offered commercially will support
focus formation by RSV in tissue culture, and samples from several different lots should be tested before larger quantities are purchased. Satisfactory sera have been obtained from Biocell Inc., Venice, California;
Flow Laboratories, Bethesda, Maryland; and Grand Island Biological
Company, Grand Island, New York. Newborn calf serum is usually not
required for cultivation of chick cells. In fact, bovine cadet serum, collected from animals up to 6 months of age, has proved highly satisfactory.
Calf sera are not inactivated. They may be stored for as much as 1 year at
- 2 0 .
b. Chicken Serum. Chicken serum also may be purchased from the
suppliers mentioned above. It should not contain antibody to RSV. Since
these sera are not usually obtained from leukosis-free flocks, they must be
inactivated at 56 for 2 hr prior to use in order to destroy any infectiousleukosis viruses that may be present. Storage up to I year at - 2 0 is
permissible.
3. Media and Reagents
a. Cell Culture Media. Several media are satisfactory both for the
growth of chick embryo cells and assay of RSV. Nutrient mixture F108 is
commonly used, but Minimal Essential Medium, 9 and Medium 1991may
be substituted. Single- and double-strength concentrations are required.
Commercially available solutions and powders are acceptable. The latter
are particularly suitable where large volumes of media are used, since
each batch can be tested in advance for its ability to support cell growth
and focus formation. Sterilization of tissue culture media is achieved by
pressure filtration through 0.22-/zm (average pore size) filters. Penicillin
s R. G. Ham, Exp. Cell Res. 29, 515 (1963).
g H. Eagle, Science 130, 432 (1959).
10 j. F. Morgan, H. J. Morton, and R. C. Parker, Proc. Soc. Exp. Biol. Med. 73, 1 0950).
[32]
383
384
SPECIALIZED
[32]
TECHNIQUES
TABLE I
COMPLEX MEDIA FOR PRIMARY AND SECONDARY CULTURES OF CHICK EMaRYO CELLS
Media
Nutrient mixture FI0, a 1 concentration
Nutrient mixture F10, a 2 concentration
Calf serum
Chicken serum
Tryptose phosphate broth
Sodium bicarbonate (2.8%)
Purified agar c
Bacto agar
Dimethyl sulfoxide
PGM
POL
SM
GM
OL
80 ~
-8
2
10
2.0
--
40
20
8
2
10
2.0
20
80
-1
-10
2.0
--
80
-5
-10
2.0
--
-40
5
-10
2.0
--
.
--
.
--
.
--
1.0 d
40
1.0
a Other culture media may be substituted for nutrient mixture F10; see text.
b E x p r e s s e d in p a r t s o f t o t a l v o l u m e .
c P u r i f i e d a n d B a c t o a g a r ( 1 . 8 % ) a r e m e l t e d in a b o i l i n g - w a t e r b a t h o r m i c r o w a v e
o v e n . B o t h a g a r a n d m e d i a c o m p o n e n t s a r e t h e n e q u i l i b r a t e d in a 45 . w a t e r b a t h
prior to mixing.
d G M is s u p p l e m e n t e d w i t h 1 % D M S O w h e n v i r u s is t o b e h a r v e s t e d f r o m t r a n s f o r m e d cells; s e e t e x t .
overlay medium for the RSV focus assay (OL) (Table I). The formulations
for the bottom agar (BA) and top agar (TA) media of the soft-agar-colony
assay and for cloning medium (CM) used in conjunction with this assay
are presented in Table II.
T A B L E II
COMPLEX MEDIA FOR SOFT-AGAR CLONING OF R S V
Media
BA
TA
CM
-35 ~
I0
3
15
1.0
1.0
-20
6
2
10
1.0
1.0
40
20
--
70
-10
3
15
1.0
1.0
--0.5
--
35
--
a E x p r e s s e d in p a r t s o f t o t a l v o l u m e .
b P G M h a r v e s t e d f r o m q u a i l p r i m a r y c u l t u r e p l a t e s 72 h r a f t e r s e e d i n g .
[32]
385
386
SPECIALIZED TECHNIQUES
[32]
Approximately 0.8--1.2 x 107 cells are required for tests of susceptibility to RSV infection and endogenous virus gene expression; the remainder
may be seeded in 100-mm petri dishes in l0 ml PGM (5-7 x l0 ~ ceils/
plate) for later use as secondary cultures. Plates containing PGM are
equilibrated in a 37-38 incubator, gassed with 95% air-5% CO2, for 1-2 hr
prior to seeding, and returned to it immediately after addition of the cells.
The primary cultures are overlayed with POL (12.5 ml/100-mm plate)
3 days after seeding. This stimulates the growth of the cells and maintains
them in a viable, healthy condition for up to 9 days after seeding.
The general procedure described above can be used to establish primary embryo cultures from most avian species, with only minor modifications to the method described here.
[32]
387
388
SPECIALIZED TECHNIQUES
[32]
pletely excluded from quail embryo cells, and thus provides a very sensitive system for the detection of RSV(RAV-0) pseudotypes.
For the test, a portion of the embryo cells (2 x 106/plate) are seeded
into a 35-mm petri dish containing 2 ml of PGM + polybrene (2/xg/ml-see below) and inoculated with approximately 105 focus-forming units of
RSV(RAV-7) shortly after seeding. On the second day after seeding, 1 ml
of the culture medium is removed and assayed on secondary cultures of
quail embryo cells for infectious virus as described in the following section. Control plates are inoculated with RSV(RAV-7) and appropriate
dilutions of a stock of RSV(RAV-0). Culture medium from embryos that
are chf~positive will generally induce 102 to l0 s foci in this quail focus
assay.
Quantitation of Transforming Virus: The Focus Assay
[32]
389
B. METHOD 2
The medium is removed from the primary cultures and the monolayers
washed with Tris-buffered saline as described above. Only 2.5 ml of
0.05% trypsin are added to a 100-mm plate, and this is left on the cells
until the monolayer takes on a granular appearance. The cells are suspended with a hand pipette and rubber bulb and are added to 3 ml of
transfer buffer (TB: 10% calf serum in Tris-buffered saline). After determining the cell concentration, the suspension is seeded into 60-mm tissue
culture dishes containing 3 ml SM, at 1.2 106 cells/plate. The cells
settle and spread rapidly in this medium, which is changed, 0.5-2 hr after
seeding, to 5 ml GM.
Each method has its advantages. The first generally avoids any possibility of overtrypsinizing the cell monolayer since excess trypsin is removed from the plates. It also avoids the obligatory medium change of
method 2 that can be excessively time consuming in large assays. In
general, assays of RSV set up by method 2 have somewhat cleaner background monolayers since unhealthy or dying cells are removed with the
SM.
390
SPECIALIZED TECHNIQUES
[32]
polybrene (Aldrich Chemical Co.) is the most efficient and least toxic to
cells. It may be added to the GM (2/~g/ml) on a routine basis unless
viruses of subgroup A are to be assayed, since infection by these viruses
is inhibited to a limited extent by polycations.
Focus assays on secondary cells of avian species other than chicken
are carded out in essentially the same manner. However, the OL added in
focus assays on quail embryo cells should always contain 1% chick serum
since this increases the efficiency of the assay severalfold.
3. Quantitation of Foci
The focus assay is usually counted 6--8 days after infection, although
foci may be visible from the fifth day. In order to quantitate the number of
foci a glass plate with a 2-mm2 grid (Technical Instrument Co., San Francisco, California) is attached to the slide holder on the mechanical stage of
an inverted microscope. The optimum total magnification for counting
foci is between 30 and 40 since this allows a 4 mm wide section of the
plate to be counted at one time. A 10 objective may be useful for
determining fine details of focus morphology. Cell clumps may resemble
foci, but generally can be distinguished from them by the presence of cells
radiating from the clump and by the absence of the rounded, refractile cell
type that characterizes RSV foci. Counting foci is usually facilitated if 1-2
ml of GM are added on top of the agar 2 hr prior to counting since this
appears to increase the refractile nature of the transformed cell and distinguishes it further from the background monolayer.
Cloning of R S V
The use of a homogenous virus population is crucial in many biochemical experiments. This is particularlytrue for R S V since in addition to the
natural variation that might be expected in a virus population, the growth
of nondefective sarcoma viruses in cellsis accompanied by the generation
of transformation-defective (td) mutants of RSV. 17,1sIn fact, continued
passage of a virus stock can result in a population that is predominantly
transformatiot~-defective virus. Therefore, it is important to produce
cloned virus stocks with which to perform experiments with RSV. T w o
methods are currently used to clone RSV: in the first,the virus is cloned in
the focus assay described above; in the second, the virus is cloned by
means of the soft-agar-colony assay.
17 p. K. Vogt, Virology 46, 939 (1971).
is S. Kawai and H. Hanafusa, Virology 49, 37 (1972).
[32]
391
392
SPECIALIZED TECHNIQUES
[32]
0.6% agar medium (BA) containing Japanese quail feeder cells and a top
layer of 0.36% agar medium (TA) into which infected cells are suspended.
Bacteriological dishes (60 mm) are used in preference to tissue culture
dishes in order to prevent adherence and overgrowth of the quail feeder
cells. A volume of 3.0 ml of bottom agar containing 5 x 105 quail cells is
added to each dish and allowed to harden at room temperature.
Chicken cells are infected in suspension by mixing 1-2 106 cells in 1
ml GM with virus at a multiplicity of infection of between 0.1 and 0.01
focus-forming units per cell. The mixture is incubated at room temperature for 1-2 hr, after which aliquots of dilutions of the infected cells are
added to 3 ml of TA in tubes at 45 . The agar suspension of cells is then
poured immediately onto the bottom agar medium. After the top agar has
hardened (20-30 min), the dishes are moved carefully to incubate at 37.
At day 4 and day 7, dishes are overlaid with 2 ml of OL medium. Colonies
of transformed cells are generally.visible to the naked eye by 7-12 days.
In some instances clumping of cells can occur during the 1-2 hr incubation of cells and virus. To ensure that only sifigle cells are suspended in
the top agar layer, the cell-virus mixture may be seeded into a 35-mm
plate and retrypsinized from the plate 4--6 hr later.
In order to establish transformed cell clones, colonies (about 1 mm in
diameter) are aspirated into a finely drawn-out Pasteur pipette and placed
into 16-mm wells of a 24-well cluster plate (Costar, Cambridge, Massachusetts) containing 1 ml of cloning medium. In general, no more than
12 colonies should be grown up in each 24-well plate in order to reduce the
possibility of cross-contamination. Plates are incubated at 37 and
checked every second day to determine the stage of growth. When the
cells are confluent they can be transferred to a 35-mm culture dish and
from there, via a 60-mm dish, to a 100-mm plate, from which cloned virus
can be harvested. It is important when transferring transformed cell
clones not to underseed the tissue culture dish; in general, seeding cells
onto twice the original area results in rapidly growing healthy cells. Once
established, transformed cell clones can be grown in GM containing 1%
DMSO.
Frozen Storage of Chick Embryo Fibroblasts
Chick embryo cells can be frozen and stored in a medium containing
high concentrations of serum and D M S O . The cells are trypsinizcd and
suspended in G M as described previously. After centrifugation(600-800
rpm, 5 min), the cells are resuspcnded in G M at a concentration of about
20 106 cells/ml. A n equal volume of freezing medium (11 parts G M , 5
parts calfserum, 4 parts dimethyl sulphoxide) is added drop by drop, and
[33]
GROWTH
OF
ROUS
SARCOMA
VIRUS
393
the cell suspension is added in 0.5-ml aliquots to 2-ml freezing vials (Vangard International, Neptune, New Jersey). The suspension is allowed to
freeze slowly (l/min is optimal) to - 7 0 before being transferred to a
liquid nitrogen freezer. The cells can be stored at - 7 0 , with reduced
survival.
Acknowledgments
Many of the techniques described in this article were developedin the laboratory of Dr.
P. K. Vogt, Universityof Southern California. The work of the author is supportedby Grant
VC-215 from the American Cancer Society.
[33] L a r g e - S c a l e G r o w t h o f R o u s S a r c o m a V i r u s
By RALPH E. SMITH
Biochemical investigations frequently require large quantities of cells
and virus. For example, the investigator may wish to isolate a specific
subcellular component that comprises a small fraction of the total material
present in the cell. In addition, biochemical investigations of viral structural components frequently require milligram quantities of purified virus.
Finally, the investigator may need to prepare large quantities of highly
infectious virus to take advantage of the unique ability of nondefective
Rous sarcoma virus (RSV) to rapidly and uniformly transform all cells in a
susceptible population. This article describes techniques for the growth of
transformed cells. It also outlines procedures for the preparation of
purified RSV. Certain of the biological techniques that are useful for working with the avian sarcoma viruses are presented separately in this volume
[32].
Media Reagents, Viruses, and Cells
394
SPECIALIZED TECHNIQUES
[33]
Viruses
Any of the sarcoma virus strains can be used for large-scale culture.
The most commonly used strains are the Schmidt-Ruppin strain of RSV,
the Prague strain of RSV, and the B77 strain of avian sarcoma virus.
Although most are used interchangeably, differences exist which merit
discussion. Both the Schmidt-Ruppin and Prague strains were originally
mixtures of viruses that have been cloned to reveal three viruses with
different antigenic and host-range properties. The Schmidt-Ruppin RSV
[33]
395
strain was cloned into members of the A, B, and D subgroups, while the
Prague RSV strain was purified into members of the A, B, and C
subgroups. 1
Members of the A subgroup are difficult to work with because extracellular virus does not accumulate reproducibly to a high level. The
reason for this is not clear, but seems to be related to the tendency of
viruses of subgroup A to accumulate at the cell surface in large aggre-.
gates, which are difficult to remove and purify. An occasional preparation
will show a high titer of virus, but this result is not reproducible. Viruses
of subgroups B and D are slightly cytopathogenic, and prolonged passage
of infected cells is usually difficult. In contrast, viruses of subgroup C (the
B77 avian sarcoma virus and the C subgroup of Prague RSV) are attractive, since they are not cytopathogenic and extracellular virus accumulates readily.
Cells
Chicken cells are used for most biochemical procedures. Methods for
the preparation of chicken embryo fibroblasts (CEF) will not be given,
since detailed descriptions are available 2 (see also this volume [32]). However, a brief discussion of the appropriate cell for study is useful. All
chicken cells possess an endogenous virus (RAV-0), which may be either
completely latent or expressed as infectious particles. For certain biochemical applications, it may be important to know whether the endogenous virus is being expressed in infected cells. Procedures have been published for determining the level of RAV-0 production. 3 Breeding programs
have been initiated to provide fertile eggs from flocks that are free of
RAV-0 expression. Such fertile eggs are available from commercial
sources (SPAFAS, Inc., Storrs, Connecticut; Heisdorf and Nelson, Redmon, Washington).
Fibroblasts that are free of the endogenous virus of the chicken may be
obtained from other avian species, but these cells may have endogenous
viruses of their own. Further, other avian species often have unusual
susceptibility patterns, and only certain subgroups of virus may be used
efficiently. The most common nonchicken avian species are the Japanese
quail and the Pekin duck. Both are readily available from commercial
breeders (Truslow Farms, Inc., Chestertown, Maryland) and can be purchased throughout the year. Other more exotic avian species, such as the
i j. Tooze, " T h e Molecular Biology of Tumor Viruses." Cold Spring Harbor Lab., Cold
Spring Harbor, New York, 1973.
z p. K. Vogt, in "Fundamental Techniques in Virology" (K. Habel and N. P. Salzman,
eds.), p. 198. Academic Press, New York, 1969.
3 See this volume [32].
396
SPECIALIZED TECHNIQUES
[33]
pheasant, are usually more difficult to obtain, and fertile eggs may be
available only once per year.
Mammalian cells can be transformed by avian sarcoma viruses, but
replication of the virus is blocked. Mammalian cells can therefore be used
to grow virus-transformed cells in the absence of virus replication. Infection of mammalian cells is an inefficient process, and large quantities of
highly infectious virus are usually required to obtain a few transformed
cells. Furthermore, one should keep in mind that the mammalian cells
should be frequently recloned, and cells selected for the particular attribute desired.
Preparation of Virus Stocks
The preparation of high-titer, well-characterized stocks of virus is a
key ingredient for the successful use of RSV in biochemical investigations. Stock virus preparation is usually carried out in two stages: cloning
of the virus stock and preparation of an intermediate stock.
Cloning. RSV cloning is started by performing a focus assay, in which
a monolayer (1.2 106 CEF in 5 ml GM/60-mm tissue culture dish) is
infected with 10-fold dilutions of RSV (dilutions are prepared in dilution
buffer). Infection with subgroups B, C, and D RSV are performed in the
presence of 2 /xg/ml polybrene. After 18-24 hr, the growth medium is
replaced with overlay medium, and the cells are incubated for an additional 7 days (an intermediate overlay of 3 ml of overlay medium at day 4
is usually helpful). Individual well-isolated loci are identified by visual
inspection and removed by drawing up a plug of agar directly over the
focus with a drawn-out sterile capillary pipette. The small amount of
material in the capillary pipette is transferred to 1 ml of dilution buffer.
After brief sonication in an ultrasonic bath, this material may be used to
start a second round of cloning, since sufficient virus is usually present to
obtain loci at a 10-1, 10-2, or 10-z dilution. Using this procedure, a virus
can be consecutively cloned 3 times in as many weeks. 4
Intermediate Stock Preparation. The relatively low virus titer of the
clone necessitates the preparation of a consistent, high-titer intermediate
virus stock for routine use. After investment of a large amount of time and
effort in the cloning of the seed virus, it is desirable to preserve the clone
in a pristine condition. This is accomplished by leaving the clone in its
milliliter of dilution buffer and entering the tube infrequently. To prepare
the intermediate stock, 0.1 ml of fluid from the isolated clone is put onto a
monolayer (1.2 106 CEF/60-mm dish) in the presence of 2 /zg/ml of
4 T. Graf, H. Bauer, H. Gelderblom, and D. Bolognesi,
[33]
397
polybrene, and cells are allowed to grow without overlay until all the cells
are transformed (one to two transfers). If other viruses are being grown at
the same time, it is a good practice to set aside a bottle of medium
specifically for the feeding of this stock in order to minimize the possibility of cross-virus contamination. The size of the-intermediate stock is
contingent upon the anticipated use and available storage space. One
should attempt to harvest sufficient virus to last for 1-2 years; otherwise,
the frequent removal of fluid from the original clone will necessitate recloning at an early date. Storage should always be at - 7 0 or lower.
Aliquots of the intermediate stock should be made in sufficiently small
volume that repeated freezing and thawing are not necessary, since the
titer of the virus will then drop below a useful level. An effective method
is to prepare several large containers of the virus stock (150 ml in a
screw-cap 200-ml polyallomer Sorvall centrifuge bottle works well), and
then thaw and dispense the virus into smaller samples (10 ml or less) as
the need arises.
Large-Scale Culture
Large amounts of virus are usually prepared by culture of virusproducing cells in roller culture bottles. Roller culture bottles are generally maintained at 37 , and since the bottles are sealed with screw caps,
humidification and carbon dioxide addition are unnecessary. Roller culture bottles are rotated at the rate of 6-10 revolutions per hour.
Preparation of Cells
There are two common methods for the preparation of virus-producing
cells in roller culture bottles. In method A, a small number of cells are
infected with virus in plastic tissue culture dishes, and these cells are
transferred until sufficient numbers are available to seed roller culture
bottles. The advantage of this method is that little intermediate stock
virus is used, and few cells are required for the initial seeding. Further,
the cells transform during passage in plastic dishes, and transformed cells
are easier to grow than nontransformed cells.
Specifically, method A involves the following procedure. Secondary
CEF are seeded into 150-mm plastic tissue culture dishes at 20 106
cells/dish and infected with 0.2 ml virus (undiluted) per dish. Cells are
subsequently trypsinized and replated into plastic tissue culture dishes at
20 x 106 cells/dish every 2-3 days until sufficient number of cells are
available for a roller culture bottle. In order to estimate the number of
cells required for a roller culture bottle, approximately 4 107 cells can
398
SPECIALIZED TECHNIQUES
[33]
[33]
399
prepare cells for one roller culture bottle. The embryos are decapitated,
minced, .and cells are dissociated with 0.25% trypsin in Tris-buffered
saline. A fluted trypsinization flask (Bellco) is helpful for this step. After
two consecutive trypsinizations (using 50 ml trypsin per embryo), the
cells are centrifuged at 1500 rpm for 15 min (800 g) in an International
refrigerated centrifuge, and the trypsin is decanted from the cell pellet.
The cell pellet is resuspended at the rate of 100 ml PGM per embryo, and
25 ml of a high-titer virus stock are added per embryo. Roller culture
bottles are seeded in 250 ml PGM per bottle. It is important to note that
polybrene cannot be used in this procedure, since it appears to change the
surface charge of the glass and prevents adherence of the cells. The primary cells will form a monolayer within 4-5 days, at.which time the roller
culture bottle is trypsinized (see method A) and cells are transferred to a
new roller culture bottle. Transformation is obtained within 4-5 days.
A disadvantage of method B is the requirement for large quantities of
intermediate virus stocks. It is impractical to infect a roller culture bottle
with less than 50 ml of virus-containing supernatant fluid, because transformation and high-titer production are considerably delayed when small
amounts of virus are used. The reason for the delayed appearance of
transformation probably stems from the large number of chicken cell
fragments and nonviable cells present in primary embryo fibroblast preparations that may absorb virus and prevent infection of the minority of
cells that eventually emerge to form the monolayer. Another disadvantage
of method B is that the cell monolayer is usually less. uniform than
monolayers prepared as outlined in method A. However, the rough appearance of the cell sheet is rapidly eliminated once trypsinization and
transfer of the cells in the roller culture bottles are initiated. Cells in roller
culture bottles are harvested, trypsinized, and transferred in the same
manner, whether prepared by method A or B.
400
SPECIALIZED TECHNIQUES
[33]
months. Prolonged harvest periods are only possible with RSVtransformed cells, since they will grow back into areas of the roller culture
bottle that have been exposed due to overgrowth of the cell monolayer
and loss of the cell sheet. The investigator determines when to stop harvesting by monitoring the virus yield. A good rule of thumb is that a liter
of supernatant fluid should yield 1 mg of purified virus. When production
falls below this level, it is hard to justify the time and expense required for
continued harvest.
When the investigator is interested in harvesting virus for the isolation
of 70 S RNA, daily harvesting is adequate. The duration of harvest is
usually not as long, and 1 month of production is generally all that should
be attempted. When the investigator is interested in harvesting virus for
the isolation of 35 S RNA, the roller culture bottles should be harvested at
intervals of 2 hr, but can usually be harvested for only 3 weeks. An
automated system is available for this type of harvest, and this subject
will be covered in the next section.
Virus-producing cells can be harvested at any time during the preparation or harvest of large-scale cultures, as long as the cells have not begun
to decline due to prolonged harvest. Cells in roller culture bottles are
washed with 50 ml of Tris-buffered saline and detached from the surface
of the roller culture bottle with the aid of a rubber-bladed scraper (Bellco).
The cells are rinsed into a centrifuge bottle with the aid of 200 ml of
Tris-buffered saline and centrifuged at 800 g for 15 min in a refrigerated
centrifuge. The cell pellet is then processed further for experimental purposes, or stored frozen at - 7 0 .
[33]
401
402
SPECIALIZED TECHNIQUES
[33]
virus. The most convenient method is to pellet the virus with ultracentrifugational forces. This is done in three stages. The first stage is to
remove cells and large debris by centrifugation at 800 g for 15 min. The
second stage is to centrifuge the supernatant fluid at 8000 g for 15 min to
remove subcellular debris and improve the subsequent purification steps.
The third stage is to pellet the virus. Three fluid-volume capacities are
used. For up to 210 ml, a Beckman SW27 rotor is used, and centrifuged 30
min at 24,000 rpm (80,000 g). For up to 420 ml, a Beckman 35 rotor is
used, and centrifuged 45 min at 24,000 rpm (46,000 g). For up to 1200 ml, a
Beckman 19 rotor is used, and centrifuged 105 min at 18,000 rpm (21,000
g). For even larger fluid-volume capacities, zonal rotors are available that
process several liters per hour.
The virus pellet is resuspended (at a volume ~ to z-~ of the original) in
a buffer appropriate for the use of the virus. For most applications, TE (1
mM EDTA, 5 mM Tris HC1, pH 8.6) is a suitable buffer. Two special
cases deserve mention. The first is that the virus pellet can usually be
extracted for 70 S RNA, in which case it is appropriate to resuspend the
virus in STE (0.1 M NaC1, 1 mM EDTA, 10 mM Tris, pH 7.4). The second
is that virus pellets may be used to make complementary DNA, in which
case the investigator resuspends the virus pellet in a buffer appropriate for
DNA synthesis.
Purification
Highly purified virus is obtained in a two-step process: equilibrium
(isopycnic) density gradient sedimentation, followed by velocity (rate
zonal) density gradient sedimentation. The concentrated virus pellet is
resuspended thoroughly (a brief sonication in an ultrasonic cleaning bath
is helpful) and layered onto a 15-60% (w/v) sucrose (in TE) gradient. The
Beckman SW27 rotor is used most frequently in our laboratory for this
application. The virus suspension is layered onto the sucrose gradients,
and the rotor is centrifuged at 24,000 rpm (80,000 g) for 3-6 hr, at which
time the virus is visualized near the center of the gradient. The virus band
is removed with a Pasteur pipette, diluted 4-fold with TE, and the virus is
pelleted at 24,000 rpm (80,000 g) for 35 min in a Beckman SW27 rotor. The
virus pellet is resuspended in TE, layered onto a 20-40% sucrose gradient, and centrifuged 90 min at 24,000 rpm (80,000 g) in a Beckman SW27
rotor, or 40 min at 36,000 rpm (175,000 g) in a Beckman SW41 rotor. The
virus band is diluted with TE, and the virus is pelleted as before. This
virus can be stored at - 2 0 indefinitely. Virus concentration can be mea-
[33]
403
Acknowledgments
The author wishes to express his thanks to Jann Bodie, Christine Chastain, Kay Izard,
and Sue Nebes, each of whom have substantially contributed to the development of the
procedures outlined in this review. Work performed in the author's laboratory was supported by National Institutes of Health Grant ROl-CA12323.
404
SPECIALIZED TECHNIQUES
[34]
[34] P r e p a r a t i o n o f S i m i a n V i r u s 4 0 a n d Its D N A
by GEORGE KHOURY a n d C H I N G - J U H L A I
[34]
405
studies. In addition, SV40 DNA has been used to study the physical and
chemical properties of supercoiled molecules, to assay restriction enzymes, and to evaluate DNA-protein interactions. The recent determination of the complete nucleotide sequence for SV40 a,4 will certainly enhance its value for numerous future biological and biochemical investigations. Simple methods for the production of SV40 and SV40 DNA are
described below.
A. Growth of SV40
Cell Cultures
SV40 produces a lytic infection in monolayer cultures of African green
monkey kidney cells. Several continuous cell lines are available including
BSC-1,5 CV-1,6 and Vero 7 as well as derivative clones of these lines. Titers
between 10s and 10a plaque-forming units (PFU) per milliliter of cell lysate
usually can be obtained. Primary or secondary AGMK cells apparently
yield 5-10 times this amount of virus. However, primary cells are expensive and tedious to prepare, which makes them a less satisfactory choice
for most laboratories.
Stock Virus
A number of strains of SV40 are commonly used including 776 (SVS),
777, and Va 4554. Many strains differ slightly in their DNA sequence and,
in some cases, this difference is reflected in variations in the restriction
enzyme cleavage patterns or in the morphology of the plaques that the
virus induces.8 Stocks of virus are generally prepared by infecting cells (in
Eagle's minimal essential medium, for example, supplemented with antibiotics, 0.03% glutamine, and 2% fetal bovine serum; abbreviated,
MEM-2) with less than 0.01 PFU/cell of a seed virus preparation. The low
multiplicity avoids the generation of defective virions that have been
a V. B. Reddy, B. Thimmappaya, R. Dhar, K. N. Subramanian, B. S. Zain, J. Pan, C. L.
Celma, and S. M. Weissman, Science (in press).
4 W. Fiers, R. Contreras, G. Haegeman, R. Rogiers, A. Van de Voorde, H. Van
Heuverswyn, J. Van Herreweghe, G. Volckaert, and M. Ysekaert, Nature (London) (in
press).
5 H. E. Hopps, B. C. Bernheim, A. Nisalak, J. H. Tjio, and J. E. Smadel, J. lmmunol. 91,
416 (1963).
6 F. C. Jensen, A. J. Girardi, R. V. Gilden, and H. Koprowski, Proc. Natl. Acad. Sci.
U.S.A. 52, 53 (1964).
r E. Earley, P. H. Peralta, and K. M. Johnson, Proc. Soc. Exp. Biol. Med. 125, 741 (1967).
8 T. J. Kelly and D. Nathans, Adv. Virus Res. 21, 85 (1977).
406
SPECIALIZED TECHNIQUES
[34]
[34]
407
408
SPECIALIZED TECHNIQUES
[34]
[34]
409
410
SPECIALIZED TECHNIQUES
[34]'
nitrocellulose tube. This tube and additional tubes are filled to capacity
with the medium saved from the infected cells; centrifugation is carried
out at 24,000 rpm for 3 hr (or 12,000 rpm overnight) at 4 in an SW27 rotor.
The virions, which under these conditions enter about 1 cm into the CsC1
cushion, can be visualized best with a high-intensity lamp in a dark room
as a blue translucent band. The band is removed in 1-2 ml of CsCI,
preferably by dripping through the bottom of a punctured tube. The density of this virus-containing CsC1 should be 1.33-1.34 g/ml (n = 1.3660),
and this material can be diluted directly into 7-8 ml of CsC1 of the same
density (1.34 g/ml) in a nitrocellulose tube for either an SW40 or a 50Ti
rotor. The virus is centrifuged to equilibrium, e.g., at 35,000 rpm for 16 hr
at 4 in a 50Ti rotor. After visualization and removal of the SV40 band
(which should be near the center of the tubes; 19 = 1.34), the virus is
dialyzed for 24 hr against the Tris-NaC1 buffer at 4 with 3-4 changes of
the dialysate. The protein capsids can be removed from virions by incubation at 50 for 30 min in 1% Sarkosyl. After two extractions with phenol
that is saturated with Tris-NaC1 buffer, the viral DNA, which remains in
the aqueous phase, can be either dialyzed at room temperature or precipitated in 2.5 volumes of cold ethanol prior to use. A further purification of
supercoiled SV40 molecules from relaxed molecules can be obtained as
described in the previous purification method by dye-bouyant-density
centrifugation and/or sucrose gradient sedimentation. However, when
SV40 DNA is extracted from virions these steps are frequently
unnecessary.
C. Radiolabeling and Quantitation of SV40 DNA
[34]
411
that contains approximately 100 /zCi/ml of carder-free [32p]orthophosphate. The time for harvesting radiolabeled viral DNA depends on
the method to be used for purification (see above):
In vitro Labeling of SV40 DNA
A number of laboratories now are routinely labeling DNA preparations in vitro using a method referred to as nick-translation. There are at
least two recently published procedures that describe in detail the methodology involved. ~2'13 Both procedures require DNase I and DNA
polymerase I and involve the nicking and removal of nucleotides from the
unlabeled input DNA followed by repair synthesis with radiolabeled nucleoside triphosphates. The advantages of in vitro labeling compared with
in vivo labeling include the ability to obtain radiolabeled DNA within
hours by a relatively simple procedure and the production of DNA with
extremely high specific activities, e.g., l0 s cpm//.~g using deoxynucleoside
triphosphates, two of which are o32p-labeled. Lower specific activities
also can be achieved by limiting the DNase concentration, the specific
activity of the radiolabeled precursors, or the time of incubation.
In vitro labeled SV40 DNA recently has been used to obtain qualitative
and quantitative information relating to the integrated viral genomes in
transformed cells. These high-specific-activity probes have been successfully used for reassociation kinetic analyses la and for detection of viral
DNA transferred to nitrocellulose filters t4 by the Southern blotting procedure.15
Quantitation and Assay of SV40 D N A
The yield of SV40 DNA is most easily determined spectrophotometrically. The concentration of a sample in milligrams per milliliter is approximately equal to the optical density at 260 nm divided by 20. Small
amounts of viral D N A are quantitated more accurately by reassociation
kinetics 16 or by gel electrophoresis. All three forms of SV40 DNA, i.e.,
form I (supercoiled molecules), form II (relaxed circles), and form III
(linear duplexes), are easily separated by agarose gel electrophoresis. In
order to quantitate small amounts of SV40 DNA (approximately 0.05-1.0
/zg), a DNA sample of unknown concentration is electrophoresed in parallel with serial dilutions of a DNA preparation of known concentration.
12 T. Maniatis, S. G. Kee, A. Efstratiadis, and F. C. Kafatos, Cell 8, 163 (1976).
~3 p. W. J. Rigby, M. Dieckmann, C. Rhodes, and P. Berg, J. Mol. Biol. 113, 237 (1977).
14 M. Botchan, W. Topp, and J. Sambrook, Cell 9, 269 (1976).
15 E. Southern, J. Mol. Biol. 98, 503 (1975).
~6 L. D. Gelb, D. E. Kohn, and M. A. Martin, J. Mol. Biol. 57, 129 (1971).
412
SPECIALIZED TECHNIQUES
[35]
The gel is stained with ethidium bromide solution (1 /~g/ml in the electrophoresis buffer) for 15-30 min and visualized under UV light. By comparing the intensity of staining of the unknown sample with a similar
standard, one can rather accurately assess the DNA concentration.
Perhaps the best criterion of purity for a viral DNA preparation is its
restriction enzyme cleavage pattern. Restriction cleavage maps of the
SV40 genome are known for numerous restriction enzymes 17 (also see
Kelly and Nathansa). In a typical assay, a small quantity of DNA is
cleaved with a restriction endonuclease, and the product is subjected to
agarose or polyacrylamide gel electrophoresis. If the DNA is 32p-labeled,
the ge! can be examined by autoradiography; otherwise, the restriction
pattern of the cleaved DNA can be analyzed by ethidium bromide staining
as described above.
A number of experiments have recently been carried out to directly
test the biological activity of either native SV40 DNA or segments of the
viral genome. In the first approach, permissive cells can be effectively
inoculated with the viral DNA in the presence of DEAE-dextran.18 This
technique has made it possible to assay the relative infectivity of various
viral DNA preparations. In addition, the technique has enabled investigators to propagate altered forms of viral genomes, either naturally occurring molecules or those constructed in vitro. An alternative procedure
for introducing viral DNA into nonpermissive cells involves coprecipitation of the DNA with calcium phosphate~a; this method had proven more
reliable, for obtaining morphologically transformed cells, and it has been
employed successfully to determine the limits of the viral DNA regions
required for establishment and maintenance of cell transformations. Finally, direct microinjection of viral DNA into cells has been valuable for
studying several biological interactions between viruses and cells. 2 While
this technique is limited by the number of cells that can be microinjected,
the ability to inoculate efficiently large numbers of molecules into single
cells has led to a number of important observations.
~7 K. J. D a n n a and D. N a t h a n s , Proc. Natl. Acad. Sci. U.S.A. 69, 3097 (1969).
18 j. M c C u t c h e o n and J. M. Pagano, J. Natl. Cancer Inst. 41, 351 (1968).
19 F. L. G r a h a m and A. J. v a n der Eb, Virology 52, 456 (1973).
2o A. G r a e s s m a n , Exp. Cell Res. 60, 273 (1970).
[35] P u r i f i c a t i o n a n d A s s a y o f M u r i n e L e u k e m i a V i r u s e s
By CHARLES J. SHERR and GEORGE J. TODARO
Murine type C viruses have been classified into two major groups
based on their potential for producing neoplastic disease. The mouse
METHODS IN ENZYMOLOGY, VOL. LVIII
[35]
413
414
SPECIALIZED TECHNIQUES
[35]
be propagated to high titer in the SIRC rabbit corneal cell line (CCL 60,
American Type Culture Collection [ATCC], Rockville, Maryland), in
MvlLu mink lung cells (ATCC line CCL 64), or in Cf2Th canine thymus
cells (Naval Biomedical Research Laboratories, Oakland, California). Viably frozen virus seed stocks can be obtained from the ATCC, from Pfizer
Laboratories (Maywood, New Jersey), from ElectroNucleonics, Inc.
(Bethesda, Maryland), or from the Office of Resources and Logistics of
the National Cancer Institute (Bethesda, Maryland). Seed cultures of
cells infected with virus also can be obtained frona numerous investigators
working in the field.
Virus Purification
Infected cells are grown and seeded into plastic 490-cmz roller
bottles (0.5 rpm) each containing 50--70 ml of medium. After cells attach,
[35]
415
the roller belt speed is increased to 1.0-1.5 rpm. Cells should be maintained at 60--100% confluency to insure high levels of virus production,
but should not be allowed to remain at confluency for long periods as virus
production will diminish, and cells will tend to slough from the surfaces of
the bottles. Medium containing virus is sterilely decanted at 24- to 48-hr
intervals and is rapidly cooled and kept at 4 . If maximum infectivity is
desired, roller bottles should contain only 20--40 ml of medium and should
be "rapidly harvested" at 3- to 6-hr intervals. In general, 10TM viral particles (approximately 1-2 mg of protein) can be obtained from approximately 5 liters of medium.
All further steps are performed at 4 . Virus-containing fluids are
clarified of cells and debris by centrifugation at 12,000 g for 10 min at 4.
The supernatant fluid is then centrifuged over a cushion of 20% glycerol
containing 0.05 M "Iris HC1, pH 7.8, and 0.1M KC1 (105,000 g for 90 min)
to pellet the virus. If the supernatant fluid is large, virus can be concentrated by ultrafiltration prior to pelleting (30 psi, XM 300 membrane;
Amicon Corp., Lexington, Massachusetts). Virus pellets are resuspended
in 0.05M Tris HC1, pH 7.8, containing 10 mM KC1 and 0.1 mM EDTA
using gentle passage through a #18 gauge syringe needle. The suspension
is then loaded over 20-60% (w/w) linear sucrose (ribonuclease-flee) gradients and banded isopycnically at a density of 1.16 g/cc. Visible gradient
bands can be harvested from the top of the tube using a Pasteur pipette, or
gradients can be collected by needle puncture from the bottom, and the
density of the band determined by refractometry. If the latter technique is
employed, aliquots of each tube can be assayed for viral reverse transcriptase activity (as described below) to confirm the position of the viral
band. Pooled, isopycnically banded virus is then diluted in the same buffer
used for banding and is repelleted over a glycerol cushion as described
above. For studies of viral structural proteins, viral pellets can be directly
frozen at - 7 0 or can be suspended in 0.05 M Tris HC1, pH 7.8, containing 0.1 mM EDTA prior to freezing. For maximum viral infectivity, preparation of viral RNA, or for synthesis of complementary DNA transcripts,
suspended virus particles should not be frozen but should be stored ,at 4
and used as soon as possible.
With very large volumes of virus-containing medium, continuous-flow
centrifugation is the most efficient method for purifying viral particles.
Virus fluids are clarified of cells and debris by a single pass through a
Beckman J-CF continuous-flow rotor (15 liters/hr at 12,000 rpm; calculated to retain particles with a sedimentation coefficient of -> 10,000 S).
Clarified virus fluids (20 liters maximum volume) are then passed through
a CF-32 rotor at 5 liters/hr, 32,000 rpm, and sedimented into a 20-60%
(w/w) linear sucrose gradient. Particulate and protein bands are allowed
to equilibrate for an additional 0.5 hr after cessation of virus fluid flow.
416
SPECIALIZED TECHNIQUES
[35]
The rotor contents are unloaded at 2000 rpm using a dense " c h a s e "
solution (62% sucrose) and are fractionated on the basis of optical density
(254 nm) and buoyant density (1.120-1.180 g/cc). Virus-rich fractions
(concentrated 500-1000 times at p -1.16 g/cc) are normalized to ---15%
sucrose by dilution with 0.05 M Tris HCI, pH 7.8, and centrifuged in a
Beckman type 35 rotor at 35,000 rpm for 1.5 hr (143,000 g). The virus
pellets are then suspended and stored as described above.
[35]
417
The genomes of murine type C viruses code only for a limited number
of viral proteins. These include the reverse transcriptase, the major envelope glycoprotein (gp70), and at least four additional low-molecularweight polypeptides (p30, p15, ppl2, and pl0). By convention, the virion
11 A. J. Dalton, J. L. Melnick, H. Bauer, G. Beaudreau, P. Bentvelzen, D. Bolognesi, R.
Gallo, A. Graft], F. Haguenau, W. Heston, R. Huebner, G. J. Todaro, and U. I. Heine,
lntervirology 4, 201 (1974).
12 R. Callahan, C. J. Sherr, and G. J. Todaro, Virology 80, 401 (1977).
13 E. M. Scolnick, E. Rand, S. A. Aaronson, and G. J. Todaro, Proc. Natl. Acad. Sci.
U.S.A. 67, 1789 (1970).
14 M. Green and G. F. Gerhard, Prog. Nucleic Acid Res. 14, 187 (1974).
15 W. P. Parks, E. M. Scolnick, J. Ross, G. J. Todaro, and S. A. Aaronson, J. Virol. 9, 110
(1972).
418
SPECIALIZED TECHNIQUES
[35]
structural proteins are designated " p " for protein, "gp" for glycoprotein,
and " p p " for phosphoprotein, followed by their apparent molecular
weight in thousands of daltons. 16Although standard immunological techniques such as immunofluorescence, immunodiffusion, and complement
fixation can be employed to assay for type C viral structural proteins, the
ability to purify viral proteins to homogeneity, label them to high specific
activity with 1251, and generate antisera that react specifically with them
has led to the development of extremely sensitive radioimmunoassays
(RIAs) for each of the above virion components.
Viral antigenic determinants can be assigned operationally to one of
three major classes of immunological reactivities, although in actuality
there exists a broad spectrum of different antigenic reactivities for each of
the viral proteins. Antigenic determinants that are shared in common by
type C viruses isolated from different mammalian species are designated
"interspecies-specific," while antigenic determinants shared in common
among a group of isolates from a single species (e.g., all MuLVs from M.
musculus) are classified as "species-specific" or "group-specific" determinants. In addition, "type-specific" antigenic determinants are those
that distinguish different isolates within a group (e.g., xenotropic and
ecotropic MuLVs).16
Four virion proteins (reverse transcriptase, p30, pl0, and gp70) possess predominantly group-specific and interspecies-specific reactivities
while p15 and ppl2 appear to exhibit predominantly type-specific reactivities. For general assays of MuLVs isolated from laboratory mice, p30
and gp70 are the most useful proteins for radioimmunoassay studies since
they are relatively easy to purify, are present in higher concentrations
than other proteins in virions, and contain readily detectable group- and
interspecies-specific antigenic determinants. Techniques for the purification of antigenically reactive MuLV p30,17"1sgp70,19 p15, p12, 2 and pl0 ~1
structural proteins as well as MuLV reverse transcriptase 22 have been
described in detail. Each of these proteins can be labeled with 125Ito high
specific activity (5/xCi//xg) using the chloramine T method~ and can be
used as "tracer antigens" in competitive RIAs that can detect subnanogram quantities of competing viral protein.
le j. T. August, D. P. Bolognesi, E. Fleissner, R. V. Gilden, and R. C. Nowinski, Virology
60, 595 (1974).
17 S. Oroszlan, C. L. Fisher, T. B. Stanley, and R. V. Gilden, J. Gen. Virol. 8, 1 (1970).
is E. M. Scolnick, W. P. Parks, and D. M. Livingston, J. Immunol. 109, 570 (1972).
is M. Strand and J. T. August, J. Biol. Chem. 248, 5627 (1973).
s o R. C. Nowinski, E. Fleissner, N. H. Sarkar, and T. Aoki, J. Virol. 9, 359 (1972).
21 M. Barbacid, J. R. Stephenson, and S. A. Aaronson, J. Biol. Chem. 251, 4859 (1976).
22 j. M. Krakower, M. Barbacid, and S. A. Aaronson, J. Virol. 22, 331 (1977).
23 F. C. Greenwood, W. M. Hunter, and J. S. Glover, Biochem. J. 39, 114 (1963).
[35]
419
Reagents
1. RIA buffer: 0.01 M potassium phosphate, pH 7.5, containing 0.1 M
NaC1, 0.01 M ethylenediaminetetraacetate (EDTA), and 0.5% normal
rabbit serum.
2. 125I-labeled p30 protein from MuLV (Rauscher strain or some hightiter equivalent). Specific activity should be approximately 5/~Ci//zg.
3. Rabbit antiserum to p30 protein of FeLV.
4. Goat antiserum to rabbit 7 S globulin ("second antibody").
420
SPECIALIZED TECHNIQUES
[35]
[35]
421
Remarks. All assays should include a standard competition curve generated using known concentrations of purified, unlabeled viral p30 protein
as the competing antigen. This permits quantitative estimation of the
amount of p30 antigen in any heterogeneous competing protein mixture, if
the total quantity of competing protein at each dilution is known. 25 Technical problems in RIAs for type C viral proteins are similar to those
encountered using general RIA techniques and arereviewed elsewhere. 26
Antisera to viral proteins can be prepared by immunization with
detergent-disrupted, isopycnically banded virions. 25 While such sera contain antibodies to many different viral structural proteins, the specificity
of the RIA depends only on the purity of the 125I-labeled tracer antigen,
Second antibodies from several species are commercially available, but
can be prepared at minimal cost using purified IgG as the immunizing
antigen.
Assays for group-specific determinants of p30 proteins can be designed using antiserum to the homologous p30 protein (i.e., anti-MuLV
p30) instead of antiserum to FeLV. TM In such tests, minor type-specific
differences between different MuLV p30 proteins will be reflected in the
slopes of the competition curves as well as in the final percent competition
at "saturating" levels of competing protein. 25'26
While RIAs can be used as a general screen for MuLVs from laboratory mice (Mus musculus), they also can be used for detecting MuLVs
from several other Mus species (e.g., M. caroli 27 and M. cervicolor28). The
immunological properties of some of the latter isolates are, however,
distinctly different from those of laboratory mouse MuLVs.
H. Virological Assays
A. MIXED CULTURE CYTOPATHOGENICITY(THE "XC TEST")
The XC cell line was established from a Wistar rat tumor induced with
the Prague strain of Rous (avian) sarcoma virus. 29 These cells contain the
Rous sarcoma virus genome but do not release infectious viral particles.
When infected with certain MuLVs, XC cells undergo syncytium formaz5 C. J. Sherr and G. J. Todaro, Virology 61, 168 (1974).
26 W. H. Hunter, in " T h e Handbook of Experimental Immunology" (D. M. Weir, ed.), p.
608. Davis, Philadelphia, Pennsylvania, 1967.
27 M. M. Lieber, C. J. Sherr, G. J. Todaro, R. E. Benveniste, R. Callahan, and H. G. Coon,
Proc. Natl. Acad. Sci. U.S.A. 72, 2315 (1975).
2~ R. E. Benveniste, R. Callahan, C. J. Sherr, V. Chapman, and G. J. Todaro, J. Virol. 21,
849 (1977).
29 V. Klement, W. P. Rowe, J. W. Hartley, and W. E. Pugh, Proc. Natl. Acad. Sci. U.S.A.
63, 753 (1969).
422
SPECIALIZED TECHNIQUES
[35]
tion resulting in the appearance of plaques. The number of plaques produced is a direct function of the number of infectious viral particles, a
Procedure
1. Mouse cells (see below) are seeded sparsely into 60-mm plastic
petri dishes ( - 1 105 cells/plate) and are grown at 37 in Dulbecco's
modified Eagle's medium containing 10% fetal calf serum and antibiotics.
2. One day later, the cells are exposed to medium containing 25/zg/ml
DEAE dextran (Sigma Chemicals, St. Louis, Missouri) or 2 #g/ml Polybrene (Aldrich Chemical Company) for 1 hr at 37.
3. The medium is removed, and cultures are infected with dilutions of
virus-containing tissue culture medium ( - 1 . 0 ml) for 2 hr on a rocking
platform. The medium is changed after infection, and cultures are kept at
37 for 5 additional days until the cells reach confluence.
4. The medium is removed, and the cell layers are exposed for 10-30
sec to two GE germicidal bulbs at - 1 0 cm (60 erg/mm2/sec).
5. XC cells in fresh medium (10 6 cells/plate) are added to the irradiated monolayer, and cultures are incubated at 37 until plaque formation occurs (generally about 4 days after the addition of XC cells). Medium is changed 2 days after the addition of XC cells.
6. Cultures are washed with phosphate-buffered saline, fixed with
methanol, and stained with Giemsa (Grand Island Biological Corp.,
Grand Island, New York). The plaques appear as holes in the XC cell
sheet which contain some multinucleated giant cells.
Remarks. The XC test is useful for detecting most ecotropic MuLVs
that replicate efficiently in culture, but it cannot be used for either
xenotropic or amphotropic viruses, or for certain genetically transmitted
ectropic viruses that replicate poorly. Thus the test detects only a subset
of MuLVs, and a negative XC test does not mean that virus is not present.
For routine assays, the mouse SC-1 cell line is ideal since it lacks Fv-1
restriction and can be infected with N-, B-, or NB-tropic viruses. 4 Mouse
secondary embryo cultures a may be used to type N- and B-tropic isolates, the NII-I/3T35 and BALB/3T36 cell lines can also be used as prototype N- and B-cells, respectively.
Ultraviolet (UV) irradiation of the mouse cells prior to the addition of
XC cells kills the mouse cells at a faster rate than it inactivates their
capacity to produce virus. The dose of irradiation is approximate and
should be determined empirically for each mouse cell line. In addition, the
time after infection when UV irradiation is performed must be long
enough to allow plaque development but short enough to prevent the
a0 W. E Rowe, W. E. Pugh, and J. W. Hartley, Virology 42, 1136 (1970).
[35]
423
("S+L - T E S T S " )
424
SPECIALIZED TECHNIQUES
[35]
[36]
HUMAN ADENOVIRUSES
425
[36] H u m a n A d e n o v i r u s e s : G r o w t h , P u r i f i c a t i o n , a n d
Transfection Assay
426
SPECIALIZED TECHNIQUES
[36]
infection of "permissive" human cells results in the production of approximately 200,000 virus particles per cell, and the eventual death of
the infected cell. 1 The viral genome is transcribed and replicated in the
cell nucleus. There are at least two stages of expression of the Ad genome,
"early," before initiation of viral DNA replication at 6-7 hr postinfection,
and "late. ''2 Early genes, which are transcribed by host cell enzymes, are
arranged in four noncontiguous gene blocks, two on the r-strand and two
on the 1-strand, and represent about 30% of the single-stranded genome.
Proteins encoded by early genes (as many as 12 or 14 early polypeptides
have been identified) probably function in viral DNA replication, regulation of transcription, and cell transformation. Late genes, which code
virion structural proteins, are mainly on the r-strand, and represent most
of the remainder of the genome. Most exciting, late mRNA consists of
"spliced" mRNA molecules (reviewed in Wold et al. 3), wherein the main
coding body of the RNA is covalently linked to short "leader" RNA
sequences coded by DNA regions far upstream from each gene.
Ad infection of nonpermissive or semipermissive rodent cells may
result in the stable transformation of a small fraction (10-5--10-~) of the
cells. The transforming information [presumably gene(s)] has been
localized on the left-hand end of the r-strand within map position 1 to 7.5
(the entire genome consists of 100 map units), in that cells can be transformed by transfection with DNA restriction fragments containing this
region, H and all virion-transformed cells retain this region6 and express it
as RNA 7 and protein. The transforming "genes" of Adl2 (group A) and
Ad7 (group B) are in a similar region. The transforming region lies within
an early gone block.
In this article, we describe in detail methods used in our laboratory for
growth of cells for the propagation and plaque assay of all human Ad
serotypes. We also describe the isolation of viral DNA, and the transfection assay to determine Ad DNA infectivity and to localize Adtransforming genes.
Growth of Cultured KB Cells Used for the Propagation of Adenoviruses
The 31 human Ads (prototype strains are available from the American
Type Culture Collection) are grown in suspension cultures of human KB
cells for large-scale virus propagation, and in monolayer culture (petri
dish) for plaque assay of Ad infectivity. HeLa cells are used in some
laboratories with comparable results. There is some evidence that HeLa
lM. Green, Cold Spring Harbor Syrup. Quant. Biol. 27, 219 (1962).
~1F. L. Graham, P. J. Abrahams, C. Mulder, H. L. Heijneker, S. O. Warnaar, F. A. J. de
Vries, W. Ficrs, and A. J. van der Eb, Cold Spring Harbor Syrup. Quant. Biol. 39, 601
(1974).
[36]
HUMAN ADENOVIRUSES
427
and KB cell lines are derived from the same H e L a cell parental line. Both
lines are available from the American Type Culture Collection. Below are
described the preparation of growth media and the cultivation of KB cells
for large-scale production of virus and for infectivity assay.
Preparation of Cell Culture Media
Triple-distilled water is used to prepare all media. Monolayer cultures
are grown in serum-supplemented Eagle's minimal essential medium
(MEM), TM a synthetic medium consisting of a balanced salt solution and
amino acids and vitamins essential for growth of cultured human cells; it
is often supplemented with nonessential amino acids. Joklik-modified
MEM which lacks calcium (to prevent cell clumping) and contains a 10fold higher phosphate concentration (increased buffering capacity) is used
for suspension cultures. There are several ways to prepare media: by
assembly of laboratory-prepared or commercially available concentrates
( 10 to 200) of balanced salts, amino acids, and vitamins; by purchase
of complete 1 or x 10 media; and by hydration of powdered media
(complete except for sodium bicarbonate). Most commercial powdered
media appear to be as satisfactory as laboratory-prepared media for the
growth of cells, and they cost about the same.
As a precaution against contamination and errors in media components, we routinely maintain cells on two independent media (A and B),
using sera, media, phosphate-buffer saline (PBS), and trypsin-EDTA,
from two different sources or lots.
Preparation of 20 Liters of MEM from Powdered Medium. Two packages of powdered media [for 10 liters each, Grand Island Biological Co.
(GIBCO) or KC Biologicals]are dissolved in 18 liters of water in a
stainless-steel pressure vessel (Millipore Corp., 5-gallon capacity).
Twenty milliliters of a stock x 1000 solution of antibiotics are added
(either penicillin and streptomycin, or x 1000 Gentamycin), followed by 44
g of NaHCOa for monolayer MEM or 40 g of NaHCOs for suspension
MEM. The pH is adjusted to 7.2 with 1 N N a O H or 1N HC1, made up to
20 liters with water, and sterilized with a pressure pump by filtration
through a 127-mm disc prefilter (Millipore AP2512750) placed on top of a
142-mm, 0.20-#m metricel filter (Gelman, from Fisher Scientific Co. ) or a
0.22-p.m GS filter (Minipore). Media are sterility-tested for at least a week
prior to use, using both thioglycolate broth (Difco) and blood agar plates
[tryptic soy agar containing 5% sheep blood (Gibco Diagnostics)], and
stored at 4. Before use, 5% or 10% sera (donor calf or donor horse
obtained from KC Biologicals, Flow Laboratories, Inc., or Gibco) are
added.
12H. Eagle, Science 130, 432 (1959).
428
SPECIALIZED TECHNIQUES
[36]
[36]
HUMAN ADENOVIRUSES
429
430
SPECIALIZED TECHNIQUES
[36]
[36]
HUMAN ADENOVIRUSES
431
432
SPECIALIZED TECHNIQUES
[36]
the Ti 50 rotor with an adaptor), filled with CsC1 solution (density -- 1.34
g/cm a, 56 g of CsC1 in 116 ml of 10 mM Tris HCI, pH 8.1), and centrifuged for 16-24 hr at 30,000 rpm. The visible band in the middle of the
gradient, at a buoyant density of 1.34 g/cm a, is collected from the side by
tube puncture, adjusted to 6.5 ml by the addition of CsC1 solution at 1.34
g/cm a, and recentrifuged as described above. The twice-banded virus is
collected and stored at 4 .
[36]
HUMAN ADENOVIRUSES
433
D N A 14'18
434
SPECIALIZED TECHNIQUES
[36]
times with water-saturated ether, and then dialyzed against 200 volumes
of 10 mM Tris. HC1, pH 8.1, containing 1 mM EDTA for 24 hr. The
absorbance at 250, 260, and 280 nm is determined. The amount of DNA is
calculated based on an A2~0 unit equivalent to 50/~g DNA/ml. The 260
nm/280 nm and 260 nm/250 nm ratios are about 1.9 and 1.1, respectively.
Yields of 60-80% homogeneous, intact duplex DNA molecules, sedimenting at 30-31 S, are obtained. 4 Approximately 0.5 mg of Ad2 DNA is
isolated from virus produced in 1 liter of cells.
Transfection-InfectivityAssay for Viral D N A and D N A Restriction
Fragments
Transfcction assays with purified viral D N A (rather than virions) arc
useful for several kinds of studies. For example, recent recombinant D N A
procedures, and related procedures, arc beginning to bc exploited for the
construction of A d mutants in vitro.20In addition, transfectionprocedures
using A d D N A and D N A restrictionendonucleasc fragments have been
used to map the Ad-transforrning genes. The infectivityof Ad D N A and
the transforming activity of Ad D N A and restrictionD N A fragments are
assayed with the calcium phosphate procedure, 11"21modified to include
treatment of ceils with dimethylsulfoxide (DMSO) 2~2 to enhance the
transfection activity. Ceils used for transfection-infectivity assay arc
permissive human K B cells in petri dishes as described in preceding sections. Cells (nonpermissive) used for transfection-transformation assay
are prepared from 16-day rat (Wistar) whole embryos. Cells prepared
from baby rat (6- to 7-day-old) kidneys are used by Graham et al. i~ A
permanent rat cell line for transfection-transformation experiments has
also been described. 2a
Cell procedures are performed under aseptic conditions. Several embryos are minced with scissors, washed 2 or 3 times with PBS lacking
calcium and magnesium, and digested with warmed (37 ) trypsin-EDTA
for 10-20 min at room temperature. Cells are centrifuged for 10 min at
230 g, chilled, suspended in MEM with 10% calf serum, and filtered
through two layers of gauze. Cells are collected from the filtrate by centrifugation, a 0.5% suspension is prepared in MEM containing 10% calf
serum, and 60-ml amounts are seeded in 32-ounce culture flasks; usually
one embryo provides sufficient cells for one flask. When monolayers are
nearly confluent (3-4 days), cells are subcultured by trypsin-EDTA treat20 G. Chinnadurai, S. Chinnadurai, and M. Green, J. Virol. 26, 195 (1978).
2~F. L. Graham, J. Smiley, W. C. Russell, and R. Nairn, J. Gen. Virol. 36, 59 (1977).
22N. D. Stow and N. M. Wilkie, J. Gen. Virol, 33, 447 (1976).
2a G. Kimura, A. Itagaki, and J. Summers, Int. J. Cancer 15, 694 (1975).
[36]
HUMAN ADENOVIRUSES
435
ment into 60-mm dishes in MEM wlth 10% fetal calf serum for transfection (5 105 cells/dish). When the cell sheets are 80-90% confluent, 0.5 ml
of solution, containing either Ad DNA or DNA restriction fragments, is
added to each dish without removing the medium. Ad DNA is prepared as
described above. Preparative isolation of Ad DNA restriction endonuclease fragments is described in detail in Wold et al. 24 The viral DNA
solution is prepared as follows.
(1) Ad DNA (0.1/xg/ml to 0.5/zg/ml for DNA fragments smaller than
2-3 x 106 daltons, or 1.0/zg/ml to 10/xg/ml for larger fragments) is diluted
into HEPES-buffered saline (a 2 solution is prepared from 8 g of NaC1,
370 mg of KC1, 125 mg of Na2HPO4 2 I-I20, 1 g of glucose, and 5 g of
N-2-hydroxyethyl-piperazine-N'-2-ethanesulfonic acid, made up to 500
ml and pH 7.05, and sterilized by filtration) containing 10/~g/ml of salmon
sperm DNA.
(2) CaC12 is added (from a 2.5 M stock solution sterilized by autoclaving) to a final concentration of 125 mM. After 15-20 min at room temperature, the mixture becomes slightly turbid and is added to petri dishes
containing rat embryo cells. After 4 hr of incubation at 37 in a CO2
incubator, the medium is removed, the cultures washed once with medium, and 1 ml of 25% DMSO in HEPES-butfered saline is added to each
plate. After 2-4 min, the DMSO is removed, the cultures washed with
medium, and 5 ml of fresh MEM with 10% fetal calf serum are added.
Three or four days later the medium is replaced with calcium-free MEM
with 5% fetal calf serum. Incubation is continued (the medium is changed
twice weekly) until transformed cell foci appear (positive controls are
always included in assays). Generally, about 2--4 weeks are required for
foci to develop. Foci appear macroscopically as raised colonies, and microscopically as multiple layers of densely packed, criss-crossed cells.
Transformed colonies can be quantitated under a dissecting microscope at
a magnification of x30 to 40. Alternatively foci can be counted after the
cultures have been fixed by a treatment with absolute methanol for 5 min,
air dried, and stained with Giemsa. The efficiency of transfection of an
Ad2 DNA HindlII digest is approximately 10 foci per microgram of DNA.
As final proof that foci represent cells transformed by viral DNA, clonal
lines must be isolated and shown to contain integrated viral DNA and to
synthesize viral RNA. For this purpose, individual foci are taken up into a
Pasteur pipette by gentle aspiration with a rubber bulb and transferred
into a new petri dish containing calcium-free MEM with 10% fetal calf
serum. Clonal lines are then developed by selection and subculture and
used for subsequent biochemical analyses.
24 W. S. M. Wold, M. Green, and J. K. Mackey, Methods CancerRes. 15, 69--161"(1978).
[37]
SOURCES
OF
STABLE
CELL
LINES
439
[37] S o u r c e s o f S t a b l e C e l l L i n e s
B y GERARD J. MCGARRITY
I. Introduction
T. C. Hsu summarized the advantages of cell culture systems: 1
1. They provide a continuous supply of homogenous cellular material
for biochemical experiment~ as well as for practical use in medical and
public health work.
2. The cells in vitro can be manipulated advantageously in many ways.
This cannot be done with cells in vivo.
3. They can be stored in a deep-frozen state without changing their
growth rate and genetic composition, and they c a n b e revived at will.
4. Using cell cultures is more economical than rearing animals and
performing experiments with intact animals.
5. They save lives of animals.
To obtain full benefit of these advantages, cell cultures used in research and diagnosis should be standardized as fully as possible and subjected to extensive quality-control procedures. (For detailed methods on
this topic, see this volume [2] and [13].) Quality control should include
selection of the cell culture to be used and the source of the culture. Many
cell cultures contain undetected adventitious agents such as bacteria,
yeast, viruses, and mycoplasmas. In this laboratory the annual mean
infection rate of cell cultures with mycoplasmas has ranged from 3.414.9%. This is based on testing of more than 6000 cell cultures.
Mycoplasma-infected cell cultures are useless in controlled standardized
procedures. Introduction of a mycoplasma-infected culture into a laboratory generally results in spread of the infection to all cultures in the
laboratory. 2
A significant number of cell cultures are contaminated with other cells,
especially HeLa. Gartler concluded that many of the permanent human
cell lines may in fact be HeLa, based on electrophoretic variant forms of
isoenzymes as genetic markers; a this observation has been reinforced. 4,5
More recently, Nelson-Rees and Flandermeyer have reported that 41 of
1T. C. Hsu, in "The Future of Animals,Cells, Models,and Systemsin Research Development, Educationand Testing," p. 180. Natl. Acad. Sci., Washington,D.C., 1977.
2 G. J. McGarrity,In Vitro 12, 643 (1976).
a S. M. Gartler, Natl. Cancer Inst., Monogr. 26, 167 (1967).
40. J. Miller,D. A. Miller,P. W. Allderdice,V. G. Dev ,and M.S. Grewal,Cytogenetics 10,
338 (1971).
5 W . Nelson-Rees, R. R. Flandermeyer,and P. K. Hawthorne,Science 184, 1093 (1974).
ME~FHODS IN ENZYMOLOGY, VOL. LVIII
440
[37]
253 cell cultures examined (16%) were not as purported; HeLa cell contamination and wrong species accounted for 36 of these 41 errors. 6
To protect against mycoplasma infection, interspecies and intraspecies
contamination, and other problems, cell cultures should be acquired only
from sources that have performed reliable quality-control checks. The
common practice of acquiring cultures from colleagues is risky and is
discouraged; several episodes of mycoplasma infection have been traced
to this practice. Even reassurances of negative mycoplasma assays made
months, even weeks, before is no guarantee of a culture's present status.
The purpose of this article is to list the major sources of cell cultures
and information pertinent to each source.
[37]
441
442
[37]
known, McKusick numbers and HL-A antigens are listed after appropriate cultures.
Starter cultures are supplied. These are obtained by inoculating a
frozen ampule into a T25 flask and growing for several days prior to
shipping. The cost per T25 flask is $20 plus shipping. The catalogue can be
obtained by writing to the Human Mutant Cell Repository, Institute for
Medical Research, Copewood Street, Camden, New Jersey 08103.
[37]
443
444
[38]
[38] I s o l a t i o n o f F i b r o b l a s t s f r o m P a t i e n t s
[38]
ISOLATION OF FIBROBLASTS
445
nitrogen. For a small charge, samples are thawed, started in culture, and
shipped to investigators on request. Investigators contemplating initiating
a cell culture laboratory for studies on normal or mutant human fibroblasts would be well advised to obtain the catalogue of existing strains.
1. Obtaining a Skin Biopsy
Materials
Biopsy tray with 2 4-mm punch biopsy instruments, 2 curved scissors, and 2 forceps. A second set of instruments is provided in
case one gets contaminated accidentally while attempting the
biopsy.
Sterile gauze
Isopropyl alcohol, 70%, 30 ml
Lidocaine without epinephrine, 1%
1 tuberculin syringe
1 screw-capped tube containing 10 ml of medium
Note: All glassware items--dishes, tubes, coverslips--are acid
washed with concentrated HNO3 and rinsed extensively with
deionized water before sterilizing by autoclave.
The site of removal of tissue depends on the purpose. Studies of testosterone binding and/or metabolism should be done on fibroblasts from
genital skin. 4 For nearly all other general purposes, we take a biopsy from
the subdeltoid area of the upper arm (the lateral aspect of the arm ~ the
distance from the shoulder to the elbow). Others use the anterior aspect of
the forearm, 2 inches below the elbow crease. 5 The area is first scrubbed
twice thoroughly with a sterile gauze fiat moistened with 70% isopropyl
alcohol. Then an area about 1-1.5 cm 2 is anesthetized by injection of 0.2
ml of 1% Lidocaine without epinephrine intradermally with a tuberculin
syringe. After 2-3 min, a 4-mm sterile punch biopsy instrument is rotated
with pressure to cut a circular flap through the epidermis from the center
of the anesthetized area. The edge of the flap is raised with a sterile
forceps, and the flap is removed by cutting under it with a small curved
scissors. The removed flap is dropped into a sterile 125-mm screw-capped
tube containing 10 ml of tissue culture medium (MEM-Earle's containing
15% heat-inactivated fetal calf serum, penicillin, and streptomycin, see
below).
Biopsies are usually "planted" immediately, but they may be planted
up to 4 days after the biopsy, if stored at room temperature in cell culture
4 j. E. Griffin, K. Punyashthiti, and J. D. Wilson, J. Clin. Invest. 57, 1342 (1976).
5 j. T. Cooper and S. Goldstein, Lancet 2, 673 (1973).
446
[38]
This operation is carried out in a laminar-flow hood to avoid contamination of the sample. The aim is to cut the sample into small pieces,
distribute it into two dishes that will be fed from separate media sources
to reduce chances of loss by contamination, and to hold the tiny pieces in
place by placing them under a coverslip which in turn is held in place by
silicone grease (see Fig. 1). 5,6
Pour the biopsy and medium from the tube to a sterile glass petri dish.
Use two round-bladed scalpels held opposing each other like scissors
blades. Bisect the skin biopsy by pressing the two blades down against the
glass and rolling the edges as if closing them in opposite directions (as a
6 R. J. Warren and C. De La Cruz, Exp. Cell Res. 71, 238 (1972).
[38]
ISOLATION OF FIBROBLASTS
.SK,
447
REASE
60 mm DISH
FIG. l. View of tissue culture dish in which explants (fragments from a skin biopsy) have
been "planted" beneath 25-mm coverslips that are held in place with silicone grease.
closing scissors). Then each piece is further divided in two and subdivided
until one has 12-16 pieces of 1 mm 3 or less.
Transfer the pieces to two dry 60-mm tissue culture dishes. The samples can easily be transferred with the tip of the blade of one of the
scalpels and will adhere to the dry dish. Arrange samples to fit under a
round 25-mm coverslip as shown. Use a sterile wooden applicator or a
disposable needle to place a spot of sterile silicone grease for one quadrant of the coverslip. Lower a coverslip into place with a forceps so that it
covers the skin pieces and the silicone grease. If skin pieces lie beyond it,
tuck them under the coverslip with a scalpel blade before pressing the
coverslip down tightly enough to secure them. Add about 0.2 ml medium
with a small pipette (1 ml) at the margin of the coverslip and allow it to
move across by capillarity to displace the air beneath the coverslip. Once
e coverslips have medium beneath them (and no large air bubbles), 5 ml
medium are added to the dishes directly to the top of the coverslips.
e dishes are placed in trays, incubated in a humidified CO2 incubator
with 5% CO2 in air, and not touched for 10 days. At 10 days, the dishes are
examined with an inverted microscope. By this time an epithelial outgrowth may be evident at the margin of the skin pieces. From this point,
dishes are " f e d " 3 times a week. Medium is aspirated from the edge of the
tilted dishes and replaced with 5 ml of fresh medium. By 2 weeks, fibroblasts are evident at the margins of the epithelial layer and moving out
from the explant. Four to six weeks from the time the biopsy was taken,
the cells will have grown well beyond the coverslip and be ready for
transfer (splitting).
At this point, the medium is removed, and the dishes are washed with
2 ml of 0.25% trypsin, following which 1.0 ml of 0.25% trypsin is added
and the dishes placed at 37 for 5-7 min. The cell layer will be seen
detaching from the dish. Then the coverslips are dislodged to be certain
448
[38]
the trypsin has had access to cells beneath them, and the cell layer is
dispersed by adding 4 ml of serum-containing medium and pipetting up
and down 3-4 times. The cells are sedimented in a sterile tube by centrifuging at 1000 rpm, resuspended in 5 ml of serum-containing medium,
and added to a 60-mm dish. Within 2-4 days, this dish should be confluent
and ready to split further. The trypsinization procedure outlined in this
paragraph is repeated. Resuspended cells from each 60-mm dish are distributed into 2 100-mm dishes, i.e., a 1:5 split, which are fed 3 times
weekly with 10 ml of medium. Within 7 days, the dishes should be confluent and may be propagated further by splitting 1:5 to 1:8 (see Section 4)
or prepared for storage by freezing (Section 3).
3. Freezing Cells for Storage and Recovering Them from Frozen Storage
Materials
[38]
ISOLATION OF FIBROBLASTS
449
- 7 0 freezer to allow them to freeze. The vials of frozen cells are transferred to a liquid nitrogen storage tank the next working day. We routinely
thaw one vial and initiate the culture to be certain that the cells are
recoverable from frozen storage and not contaminated. An alternate
method to freezing in a -70 freezer is use of a BF-5 cap attachment for a
Linde Liquid nitrogen tank which allows slow freezing of nine vials at a
time in the vapor phase. They are held here for 90 min before transfer to a
cannister for storage.
To recover cells from frozen storage, one warms the bottom of the vial
to 37 in a water bath immediately on removal from the freezer. As soon
as the sample has melted (which is obvious as the sample turns from ice to
liquid) the vial is wiped with 95% ethanol to sterilize the outside, air dried,
and the cells removed from the opened vial with a sterile 3-cc syringe with
a 19-gauge needle. The cells are diluted with 10 ml of serum-containing
medium to dilute out the DMSO; cells are sedimented, resuspended in 5
ml of serum-containing medium, and placed in a 60-mm dish. Within 1-2
days at 37 , this sample is usually ready to propagate and is "split" 1:5
into two 100-mm dishes.
From that point, most cell lines can be split every 7-8 days from 1:5 to
1:8 to produce large amounts of cells.
Diameter of
growth surface (mm)
Area of
growth surface (cm)
Media per
feeding (ml)
100 mm x 20 mm
60 mm x 15 mm
35 mm x 10 mm
81
51
32
51.5
20.4
8.5
10
5
2
450
SPECIFIC
CELL
LINES
[39]
[39] Cell L i n e s f r o m I n v e r t e b r a t e s
By W. FRED HINK
Cell lines from about 70 different species of invertebrates have been
reported. Most are from insects, but several lines have been established
from ticks and snails. The lines have developed from primary cultures of
embryos, hemocytes, ovaries, imaginal discs, fat bodies, and macerated
larvae, pupae, or adults. This article presents general information that is
applicable to the culturing of most of these lines. It does not deal with
primary cultures or details about each specific cell l i n e .
Maintenance of Cell Lines
[39]
INVERTEBRATES
451
Subculturing
While almost any cell culture vessel is adequate (TC petri dish, glass
T-flask, polystyrene disposable TC flask, etc.), most lines are maintained
in TC flasks with 5-ml volumes of media and 25-cm 2 growth surfaces, and
this discussion will center around the use of this size flask. Subculturing is
done by transferring an aliquot of cells from a parent culture to a new flask
containing fresh medium. New cultures are initiated with 1-3 x 105cells/ml.
The ratio of the volume transferred from the parent culture to the volume
of the new culture is termed the split ratio. A 0.5-ml aliquot of suspended
cells from a 5.0-ml parent culture transferred to 4.5 ml fresh medium is a
1:10 split ratio. Split ratios for different lines vary between 1:2 and 1:25,
and subculture intervals range from 2 days to several weeks. When one
first receives a specific line and attempts initial maintenance, it is advisable to follow the split ratio and subculture interval as used by the parent
laboratory.
If difficulty is encountered in keeping cells in good condition, the
subculture techniques may be modified. Cells should be subcultured while
in the exponential growth phase or just as they are entering the stationary
phase. To determine when to subculture, growth curves should be done
soon after receiving cells. The single cell suspensions are counted with a
hemocytometer. Growth curves from representative insect cell lines show
that after initiation of new cultures, there are lag periods of several hours
to 3-4 days. Exponential growth may last for 2-7 days after which the cell
population numbers level off or decline. Population doubling times during
exponential growth range from 16 hr for T. ni to 48 hr for Antheraea
eucalypti, and maximum cell densities are from 1 x 106 to 1 x 107 cells/ml.
Many insect lines grow loosely attached or in suspension, and these
are gently agitated to obtain a homogenous suspension prior to
subculturing.
Attached cell lines must be released from the culture flask and dispersed as single cells before subculturing. For most attached lines, the
cells are removed by scraping with a rubber policeman and resultant
clumps dispersed by gentle pipetting, xa Other techniques employ a small
i p. E. Eide, J. M. Caldwell, and E. P. Marks, In Vitro 11,395 (1975).
J. L. Vaughn, R. H. Goodwin, G. J. Tompkins, and P. McCawley,In Vitro 13, 213 (1977).
452
[39]
TABLE I
R I N A L D I N I ' S SOLUTION a
Compound
mg/100 ml solution
NaC1
KCI
Na.zPO4 H20
D-glucose
NaHCOz
NaaC6H507 "2 H20
Demineralized water
800
20
5
100
100
67.6
to 100 ml
a Rinaldini.3
[39]
INVERTEBRATES
453
Storage
If cells are not transferred to fresh medium when they reach maximum
density, they begin to die and, depending on the cell line, all will be dead
in 10-20 days. The longevity of cell viability can be increased by transferring newly subcultured cells to 5. In my laboratory, the following lines
were stored at 5 in fresh media and samples taken every week to determine if they would recover: T. ni (TN-368) can be stored 14 days; A.
eucalypti for 21 days; Grace'sAedes aegypti for 40 days; and Lasperesia
pomonella (CP-1268 and CP-169) for 90 days. In all cases, cell growth was
slower after storage, requiring about five subcultures for the growth rate
to return to normal.
As with vertebrate cells, insect cells also may be frozen in liquid
nitrogen or ultra-low temperature freezers at - 7 0 to - 9 0 . The cell culture
media are supplemented with either sterile 10% glycerol or 8% dimethysulfoxide. Cells from a culture in late stage of exponential growth
are centrifuged at 380 g for 10 rain and resuspended in glycerolsupplemented medium at one-half the original volume. The cell suspension
is dispensed in 1.0-ml volumes into glass ampules and immediately sealed
by use of a propane torch and glass rod. Animal cells should be slowly
frozen at rates of 1-3 per minute to about -30; programmable freezers
may be used for this purpose. We obtain slow freezing by wrapping the
ampules in insulation before placing them in a freezer and, after 2 hr,
transferring them to liquid nitrogen. Cells are thawed quickly by removing
them from the nitrogen and plunging the ampules in a 30 water bath
where they are agitated to accelerate melting. The content of one ampule
is added to 4 ml of fresh medium in a TC flask. For most lines, the cells
will attach within 2-4 hr at which time the medium with the glycerol
supplement is removed by pipetting and 5 ml of fresh medium added. If
cells do not attach, the cryoprotective agent may be removed by centrifuging freshly thawed cells, discarding the supernatant fluid, and resuspending cells in fresh medium.
Quality Control
Occasionally, a batch of basal medium, serum, hydrolysate, or other
ingredient will be toxic or will not support normally expected growth
rates. For this reason, the lot number of all ingredients in the complete
medium should be recorded. When an ingredient with a new lot number is
to be employed and before supply of the previously used batch is depleted,
it should be incorporated in a small batch of complete medium that contains no other new ingredients. This should be used to culture cells for at
454
[39]
[39]
INVERTEBRATES
4.55
Gibco MEM nonessential amino acids. The pH was adjusted to 6.9 and cultures gassed with 5% CO2 in air. A series of manipulations consisting of
addition of various supplements and culturing in roller bottles were required to obtain suspension cultures. Cells in suspension reach densities
of 1-4 106 cells/ml with a generation time of 30 hr. Another D.
m e l a n o g a s t e r line, (GMz), 14 was grown in insect cell culture media in
Erlenmeyer flasks on a gyratory shaker at 180 rpm with a 1-inch radial
stroke.15
The moth cell line (T. n i , TN-368) 16 grew in suspension in 100-ml spin
flasks and 4-liter fermentors. 17 The culture medium (TNM-FH, Table IV)
was modified by addition of 0.1% methylcellulose (50 cps) to prevent cell
clumping. Aeration of cultures produced higher growth rates and final cell
densities. Periodic adjustment of medium pH also resulted in higher maximum cell densities.
Media
According to my most recent tabulation, there are 34 different formulations of media for culturing invertebrate cell lines. It is impractical to
consider all of these here, and I will therefore discuss only the most
common media. Before dealing with specific media, some general comments will be made.
Insect cell culture media differ from media designed for vertebrate
cells by having different ion balances, elevated amino acid concentrations,
generally phosphates as buffers rather than carbonates, usually lower pH,
and higher osmotic pressures. There is some question as to the significance of these differences since a few insect cell lines grow in vertebrate
media. 8,1a,18,19 Established insect cell lines appear to be rather insensitive
to Na/K ratios altered from 0.67-1.38 and calcium and magnesium levels
reduced by one-half and three-fourths, respectively. 2 However, as with
vertebrate cells, excess potassium is toxic. 21 The increased amounts of
amino acids are present in some formulas because insect blood has high
concentrations of these compounds. Requirements for specific amino
acids vary with different lines, and patterns of utilization are different
14G. Mosna and S. Dolfini,Chromosoma 38, 1 (1972).
15T. Miyake, K. Saigo, T. Marunouchi, and T. Shiba, In Vitro 13, 245 (1977).
18W. F. Hink, Nature (London) 226, (1970).
17W. F. Hink and E. Strauss, in "Invertebrate Tissue Culture" (E. Kurstak and K.
Maramorosch, eds.), p. 297. Academic Press, New York, 1976.
18M. Pudney, personal communication.
~9A. H. Mclntosh, K. Maramorosch, and C. Rechtoris,In Vitro 8, 375 (1973).
20j. L. Vaughn,In Vitro 9, 122 (1973).
21T. J. Kurtti, S. P. S. Chaudhary, and M. A. Brooks,In Vitro 11,274 (1975).
456
[39]
even between cell lines from the same insect species, z2 In fact, some
amino acids essential for growth of living intact insects are not essential
for cells in vitro .23
Osmotic pressure and pH are important parameters in media formulation. Studies with moth cell lines indicate that growth is reduced when
osmotic pressure varies more than about 40 msM/kg on either side of the
osmotic pressure (316 msM/kg) of the "normal" medium. 24 Our policy is
to have less than _ 10 msM/kg variation in batches of media. The pH of
most media range from 6.2-7.0 with media for som.e cockroach lines being
as high as 7.4. The optimum initial pH for moth (Heliothis zea) cells is
between 6.5-7.025 and for two leafhopper cell lines it is 6.3-6.4. 26 When
the pH of the medium was held at preset levels of 5.8, 6.0, 6.3, 6.5, and 6.7
for the life of suspension cultures ofT. ni cells, the optimum was between
pH 6.0-6.5, whereas pH 6.7 was clearly detrimental.
Supplements and Additives to Basal Media
For purposes of discussion, the basal medium is that portion of the
complete medium that contains synthetic chemically defined compounds.
There are no chemically defined media for invertebrate cell lines, and all
basal media are supplemented with undefined natural ingredients. Fetal
bovine serum, at concentrations of 5-20%, is used in nearly all media, and
it is usually heat inactivated at 56 for 30 rain. Other complex ingredients
such as lactalbumin hydrolysate furnish amino acids, and yeast hydrolysate provides both amino acids and vitamins.
Antibiotics are often added, but their use should be discouraged because they can mask microbial contamination. If contamination is present, it is better to know it immediately instead of carrying undetected
low-level contaminants and having them affect experimental results. It
appears that dependence on the presence of antibiotics leads to the development of rather careless nonsterile techniques. The two most common
antibiotics and concentrations are penicillin G at 100 U/ml and streptomycin sulfate at 100/xg/ml. A 100 stock solution is prepared by dissolving
10,000 U penicillin and 10 mg streptomycin/ml PBS, filter-sterilized; 1.0 ml
is added to 99 ml of complete medium just before use.
22 W. F. Hink, B. L. Richardson, D. K. Schenk, and B. J. Ellis,Proc. Int. Colloq. lnvertebr.
Tissue Cult., 3rd, 1971 p. 195 (1973).
23 j. Mitsuhashi, J. Insect Physiol. 22, 397 (1976).
z4 T. J. Kurtti, S. P. S. Chaudhary, and M. A. Brooks, In Vitro 10, 149 (1974).
25 T. J. Kurtti and M. A. Brooks, in "Insect and Mite Nutrition" (J. G. Rodriguez, ed.), p.
387. North-Holland Publ., Amsterdam, 1972.
26 G. Martinez-Lopez and L. M. Black, In Vitro 13, 777 (1977).
[39]
INVERTEBRATES
457
M e d i a Sterilization
GRACE MEDIUM
This medium (Table 11)27 is designed to resemble the chemical composition of silkworm, B o m b y x m o r i , h e m o l y m p h and is a modification o f
W y a t t ' s medium, z8 The basal medium is supplemented with various combinations and amounts of FBS, chicken egg ultrafiltrate, yeast extract,
lactalbumin hydrolysate, and bovine serum albumin prior to use. It is
used to culture cell lines from the Australian emperor gum moth, A.
eucalypti; zr silkworm, B. Mori; 29 spruce b u d w o r m , Choristoneura
f u m i f e r a n a ;3o cotton bollworm, H . zea ;31 codling moth, L. p o m o n e l l a ;32
g y p s y moth, L y m a n t r i a dispar; 33 forest tent caterpillar, M a l a c o s o m a disstria; 34 t o b a c c o hornworm, M a n d u c a s e x t a ; 1 cynthia moth, S a m i a
cynthia ;35 cabbage looper, T. ni ;16 mosquito, A . aegypti ;36 mosquito, A e d e s
v e x a n s ;37 and mosquito, Culiseta inornata .37
PROCEDURE FOR MAKING l0 LITERS OF GRACE MEDIUM 3s
1. Weigh out amino acids (B) and combine them in a 400-ml beaker.
2. Weigh out sugars (C) and combine them in a 400-ml beaker.
27T. D. C. Grace, Nature (London) 195, 788 (1962).
2s S. S. Wyatt, J. Gen. Physiol. 39, 841 (1956).
29T. D. C. Grace, Nature (London) 216, 613 (1967).
30S. S. Sohi, Proc. Int. Colloq. Invertebr. Tissue Cult., 3rd, 1971 p. 75 (1973).
31W. F. Hink and C. M. Ignoffo,Exp. Cell Res. 60, 307 (1970).
32 W. F. Hink and B. J. Ellis, Curr. Top. Microbiol. Immunol. 55, 19 (1971).
33j. M. Quiot, personal communication.
34 S. S. Sohi, Proc. Int. Colloq. lnvertebr. Tissue Cult., 3rd, 1971 p. 27 (1973).
35j. Chao and G. H. Ball, Curr. Top. Microbiol. Imrnunol. 55, 28 (1971).
36T. D. C. Grace, Nature (London) 211, 366 (1966).
37 B. H. Sweet and J. S. McHale, Exp. Cell. Res. 61, 51 (1970).
38W. F. Hink, unpublished data.
458
[39]
TABLE II
GRACE INSECT TISSUE CULTURE MEDIUM (g/lO liters)a
Salts
NaH2PO4 H~O
NaHCO3
(A) KCI
MgCI2 6 1-120
MgSO4 7 H~O
(I) CaCI~.2 H20 (sep.)
I 0.08
3.50
22.40
22.80
27.80
13.25
Amino acids
L-arginine HCI
L-aspartic acid
L-asparagine
L-alanine
B-alanine
L-glutamic acid
L-glutamine
L-glycine
L-histidine
L-isoleucine
L-leucine
L-lysine HC1
L-methionine
L-proline
L-phenylalanine
DL-serine
L-tryptophan
L-threonine
L-valine
L-cystine HCI (sep.)
L-tyrosine (sep.)
7.0
3.5
3.5
2.25
2.0
6.0
6.0
6.5
25.0
0.5
0.75
6.25
0.5
3.5
1.5
11.0
1.0
1.75
1.0
0.25
0.5
(B)
(G)
(H)
Sugars
Sucrose
Fructose
Glucose
266.8 /
4.0
7.0
(C)
Organic acids
Malic
Alpha-ketoglutaric
Succinic
Fumaric
6.7 1
3.7
0.6
0.55
(D)
Vitamins
Thiamine HC1
Riboflavin
Ca pantothenate
Pyridoxine HC1
p-Aminobenzoic acid
Folic acid
Niacine
Isoinositol
Biotin
Choline chloride
0.0002
0.0002
0.0002
0.0002
0.0002
0.0002
0.0002
0.0002
0.0001
0.002
(E)
Antibiotics
Penicillin G, Na salt
Streptomycin sulphate
0.3 "~
1.0 /
(F)
a Grace.2r
3. Weigh out organic acids (D) and combine them in a 100-ml beaker.
4. Weigh out L-cystine (G) and L-tyrosine (H) and combine them in a
50-ml beaker.
5. Weigh out calcium chloride (I) and place in a 100-ml beaker.
6. A x 1000 stock solution of vitamins (E) is made up by adding 2 mg
thiamine, 2 mg riboflavin, 2 mg Ca pantothenate, 2 mg pyridoxine, 2 mg
p-aminobenzoic acid, 2 mg folic acid, 2 mg niacin, 2 mg isoinositol, 1 mg
biotin, and 20 nag choline chloride to a 100-ml volumetric flask and bringing up to volume with water. Filter-sterilize, dispense in 10-ml portions,
and store at - 2 0 .
7. If antibiotics (F) are to be used, they are made up in a x 100 stock
[39]
INVERTEBRATES
459
Fraction
A
B
C
D
KOH
GH
I
HC!
E
Totals:
Volume occupied by
chemicals when
dissolved (ml)
37.0
53.0
170.0
5.0
2000.0
2000.0
2OOO.0
2000.0
--
0.5
3.0
--268.5
Volume of
liquid (ml)
175.0
15.0
50.0
27.0
10.0
8277.0 =
8545.5 m l
460
[39]
4.0
5.0
19.5
2.0
4.0
mg
mg
mg
mg
mg
82.3 mg
53.2 mg
[39]
INVERTEBRATES
461
TABLE III
YUNKER, VAUGHN, AND CORY MEDIUMa
90.0 ml Grace medium
10.0 ml FBS
10.0 ml Egg ultrafiltrate
1.0 g Bovine plasma albumin
a Yunker
e t a l . asa
TABLE IV
HINK (TNM-FH) MEDIUMa
90.0
8.0
0.3
0.3
ml Grace medium
ml FBS
g Lactalbumin hydrolysate
g TC Yeastolate
TABLE V
GOODWIN IPL-52 MEDIUM (mg/liter)a'b
I(NH4)MorO24 4 H20
[ C o C l 2 6 H20
AJCuClz 2 H20
[MnC12 4 H20
[ Zn C12
B J'FeSO4_ 7 HzO
LAspartic acid
L-Arginine hydrochloride
L-Aspartic acid
L-Asparagine
L-Cystine (dissolve in
10% NaOH)
L-Glutamic acid
L-Glutamine
L-Glycine
L-Histidine
Hydroxy-L-proline
L-Isoleucine
L-Leucine
L-Lysine hydrochloride
L-Methionine
L-Proline
L-Phenylalanine
a Goodwin?9
b Goodwin.40
0.040
0.050
0.195
0.020
0.040
0.0551
0.0356
800
1000
1300
I00
1300
1000
400
200
800
500
400
700
1000
600
1000
DL-Serine
L-Threonine
L-Tryptophan
L-Tyrosine (dissolve in
10% NaOH)
L-Valine
Dextrose
Maltose
MgSO4 7 H20
KCl
NaHC03
NaHzPO4 " H20
TC Yeastolate
Acetyl B-methyl choline
chloride
Cyanocobalamin (B~2)
Isoinositol
Folic acid
CaCI2
Turkey serum
Chicken serum
Calf serum
600
200
100
250
500
5000
1000
1880
2600
350
1160
5000
250
1
10
1.2
50O
3%
3%
3%
462
SPECIFIC
CELL LINES
[39]
TABLE VI
MITSUHASHI AND MARAMOROSCH MEDIUM (g/liter) a
NaX-X2VO4 1-120
MgC12 6 1-120
KCI
CaC12 2 H20
NaC1
NaHCO3
0.20
0.10
0.20
0.20
7.00
0.12
D-glucose
Lactalbumin hydrolysate
Yeastolate
FBS
Penicillin
Streptomycin
4.00
6.50
5.0
20%
100 U/ml
100/,~g/ml
9. Before use, the above basic medium is supplemented with heattreated (60 for 30 min) sera as given in Table V.
MITSUHASHI AND MARAMOROSCH M E D I U M 41
Many mosquito cell lines are grown in this medium (Table VI) which
contains relatively few ingredients. It supports growth of cell lines from
mosquitoes A. aegypti, r A. albopictus, 7 Aedes novalbopictus, 42 A.
taeniorhynchus, 43Aedes w-albus, 44 and Armigeries subalbatus. 43 This medium also is used for cell lines from the leafhopper, Agallia constricta, 45
and the potato tuber moth, Gnorimoschema operculeUa. 4e
PREPARATION OF MITSUHASHI AND MARAMOROSCH M E D I U M 43'47
[39]
INVERTEBRATES
463
TABLE VII
LEIBOVITZ L-15 MEDIUM (mg/liter) a
Amino acids
DL-alpha alanine
L-Arginine (free base)
L-Asparagine
L-Cysteine (free base)
Glycine
L-Histidine (free base)
DL-Isoleucine
(A). L-Leucine
L-Lysine
DL-Methionine
DL-Phenylalanine
L-Serine
DL-Threonine
L-Tryptophane
DL-Valine
(B) L-Glutamine
(C) L-Tyrosine
Salts
[ NaCI
(D) ~! KCI
| MgCI2 6 H20
[ MgSO4 " 7 H20
/
450.0
500.0
250.0
120.0
200.0
250.0
250.0
125.0
75.0
150.0
250.0
200.0
600.0
20.0
200.0
300.0
300.0
8000.0
400.0
200.0
200.0
CaC12
KH2PO4
Na~HPO4
Phenol red
Sodium pyruvate
140.0 (E)
60.0 ]
50.0 / (~
10.01
550.0 (G)
Vitamins
DL-Ca pantothenate
Choline chloride
Folic acid
Inositol
Nicotinamide
Pyridoxine HCI
Riboflavin-5' phosphate
Thiamine monophosphate
Na2HPO4
1.0
1.0
2.0
1.0 ' (H)
1.0
0.1
1.0
140.0
Sugar
Galactose
900.0
FBS
1.0"
10%
a Leibovitz.4S
L E I B O W l T Z L - 1 5 M E D I U M 48
This medium (Table VII), which was originally developed for vertebrate cells, supports cell lines from the hard tick, Rhipicephalus appendiculatus, 49 three other tick species, 5 and the blood-sucking bug,
Triatoma infestans. ~s The medium is usually supplemented with 10% tryptose phosphate broth and 10% FBS. The final pH of L-15 is 7.6 and must
be adjusted to pH 7.0 before use for tick cell lines.
PREPARATION OF L - 1 5 M E D I U M 48
464
[39]
750.0
10.0
10.0
10.0
100.0
100.0
10.0
ml
ml
ml
ml
ml
ml
ml
This medium (Table VIII) is for culturing cell lines from fruit flies D.
melanogaster, 12 Drosophila immigrans, ~ and Drosophila virilis. 5z There
are several other media, not discussed here, that are used for other D.
melanogaster lines.
PREPARATION OF SCHNEIDER M E D I U M 43
[39]
INVERTEBRATES
465
TABLE VIII
SCHNEIDER MEDIUM (mg/100 ml)a'~
Salts and
organic acids
~NaCI
Na2HPO4
KH, PO4
KC1
(A), MgSO4.7 H20
a-Ketoglutaric acid
Succinic acid
Fumaric acid
Malic acid
2 l0
70
45
160
370
20
10
l0
10
Sugars
_. fGlucose
B)'~Trehalose
(C) TC Yeastolate
200
200
200
Amino acids
/fl-Alanine
~ L-Arginine
(D) | L-Aspartic acid
[ L-Cysteine
50
40
40
6
L-Cystine
L-Giutamic acid
L-Glutamine
Glycine
L-Histidine
L-Isoleucine
L-Leucine
L-Lysine HCl
L-Methionine
L-Phenylalanlne
L-Praline
L-Serine
L-Threonine
L-Tryptophan
L-Tyrosine
L-Valine
CaCl2
NaHCOa
l0
80
180
25
40
15
15
165
~(D)
80
15
170
25
35
l0
50
3(
60 (E)
40 (F)
a Schneider.43
b Schneider.51
4. All a m i n o acids (D) e x c e p t c y s t i n e and t y r o s i n e are dissolved in 40
ml water. Dissolve c y s t i n e in 5.0 ml hot acidic w a t e r and t y r o s i n e in 5.0 ml
alkaline water. A f t e r t h e s e t w o amino acids are in solution, t h e y are a d d e d
slowly to the rest o f the a m i n o acid solution.
5. CaClz (E) is dissolved in 5.0 ml water.
6. N a H C O s (F) is dissolved in 4.0 ml water.
7. T h e c o m p o n e n t s in (B), (C), (E), and (F) dissolve quickly. This is
not so with (A) a n d (D) w h i c h require stirring for a b o u t 30 min with a
m a g n e t i c stirrer.
TABLE IX
HANSEN S-301 MEDIUMa
Schneider's medium diluted to 22%
Galactose
1.3 g/liter
Lactalbumin hydrolysate
4.5 g/liter
Fetal bovine serum
13%
a Hansen.~
466
[40]
This medium (Table IX) is for culturing a cell line (Bge) from the snail,
Biomphalaria glabrata. This is the first, established, rapidly growing cell
line from an invertebrate other than the arthropods. The pH is 7.1-7.3,
and osmotic pressure is much lower than insect cell culture media.
E. H a n s e n , in " I n v e r t e b r a t e Tissue Culture: R e s e a r c h Applications" (K. M a r a m o r o s c h ,
ed.), p. 75. A c a d e m i c Press, N e w York, 1976.
[40] C o l d - B l o o d e d V e r t e b r a t e Cell a n d T i s s u e C u l t u r e
By KEN WOLF
Introduction
Methods of cell and tissue culture I range from the very simple to the
highly complex and demanding. In general, many of the methods can be
routine, but cultures are biological systems that can and at times do evade
absolute control. The biochemist planning to use cell cultures will find
that cultures at time show greater variability than the biochemical determinations that are performed.
Most of today's animal cell and tissue culture involves materials of
mammalian and secondarily, of avian origin. Reptiles, amphibians, and
fishes--the poikilotherm vertebrates--are used the least. Without doubt
the present status of homeotherm cell and tissue culture reflects healthrelated research on man and his domestic livestock. Lower vertebrate cell
and tissue culture has much to offer the researcher; it extends the
phylogeny of vertebrates, allows work to be done through a wide range of
temperatures, and is less demanding than culture of homeotherm
materials.
Fortunately for all concerned, the physiology and functional systems
of the foregoing five classes of vertebrates have many similarities. Within
the limits to be described, the methods can be considered realistically as
simply being vertebrate cell and tissue culture.
1 This article e m p l o y s t h e standard terminology o f the Tissue Culture Association as set
forth by S. Federoff, T C A M a n u a l 1, 53 (1975).
[40]
COLD-BLOODED VERTEBRATE
467
468
[40]
the risk of contamination in such material is low. In contrast and for all
practical purposes, the reptiles, amphibians, and all but aquarium or
hatchery-reared fishes are wild animals, and their health history is unknown or uncertain. Such feral animals can harbor systemic parasites,
microorganisms, and viruses that can contaminate primary cell cultures.
On the other hand, there are differences between homeotherms and
poikilotherms that are to the investigator's advantage. For example, cell
cultures from lower vertebrates usually require less frequent handling and
attention than homeotherm cultures, and they are far more tolerant of
neglect. Intrinsically, the coldblooded vertebrates are adapted to and
have a range of body temperatures that cannot be matched by
homeotherms. Some cold-water fish cells metabolize at temperatures
from near 0--25 whereas many warm-water fishes, reptiles, and amphibians span the range of 10-37 . If rigid temperature control need not be
maintained, many kinds of cells are grown on the bench at room temperature, i.e., 20-25 .
Physiological Salines
The usual composition of physiological salines or balanced salt solutions (BSS) for vertebrates is an isotonic salt solution containing the essential physiologic ions, a system for maintaining pH in the physiological
range, an energy source (usually glucose), and, unless contraindicated,
phenol red as a pH indicator. With appropriate adjustment of osmolarity
the common BSS are appropriate for cell cultures of mammals down
through bony fishes (Tables I).
Virtually all routine coldblooded vertebrate cell culture needs will be
met with Earle's or Hanks' BSS and Dulbecco and Vogt's phosphatebuffered saline (PBS). All are available commercially. Without their
sodium bicarbonate, Earle's and Hanks' BSS are also available as
nonsterile powders that can be dissolved in high-purity water and decontaminated by membrane filtration (0.22/zm mean pore diameter). Alternatively, these solutions are easily prepared and decontaminated or
sterilized in one's own laboratory.
Physiological salt solutions find many applications in cell culture.
Earle's BSS with 2.2 g/liter of sodium bicarbonate is intended to equilibrate in the physiological range in an atmosphere containing about 5% COs.
In normal atmosphere, the pH of Earle's BSS rises comparatively
rapidly--the quickest of the three. Hanks' BSS has a phosphate buffer
and much less sodium bicarbonate; therefore its pH changes less rapidly
in normal atmosphere. For the easiest maintenance of pH, PBS is recommended. Physiological salines may be buffered with about 15 mM Tris,
[40]
C O L D - B L O O D E D VERTEBRATE
469
TABLE I
OSMOLARITY ADJUSTMENTS SUGGESTED FOR PHYSIOLOGICAL SALINES AND MEDIA
USED FOR CULTURING LOWER VERTEBRATE CELLS AND TISSUES
Vertebrate
class
Reptile
Amphibian
Teleost
Elasmobranch
Cyclostome
Habitat
Terrestrial or
freshwater
Marine
Terrestrial or
freshwater
Freshwater
Marine
Freshwater
Marine
Freshwater
Marine
Adjustment
None
Add 0.06 M NaC1 a
Dilute 20% with water
or NaCl-free BSS
None
Add 0.07 M NaCI ~
Add urea to isotonic level
Add 2 M urea
None
Not determined
470
[40]
[40]
COLD-BLOODED VERTEBRATE
471
TABLE II
PROXIMATE GUIDELINES FOR INCUBATION TEMPERATURES FOR LOWER VERTEBRATE
CELL AND TISSUE CULTURES
Elasmobranch
Cyclostome
Nearoptimal
Environment
Low
High
Temperate
Tropical
Temperate
Tropical"
Temperate
Cold water
Warm water
Tropical
Temperate
Cold water
Warm water
Temperate
20
not known
15
not known
23-25
30
25-26
28
37
37
37
37
4
13
20
20
25
25
26
30-37
37
4
not known
4
10-15
not known
15
not known
not known
20
472
[40]
II). Cells from donors of a temperate climate are incubated at room temperature and those from tropical latitudes at about 30.
Leukocyte Culture
Poikilotherm vertebrate leukocytes lend themselves to the usual in
vitro applications: karyology, migration inhibition studies, investigations
of antibody production, and other immunologic research. Viable leukocytes are readily obtained from sterile whole blood drawn with either
plastic or siliconized glass syringes containing cold heparin solution in
physiological saline at a final concentration of 10 IU per milliliter of blood.
If one is unfamiliar with the anatomy of a particular animal, it will be
advantageous to dissect a specimen and locate the heart and major blood
vessels and to orient them with external anatomy. Direct cardiac puncture
with a needle of appropriate gauge works well with normally scaled reptiles. Easy access to the heart of terrapins and turtles, of the size conveniently used in laboratories, is gained by first drilling a small hole in the
plastron immediately beneath the heart. Amphibians can be bled from the
heart, and the frogs and toads also can be bled from the femoral artery.
For best results in culturing amphibian leukocytes, the donors should be
kept at normal temperature prior to bleeding; in that way the lymphocyte
population is maintained.
Fishes also can be bled from the heart, but if the animal is to remain
alive cardiac puncture does run the risk of tamponade formation. Fishes
may be bled by opening the mouth and using a shallow approach to enter
the dorsal aorta immediately behind the last gill arch. Fishes are perhaps
bled most safely, most easily, and repeatedly by lateral or ventral approach in the region behind the vent, the caudal peduncle. By the last
method, either the dorsal aorta or the caudal vein will be entered.
When drawn, blood is mixed with the heparin solution and centrifuged
in the cold at 200-500 g or until the "buffy coat" of leukocytes is clearly
visible above the packed erythrocytes. It is convenient to centrifuge the
blood in an inverted but locked syringe. Locks are easily made from split
plastic tubing that fits around the plunger; during centrifugation the tubing
prevents the barrel from moving down and expressing blood. After centrifugation, a needle is refitted, bent to a 90 angle, and the plasma or
"buffy coat", or both, are gently expressed into culture medium.
Obtaining Tissues
Either external or internal tissues can be cultured, but the latter have
the lower risk of contamination. Almost universally, ovarian tissues do
[40]
COLD-BLOODED VERTEBRATE
473
474
[40]
[40]
COLD-BLOODED VERTEBRATE
475
orifice or spatulas are convenient for planting the tissues. Fragments are
uniformly distributed o v e r the growth surface at a rate of two to four
pieces per cm 2. The culture vessels are then closed and placed on edge for
an hour or so at a favorable incubation temperature. After fluids drain
from the tissue and natural adhesion occurs, the liquid is aspirated, the
vessels inverted, medium is added, and the cultures are m o v e d to an
incubator. The vessels are then oriented so that the medium covers the
tissue. Most fragments adhere and many will send out cellular processes
or extend areas o f epithelial-like cell sheet within a day or two. This initial
growth helps the fragments adhere more tightly. After the first growth
occurs the cultures can be examined safely microscopically. Confluent
sheets o f cells may be formed within a few days, but it may take as long as
2 weeks; it depends on donor age, the organ used, pH, temperature, and
other variables. When confluency is attained and growth is obviously
heavy, the cultures may be divided. Requiring only 2 or 3 hr, this method
is undoubtedly the simplest and least time-consuming means o f setting up
primary monolayer c u l t u r e s ) The cultures thus developed are typically
less homogeneous than monolayers that have been initiated from trypsinized tissues.
476
[40]
[40]
COLD-BLOODED VERTEBRATE
477
478
[41]
[41] P l a n t C e l l L i n e s 1
[41]
479
480
[41]
z T. Murashige and F. Skoog, Physiol. Plant. 15, 473 (1962). The salts, in mg/1 are:
NH4NO3, 1650; KNOz, 1900; CaClz. 2 H20, 440; MgSO4.7 HzO, 370; KH2PO4, 170;
Na2EDTA, 37.3; FeSO4 7 H20, 27.8; HzBO3, 6.2; MnSO, H20, 16.9; ZnSO4 7 H20,
8.6; KI, 0.83; Na2MoO4 2 H20, 0.25; CuSO4 ' 5 H~O, 0.025; and CoC12 6 HzO, 0.025.
8 This is agar specially prepared for plant tissue culture by Grand Island Biological Co.,
Grand Island, New York.
9 Examples of plastic caps available are Kimble Kimkap, Bellco Kaput, and Bacti Capalls.
10 Use 9- inch dressing forceps, Model MX6-158, obtainable through Miltex Instrument
Co., New York, New York.
[41]
481
482
[41]
[41]
483
484
[41]
i.e., after 4-5 days of a given passage. ~s Collect cells on sterile Kimwipe
tissue. Transfer 0.5-g portions to 50-ml Delong flasks, each containing 5 ml
of protoplast-release solution.19 Place the suspension on a gyratory shaker
and agitate continuously for 2-3 hr at 50-70 rpm. Filter through nylon
cloth of 50-60/~m. Centrifuge the filtrate at 100 g for 2 min. Decant and
resuspend in 0.7 M mannitol and recentrifuge. Repeat the process of
washing 3 times, by suspending in mannitol and centrifuging. Finally,
suspend the rinsed protoplasts in 2 ml of nutrient medium that has been
prepared earlier and held in a 40 water bath. This medium should contain, in mg/l: Murashige and Skoog salts; sucrose, 15,000; mannitol,
110,000; NAA, 0.6; kinetin, 0.1; thiamine. HCI, 10; pyridoxine. HC1,
10; nicotinic acid, 5; myo-inositol, 100; glycine, 2; and prewashed
Phytagar, 8000. Mix protoplasts and medium gently, but thoroughly, and
pour into 40-mm petri dish by following a procedure similar to cell plating.
Incubate plated protoplasts as done with cells. Cell-wall regeneration will
occur within several hours, and cell division will be observable within a
few days. Transferable cultures will be obtained in 2-4 weeks.
Comments
Some serious misconceptions regarding cultured plant cells need correction. Cultured plant cells are not undifferentiated as often claimed.
They are larger, more vacuolated, lower in cytoplasm content, thicker
walled, and smaller of nuclei than those of the embryo or apical meristem,
where the more truly undifferentiated cells can be found. Very often
phloem and xylem elements may be apparent among them. The cells in
culture may lack organization and are therefore identifiable as being unorganized.
Uniformity also is not the usual trait among cells of callus or liquid
suspension. Obvious variations include shape and size, cytoplasm and
organelle contents, wall thickness, and ergastic deposits. More important,
the cells within a culture may be genetically diverse. For example, variations in chromosome number have not been uncommon. 2 Subculturing,
furthermore, has tended to accentuate the incidence of variants. Retention over long periods of a cell line's characteristics will require employment of cryogenic proceduresY 1
is H. Uchimiya and T. Murashige, Plant Physiol. 54, 936 (1974).
19 Solution containing 1% cellulysin, 0.2% macerase, and 0.7 M mannitol. The enzyme
preparations are obtainable through Calbiochem, La Jolla, California. The pH of the
solution is set at 5-7, and the solution should be filter-sterilized.
20 F. D'Amato, Int. Biol. Programme 2, 333 (1975).
21 y. p. S. Bajaj, Physiol. Plant. 37, 263 (1976).
[41]
485
486
[42]
lution of this problem. The common practice has been simply to set a
nutrient medium's starting pH at between 5 and 6, assuming that this is
within optimum range, and maintaining this pH range during the course of
culture.
Acknowledgments
This work was supported in part by the Elvenia J. Slosson Fellowship in Ornamental
Horticulture and NSF Grant OIP 75-10390 awarded to T. M. We thank S. Hamman and
S. Kearns-Sharp for typing the manuscript.
[42] L y m p h o c y t e s as R e s t i n g Cells
By SHELBY L. BERGER
The human peripheral blood lymphocyte is an ideal system for the
study of both resting and growing cell populations. As they are isolated
from the blood, lymphocytes are quiescent. DNA synthesis measured in
such cells reflects mainly ongoing repair processes since less than one cell
in 5000 is engaged in mitosis.l In v&o, the cells are capable of surviving in
the nondividing state for months or years. ~ In culture, resting lymphocytes can be maintained in excellent condition for several days after
which they deteriorate and die. It should be emphasized that these are
physiologically nongrowing cells; despite the presence of autologous
plasma and a complete medium such as Eagle's minimal essential medium
(MEM) or RPMI-1640, the cells remain in Go, outside the mitotic cycle.
Since most of our knowledge of cellular processes derives either from
continuously dividing cells adapted for cell culture, from density-inhibited
cells, or from arrested cells deprived of essential nutrients, it is essential
to expand our understanding by including the great bulk of normal cells
which resemble none of these examples. The lymphocyte provides such
an opportunity.
When confronted with a stimulus, the resting lymphocyte undergoes
changes in virtually every aspect of cellular metabolism culminating in
DNA synthesis and mitosis. In vivo, the challenge is an immunological
one, and the responding cells are those having specific mechanisms for
antigenic recognition. In vitro, growth induction of a large number of cells
Reviewed by H. L. Cooper, in "Drugs and the Cell Cycle" (A. M. Zimmerman, G. M.
Padilla, and I. L. Cameron, eds.), pp. 137-194. Academic Press, New York, 1973.
2 N. B. Everett and R. W. Tyler, in "Formation and Destruction of Blood Cells"
(T. Greenwalt and G. Jamieson, eds.), pp. 264-283. Lippincott, Philadelphia, Pennsylvania, 1970.
[42]
487
488
[42]
rain at room temperature. On removing the blood pack from the bucket, a
dense layer of erythrocytes can be seen with overlying plasma. The lymphocytes are found as a diffuse band, suspended in the lower region of the
plasma layer. Without disturbing the bands, the unit is carefully hung in
an open plasma extractor by the holes provided in the blood pack and
connected to the 4R2001 Transfer Pack by plugging the attached coupler
into the center port of the blood pack. The blood pack is also connected
by the side port to a receiving vessel, usually a sterile 32-ounce prescription bottle with a foil cover, by means of the 4C2243 Transfer Set. The foil
can be crimped around the coupler to hold it in plfice near the rim of the
bottle. Sterile technique is used throughout, and all steps are performed at
room temperature unless noted otherwise. With the tubing of the transfer
set closed, 50 g of plasma are expressed from the unit into the transfer
pack by releasing the plasma extractor. Then, with the transfer pack
tubing squeezed shut, the remaining plasma and the region of the interphase are expressed into the bottle. To insure complete recovery of the
lymphocyte-rich layer, several milliliters of red cells are expressed into
the bottle as well. Afterwards, 90-100 g of the remaining red cell layer in
the blood pack are added to the transfer pack. As a result of this procedure, most of the lymphocytes are confined to the prescription bottle
while the remainder, which were either trapped in the red cell layer or
suspended in the plasma, are in the transfer pack. Both the blood pack
and the transfer pack are mixed well and centrifuged again under the same
conditions to reextract the plasma-erythrocyte interphase. This step
necessitates removing the transfer set coupler from the receiving bottle,
but not from the side port of the blood pack, and capping it with the
original covering to maintain sterility. Other tubing connections are
clamped but left in place, and the tubing itself is tucked between the two
packs in the bucket during sedimentation. After the centrifugation step the
aim is to combine the lymphocytes in a single container. If plasma is
visible in the blood pack when subsequently squeezed by the plasma
extractor, it is collected into the receiving bottle. The plasma layer and a
few milliliters of erythrocytes from the transfer pack are also expressed
into the prescription bottle. As a result, virtually all the lymphocytes, in
partially purified form, are recovered. To insure against clotting, 5000 U
of heparin (1 ml of Liquaemin Sodium "50 "8) are added at this time.
Many investigators preparing lymphocytes have observed the formation of a "buffy coat" in the discarded blood pack after several hours of
standing. When examined microscopically, this band was found to be rich
in granylocytes and almost devoid of lymphocytes. Thus the preliminary
8 Available from Organon Inc., West Orange, New Jersey.
[42]
489
separation described above removes some, but not all, of the contaminating granulocytic leukocytes.
490
[42]
effect the desired flow rate. A tight-fitting sterile stopper with a sterile
cotton plug is required to couple the pump to the column aseptically.
When the cell suspension has been thoroughly drained or pumped
from the column, it is held at 37 while the nylon is rinsed. Approximately
100 ml of warmed MEM are passed through the column at the same rate of
flow to release trapped cells. The wash solution is reserved in a separate
vessel.
Removal of Platelets
The partially purified cell suspension is freed of platelets by differential
centrifugation at 1000 rpm (200 g) for 10 min. Sterile, plastic conical
centrifuge tubes with caps, holding 50 ml, such as Coming (No. 25335),
Falcon (No. 2074), Kimble (No. 58331), or the equivalent are satisfactory
and can be centrifuged in an HL-8 (Sorvall) or a No. 269 (International)
rotor. The plasma layer, containing platelets, is carefully decanted or
removed with a sterile pipette without mixing the two layers. The deep
red pellet contains the lymphocytes. At a later time the platelets are
separated from the plasma centrifugally (2000--3000 g) in 50-ml sterile
tubes at 20 for 15-20 rain. Although room temperature should be acceptable for this process, in practice, the plastic tubes become deformed at
high speeds in a warm centrifuge. The supernatant fraction, composed of
cell-free plasma, is filtered through a Nalge, 0.45-/zm filter9 to insure
sterility and retained for use as a supplement in the final culture medium.
[42]
491
The dense red cells penetrate the LSM layer and sediment to the bottom.
Lymphocytes band at the interphase while granulocytes, if present, are
found as a whitish layer immediately above the erythrocytes. Contaminating platelets band with lymphocytes.
The lymphocyte layer is recovered with a pipette and separated from
LSM by centrifugation at 1500 rpm for 15 min. The final purified preparation is resuspended in 30 ml of MEM and combined in a single tube. The
entire procedure takes 4 hr, once the blood is drawn.
YieM
For counting, approximately 0.2 ml are withdrawn to a spot plate, and
an aliquot is diluted 20-fold with 0.02% crystal violet in 1% acetic acid. A
white blood cell diluting pipette is convenient for this purpose, and a
hemocytometer is required for quantitation.
The yield from one unit of blood (1 pint; 500 ml) is approximately
4-12 x 108 small lymphocytes. Immediately after purification polymorphonuclear leukocytes account for 0--2% of the total purified leukocyte
population. However, these contaminating cells do not survive well in
culture and are gradually eliminated during incubation. An occasional
monocyte is evident on microscopic examination; such cells are essential
for mitogen stimulation of lymph node lymphocytes from guinea pigs TM
and are believed to be necessary for activating other types of lymphocytes
as well.
492
[42]
Impurities
The major contaminants in lymphocyte preparations are platelets and
erythrocytes. Both are destroyed by the dilute acetic acid in the staining
solution used for counting the cells but can be viewed microscopically in
unstained samples. On the average, one erythrocyte is seen per 20 lymphocytes after incubating the cells overnight.
Platelet contamination is more difficult to assess. In this laboratory,
platelets account for 8-25% of the packed cell volume. However, such
contamination can be reduced by repeating the centrifugation step in
100% plasma. For best results the cell pellets, containing predominantly
lymphocytes and erythrocytes, should be resuspended in a small amount
of medium while the recovered plasma is cleared of platelets by sedimentation. When the plasma is ready, the impure lymphocytes are mixed with
~4 For a more complete list of mitogens and their properties, see B. A. Cunningham, B.-A.
Sela, I. Yahara, and G. M. Edelman, in "Mitogens in Immunobioiogy" (J. J. Oppenheim
and D. L. Rosenstreich, eds.), pp. 13-30. Academic Press, New York, 1975, or see
N. Sharon, ibid. pp. 31-41.
15 j. L. Bernheim and J. Mendelsohn, in "Regulatory Mechanisms in Lymphocyte Activation" (D. O. Lucas, ed.), pp. 479-505. Academic Press, New York, 1977.
[42]
493
the plasma and separated from remaining platelets by differential centrifugation as indicated above.
In the procedure detailed by Cooper, 5 platelets are removed by
agglutination with adenosine diphosphate.16 This method is capricious in
its usefulness; it ranges from highly effective to completely ineffective
depending on the donor. If platelet-free cultures are essential, defibrination with glass beads is recommended. Although some loss of lymphocytes from clotted blood cannot be avoided, the fibrin clots trap and bind
virtually all of the platelets, lr
Comments
The inconvenient and tedious preparation of nylon can be circumvented by purchasing this material from Fenwall. The Leuko-Pak (No.
4C2401) contains sufficient teased nylon for 3-4 units of blood. There are
two disadvantages: processed nylon is considerably more expensive than
the cruder product from Dupont, and one becomes dependent on the
availability of Leuko-Paks, which can be out of stock for months.
The separation of erythrocytes by LSM solution has been adopted as a
convenience. The Ficoll-Hypaque method as described by Perper et al. 18
and used by Coope# is equally satisfactory.
The composition of the lymphocytes produced by nylon-column filtration has been a matter of conjecture. It has been suggested that nylon may
deplete the population of B cells (bone marrow-derived lymphocytes)
leaving predominantly T cells (thymus-derived lymphocytes). When the
distribution of the two types of lymphocytes was measured by rosetting
with sheep erythrocytes treated with S-2-aminoethylisothiouronium
bromide hydrobromide, TM approximately 85% were nominally T cells in
our preparations. Studies of the composition of lymphocytes purified
without recourse to nylon gave similar results. 19 There is therefore no
evidence for depletion of B cells during the brief exposure to nylon which
forms part of this procedure for purifying lymphocytes.
A major disadvantage of the human peripheral blood lymphocyte in
research is the difficulty of obtaining large amounts of material. For most
purposes, pooled cells are unsatisfactory even if mitogen treatment is
anticipated. This problem is solved in part by leukophoresis, an alternate
16 A. Gaarder, J. Johnsen, S. Laland, A. Hellem, and P. A. Owren, Nature (London) 192,
531-532 (1961).
17 S. Niewiarowski, E. Regoeczi, G. J. Stewart, A. Senyi, and J. F. Mustard, J. Clin. Invest.
51, 685-700 (1972).
18 R. J. Perper, T. W. Zee, and M. M. Mickelson, J. Lab. Clin. Med. 72, 842-848 (1968).
19 M. E. Kaplan and C. Clark, J. lmmunol. Methods 5, 131-135 (1974).
494
[43]
yielding large numbers of white cells which must then be purified as described. However, autologous plasma sufficient for culturing the entire
yield is not provided so that pooled AB serum must be used as a substitute. The effect of foreign substances in serum on the cellular biochemistry of cultured cells has not been fully investigated.
[43] M a c r o p h a g e s
By DOLPH O. ADAMS
Macrophages are currently studied in vitro in almost all fields of biology. The mononuclear phagocyte system, which includes macrophages, is
a host-wide system of phagocytic cells with similar properties. 1 Mononuclear phagocytes arise in the marrow, circulate briefly in the blood as
monocytes, and immigrate into the tissues and inflammatory foci where
they mature into macrophages. Macrophages can be further stimulated to
develop altered function and metabolic characteristics--a state often
termed activation. Activation in this broad sense can be induced by many
stimuli and is characterized by the presence of certain markers 2 as well as
by increases in size, adherence, secretory capacity, content of lysosomes,
and various functions such as phagocytosis and chemotaxis. 1 Since most
activated states are not identical functionally, activated macrophages
should be identified by both the eliciting stimulant and the altered capacity
tested.
Macrophages as cultivated cells offer the following advantages: They
are easy to obtain, can be cultured in relatively pure form, are primary
cultures, and are available in numbers sufficient for analytical biochemical
manipulations. On the other hand, macrophages do not generally replicate
in culture, are relatively short-lived, and may be difficult to obtain in
numbers sufficient for preparative biochemistry, It is important to note
that macrophages also are very sensitive to small changes in their environment and are thereby modified considerably from their native state in
vivo, even when delicately handled and observed after very short periods
of culture.
1 D. O. Adams, Am. J. Pathol. 84, 163 (1976).
z p. Edelson and Z. A. Cohn, in " I n Vitro Methods in Cell Mediated and Tumor Immunity"
(B. R. Bloom and J. R. David, eds.), pp. 333-340. Academic Press, New York, 1976.
[43]
MACROPHAGES
495
Sources of Macrophages
General
Macrophages can be obtained from blood, lung, spleen, liver, and the
peritoneal cavity, t2-s} However, mononuclear phagocytes from most of
these sources must be used in short-term experiments since they do not
survive much more than 24-48 hr in culture. Alveolar macrophages from
all sources and peritoneal macrophages from small rodents other than
mice are particularly difficult to maintain. The spleen does not yield many
macrophages, and those obtained are often contaminated by other adherent cells. Hepatic macrophages are considerably altered by the techniques necessary for their isolation. Consequently, peripheral blood
monocytes and peritoneal macrophages are the commonly employed
sources of mononuclear phagocytes.
Monocytes
Monocytes are generally obtained from humans, but equine monocytes have been studied, z Monocytes are separated from the blood by
differential centrifugation and subsequent adherence to culture vessels.
Approximately 108 monocytes can be obtained from 500 ml of blood. This
represents the most readily accessible source of human mononuclear
phagocytes. However, these cells are extensively manipulated before
plating, are difficult to culture, and may be contaminated with large numbers of platelets. A detailed protocol for the culture for human monocytes
is available. 2
Peritoneal Macrophages
The peritoneal cavity offers a ready source of mononuclear phagocytes. The unstimulated peritoneal cavity of mice contains usable numbers (2-3 108) of resident macrophages. These may be washed out directly and placed into culture. The resident cells should not possess any of
the characteristics of inflammatory or activated macrophages. 2 If they do,
the likelihood that the mice are infected should be strongly considered.
3 Z. A. Cohn, this series, Vol. 32, pp. 758-765.
4 G. D. Wasley and R. John, in "Animal Tissue Cultures" (G. D. Wasley, ed.), pp. 101-137.
Butterworth, London, 1972.
5 A. E. Stuart, J. Habeshaw, and A. E. Davison, in "Handbook of Experimental Immunology" (D. M. Weir, ed.), pp. 24.1-24.36. Blackwell, Oxford, 1973.
496
[43]
Alveolar Macrophages
Alveolar macrophages can be obtained from guinea pigs, rabbits, and
humans, z In the former two circumstances, 20 106 macrophages can
normally be lavaged from the lungs. Up to 20 x 108 macrophages are
obtainable if the animals are given Freund's adjuvant or avirulent tubercle
bacilli intravenously 3-4 weeks before harvest. Human alveolar mac-
6 S. Gordon, Z. Werb, and Z. A. Cohn in "In Vitro Methods in Cell Mediated and Tumor
Immunity" (B. R. Bloom and J. R. David, eds.), pp. 341-352. Academic Press, New
York, 1976.
r T. P. Stossel and Z. A. Cohn, Methods lmmunol, lmmunochem. 5, 261-292 (1976).
[43]
MACROPHAGES
497
rophages can be obtained by direct bronchial lavage of both normal volunteers and patients. 8
Separation and Purification of Macrophages
Only techniques for enrichment of macrophages will be described here.
Procedures for depletion of mononuclear phagocytes are available elsewhere. 9 Enriched populations of macrophages generally are obtained by
centrifugation, adherence, or combinations of the two.
Centrifugation
A fraction of mononuclear leukocytes containing both lymphocytes
and monocytes can be separated from other leukocytes and from red cells
by centrifuging in albumin or in a gradient of Ficoll-Hypaque. 2 Contaminating platelets are then partially removed from the mononuclear
fraction by differential centrifugation. This technique, which is necessary
to obtain monocytes, may injure or alter the cells due to the extensive
separation procedure.
Adherence
Most mononuclear phagocytes, particularly mature or elicited ones,
adhere avidly to culture vessels. 1 This property is effectively exploited in
purification by adding populations of leukocytes to culture vessels and
incubating them in medium containing serum for several hours. The
nonadherent cells are removed, and the adherent cells are washed. The
washings must be vigorous to dislarge other adherent leukocytes such as
neutrophils and stimulated T cells. The adherent cell population can then
be trypsinized to remove fibroblasts or tumor cells. 11 The remaining cells
should be macrophages of high purity (95% or greater). The proportion of
the cell population that is macrophages is determined routinely as described below.
Adherence does possess certain disadvantages. Some of the mononuclear phagocytes will not adhere in several hours and may be lost. However, cultures may be continued without washing for 24 hr, which permits
a larger number of the mononuclear phagocytes to adhere. This interval
also results in death and disintegration of the neutrophils, further con8 D.
K.
10 M.
11 R.
498
[43]
Phagocytic Uptake
Extensive phagocytic uptake by a mononuclear cell population is clear
evidence of a lineage in the mononuclear phagocyte system. Procedures
for determining phagocytic uptake are well detailed. 7 A test particle such
as starch, latex beads, or erythrocytes plus an opsonin (fresh serum or
specific antibody) are incubated for 30-60 rain at 37 with the mac1~ L. T. Yam, C. Y. Li, and W. H. Crosby, Am. J. Clin. Pathol. 55, 283 (1971).
[43]
MACROPHAGES
499
rophages. The cells are then washed and the percentage of mononuclear
cells containing five or more particles within the cytoplasm (not adherent to
the cells) is determined. Lab-Tek chambers (Lab-Tek Products, Napierville, Illinois) or coverslips suspended over slides with a depressed well
(Arthur H. Thomas, cat. no. 6688-D20) are particularly convenient for this
purpose.
Markers
Mononuclear phagocytes are distinguished by the presence of surface
receptors for both C3h and Fc. 13,14These may be demonstrated by use of
appropriately coated red cells. Detailed protocols for these procedures
are available) T M
Culture Techniques
The selection of appropriate culture conditions for macrophages depends upon the intent of the particular experiment and usually represents
a balance between multiple factors. First, basal media are less expensive
and will support viability of the macrophages but may not permit full
expression of all functional capabilities. Second, macrophages are easily
stimulated by alterations in their environment such as enriched media,
high concentrations of serum, trace concentrations of endotoxin, or
phagocytosis of other cells. Such endogenous stimuli can obscure differences between experimental and control cultures. Third, the inherent inconsistencies of tissue culture must be considered. In our hands, for example, Dulbecco's minimum essential medium (MEM) supports accumulation of acid hydrolases much better than does Eagle's MEM, whereas
the reverse is true for the expression of nonspecific cytotoxicity.
General Requirements
Macrophages usually are cultured in moist air containing 5% COz.
Precise regulation of CO2 content with a controller may be advantageous
for critical experiments. Acidity is common with cultured macrophages,
and pH should be checked daily. The pH can be adjusted to 7.2 by
addition of sterile sodium bicarbonate (sterilized by filtration).
Plastic vessels coated for tissue culture are generally suitable for
13 j. Michl, D. J. Ohlbaum, and S. C. Silverstein, J. Exp. Med. 144, 1465 (1977).
14 E. M. Shevach, E. S. Jatfe, and I. Green, Transplant. Rev. 16, 3 (1973).
500
[43]
Media
Medium 199 and Eagle's minimal essential medium are useful basal
media. These are generally supplemented with streptomycin, penicillin,
and fresh glutamine. HEPES buffer, 5-10 mM, aids in stabilizing pH but
may be toxic to some cultures.
NCTC-135, MEM alpha, RPMI-1640, and Ham's FI2 are richer media
that have been successfully employed in culturing macrophages.
Basal media can be selectively enriched by addition of a variety of
substances including sodium pyruvate (0.11 mg/ml), ascorbic acid (50
/zg/ml), other vitamins, and nonessential amino acids.~5
Serum-free medium may be necessary for some experiments. A variety of supplements have been tried; we have had the most success with
lactalbumin hydrolysate (Grand Island Biological Co.). The cell-free medium of Newman-Tytell is particularly useful, especially when supplemented with 10 mM HEPES. We have recently begun using the supplement described by Guilbert and Iscove of selenite, transferrin, lecithin,
and albumin TM and have found that it improves the viability and maturation
of macrophages when added to serum-free cultures.
15 C. W a y m o u t h , Int. Rev. Cytol. 3, 1 (1954).
an L. J. Guilbert and N. N. I s c o v e , Nature (London) 263, 594 (1976).
[43]
MACROPHAGES
501
Serum
Most cultures of macrophages are supported by 10-40% serum. Fetal
calf serum is routinely employed, but equine and newborn calf serum also
have been used. Newborn calf serum may contain antibodies that stimulate maturation of the macrophages. 3 Human macrophages usually are
cultured with human serum, preferably autologous. For murine macrophages, we routinely used 10% heat-inactivated fetal calf serum.
Serum can vary considerably from lot to lot in its ability to support
both survival and function of macrophages. For critical experiments,
serum may have to be screened by testing the ability of various lots to
support the function being tested. Furthermore, certain components of
serum are apparently labile. In critical experiments, we obtain frozen
serum shipped in Dry Ice from a previously screened lot. All is gently
thawed at 37, heat-inactivated with constant swirling for 30 min at 56,
apportioned into small aliquots, and stored at - 2 0 . On the day an experiment begins, one container of serum is gently thawed and used. Any left
over serum is not reused for an experiment, not even the next day, but is
saved for routine tissue culture.
Detachment of Macrophages
Detaching macrophages that have already adhered to a culture vessel
particularly if the macrophages are activated, is extremely difficult.
Neither trypsin, cold, nor chelating agents remove macrophages well, and
it is generally best to conduct experimental manipulations in the culture
vessel used for purification. Gentle scraping of macrophage cultures with
a rubber policeman will remove the cells, approximately 1/z of which may
be viable. Macrophages may be detached partially by use of a local anesthetic, a7 Purified Lidocaine, without preservatives (Astra Pharmaceuticals, Worchester, Massachusetts) is made to 360 mM in phosphatebuffered saline (PBS) and adjusted to pH 6.6 with 1 M NaOH. The stock is
diluted in medium to 12 mM, and macrophages are incubated in the
medium containing Lidocaine for 5 min at 37. The macrophages should
then appear rounded and are easier to remove, although extensive losses
may be incurred.
502
[43]
For study of macrophages in exudates or suspension cultures, an excellent method is to prepare them in a Cytocentrifuge (Shandon Southern
Instrument Co., Sewickeley, Pennsylvania) and stain the resultant
smears with a commercially available Wright's stain (Diffquik, Arthur
Thomas Co.). This stain is also convenient for macrophages cultivated on
coverslips or in Lab-Tek chambers.
Phase microscopy is extremely useful for more detailed examinations.
Macrophages are particularly advantageous to study by this method,
since they spread after adherence and thus reveal their cytoplasmic contents in the thinned cytoplasm. Cultures of spread macrophages on coverslips are washed in PBS and fixed 5 rain in 4% osmium tetroxide in
S-Collidine buffer while in a f u m e hood. The coverslips are immersed over
PBS in a trough whose shallow walls of silicone grease are applied to a
glass slide with a syringe and needle. The resultant inverted wet mount is
studied under the phase microscope and can be preserved by coating it
with clear nail polish.
Biochemical Preparation
[43]
MACROPHAGES
503
rophages can remain intact after multiple cycles of freezing and thawing.
We generally employ the detergent Triton X-100. We add ice-cold PBS
containing 0.2% Triton X-100 to washed cultures of macrophages and
incubate the cultures with the detergent for 30 min. The entire procedure
must be conducted on ice to prevent liberated lysosomal enzymes from
destroying the desired cellular constituent. After the incubation, macrophages are completely removed from the culture vessel by scraping
with a rubber policeman. The preparation of organelles from macrophages has been described in detail. 2
The study of products secreted by macrophages is conducted on medium conditioned by growing macrophages in it. 6 Since many secreted
products, e.g., neutral proteases, are inhibited by serum constituents, the
cultures are frequently conducted in serum-free medium. Siliconized
glassware is useful because many of the secreted products are extremely
labile and adhere to glass. Daily, the cultures of macrophages are aspirated and the aspirates placed in siliconized conical centrifuge tubes on ice
and immediately spun 10 min at 10,000 g at 4. If the desired secretory
product is present in low concentration, the conditioned medium can be
lyophilized or dialized against an adsorbent such as Acquacide II (Calbiochem, San Diego, California) to concentrate the constituent. In our
hands, concentration in a stirred cell against an appropriately sized UM
membrane (Amicon Inc., Lexington, Massachusetts) has proven the most
useful and satisfactory method.
Specific activity of a given constituent within macrophages can be
expressed in relation to either protein or cell number. Determination of
cell number is generally advantageous because cultivated macrophages
usually produce large amounts of protein in culture and thus specific
activity may fall in the face of a rising total amount of a particular enzyme. 21 Determining cell number by hemocytometer count is often inaccurate because a large but variable number of macrophages is lysed while
scraping them from the culture vessel. Determination of DNA content
provides an accurate, reliable, and sensitive method for quantifying cell
number. 2z
Continuous Cell Lines
Several lines of continuously replicating cells having the properties of
macrophages are now available (for review, see Defendi2Z). These neon0z. Werb and Z. A. Cohn, this series, Vol. 31, P. A, pp. 339-345.
21 Z. A. Cohn and B. Benson,J. Exp. Med. 121, 153 (1968).
z2 S. Cooksonand D. O. Adams,J. lmmunol. Methods (1978) in press.
2s V. Defendi, in "Immunobiologyof the Macrophage" (D. S. Nelson, ed.), pp. 275-286.
Academic Press, New York, 1976.
504
[43]
plastic murine lines have the morphology of macrophages and are phagocytic. Depending on the particular line, these cells may express various
other functions generally associated with macrophages. The macrophagelike lines are generally easy to grow and are passaged without difficulty,
since they detach readily from the culture vessels. They offer the obvious
advantage of providing large numbers of similar cells with minimal
difficulty.
Appendix: Protocol for Culturing Stimulated Macrophages
Animals
Two C57 B 1/6J mice of either sex (Jackson Laboratories, Bar Harbor,
Maine) weighing 25-30 g each are used.
Supplies
Small, lidded plastic bucket, containing one-haft pound of Dry Ice
Small dissecting board, covered with sterile barrier
70% ethanol
Two small forceps with teeth
One 3 inch dissecting scissors
Sterile, plugged, Pasteur pipettes with rubber bulbs
Two sterile 23-gauge 1 inch needles
Two sterile 10-ml syringes
One sterile 50-ml conical polypropylene centrifuge tube with cap
30 ml of ice-cold washout medium [Eagle's minimal essential medium
with Earle's salts (Grand Island Biological Co., Grand Island, New
York, cat. no. F-11); to this, add 10 units of heparin per milliliter]
Ice bucket filled with ice
3 cluster dishes with four 60-mm wells of tissue culture plastic
(Falcon Plastics, Los Angeles, California)
250 ml of sterile Eagle's MEM with Earle's salts to wash plates
Hemocytometer
Gilson or similar pipettes, dispensing 20 and 200/A
10 ml sterile plugged pipettes
Glass slides
Coverslips
2 ml of sterile, Brewer's thioglycolate broth (Difco Manufacturing
Co., Detroit, Michigan, cat. no. B-236; prepare and store
according to manufacturer's instructions)
[43]
MACROPHAGES
505
Collection of PEC
Lay out all equipment. Load two syringes with 10 ml of chilled washout medium and place them and the centrifuge tube on ice. Note that the
entire collection and culture procedure is to be done under sterile
conditions.
To kill the mouse, place it in the plastic bucket containing the Dry Ice.
Suspend it over the ice by its tail placed under the rim of the closed lid.
After 1 min, remove the dead mouse.
Wash the skin of the abdomen of the mouse vigorously with 70%
ethanol. Gently pick up the abdominal skin with a pair of toothed forceps
in each hand and gently manipulate the skin up and down to separate it
from the peritoneal wall. Grasp the skin with both forceps and gently tear
it, making sure not to tear the peritoneal wall. Pull the torn skin caudally
and rostrally until the carcass of the mouse is completely exposed. Do not
tear the peritoneal wall or the thorax.
Rewash the peritoneal wall with 70% alcohol. Insert the needle into
the peritoneal cavity in the midline and inject the 10 ml of washout medium. Massage the flanks of the animal for several seconds. Elevate theI
shaft of the needle to create a small tent just below the xyphoid process
and aspirate the injected fluid. At least 9 ml should be recovered.
Remove the needle from the syringe and gently introduce the collected
fluid into the centrifuge tube. Cover the centrifuge tube. Wash the second
mouse similarly. Add the fluid from the second mouse to that of the first
and mix gently. Remove a few drops of the fluid and prepare smears on
the Cytocentrifuge, keeping the centrifuge tube on ice.
506
[44]
Culture of Macrophages
Immediately centrifuge the collected peritoneal fluid for 10 rain at 250g
at 4. Carefully aspirate the supernant from the resultant cell pellet. Add
10 ml of complete tissue culture medium. Gently disperse the cell button
with a cotton-plugged sterile Pasteur pipette and take a small sample.
Return the capped centrifuge tube to the ice bucket.
Determine the cell number in a hemocytometer, using the Gilson
pipettes for dilution and counting at least 400 cells. Examine the stained
cytocentrifuge smear and do a differential count. Typically, each mouse
should yield 20-30 10n PEC, of which approximately 90% should be
large macrophages. Calculate the final dilution of the PEC. To get 3 105
adherent macrophages/cm 2, a concentration of 2.5 x 10n PEC/ml in 6 ml
will be added to each 30 cm 2 well. (2.5 10n PEC/ml 90% macrophages
~ of elicited macrophages to adhere 6 ml + 30 cm z = 3 105 adherent
macrophages/cm2).
Make this dilution and add 6 ml of the suspension to each well. Shake
the dishes laterally to disperse the cells evenly and incubate the dishes for
3 hr in a humidified CO2 incubator at 37. Aspirate the medium and wash
each well vigorously 3 times with tissue culture medium containing no
serum or supplements. The cultures at this time should consist of
monolayers of well-spread phase-dense macrophages having prominent
cytoplasmic ruffles. Add 6 ml of complete culture medium to each well
and return to the incubator.
Acknowledgments
This work was supported in part by U.S.P.H.S. (}rants CA-14236 and CA-16784.
[44] M o u s e E r y t h r o l e u k e m i a C e l l s
By T. V. GOPALAKRISHNANand W. FRENCH ANDERSON
Mouse erythroleukemia (MEL) or Friend cells grow in suspension
culture and have been extensively used as a model system for studying
erythropoiesis in vitro.l,2 When MEL cells are grown for several days in
the presence of dimethylsulfoxide (DMSO) or a variety of other chemical
agents, 3 they undergo changes similar to the normal maturation of red
1 C. Friend, H. D. Preisler, and W. Scher, Top. DeF. Biol. 8, 81 (1974).
2 p. R. Harrison, Int. Rev. Biochem. 15, 227 (1977).
3 R. C. Reuben, R. L. Wife, R. Breslow, R. A. Rifkind, and P. A. Marks,Proc. Natl. Acad.
Sci. U.S.A. 73, 862 (1976).
[44]
MOUSE ERYTHROLEUKEMIA
507
4 c. Friend, W. Scher, J. G. Holland, and T. Sato, Proc. Natl. Acad. Sci. U.S.A. 68, 378
(1971).
5 D. Kabat, C. C. Sherton, L. H. Evans, R. Bigley, and D. Koler, Cell 5, 331 (1975).
6 p. S. Ebert and Y. Ikawa, Proc. Soc. Exp. Biol. Med. 146, 601 (1974).
r H. Eisen, R. Bach, and R. Emery, Proc. Natl. Acad. Sci. U.S.A. 74, 3898 (1977).
8 y. Ikawa, M. Furusawa, and H. Sugano, Bibl. Haematol. (Basel) 39, 955 (1973).
9 A. W. Nienhuis, J. E. Barker, and W. F. Anderson, in "Kidney Hormones" (J. W.
Fischer, ed.), p. 245. Academic Press, New York, 1978.
10A. W. Nienhuis, J. E. Barker, A. Deisseroth, and W. F. Anderson, Ciba Found. Symp. 37,
329 (New Ser.), (1976).
11T. V. Gopalakrishnan, E. B. Thompson, and W. F. Anderson, Proc. Natl. Acad. Sci.
U.S.A. 74, 1642 (1977).
12D. E. Axelrod, T. V. Gopalakrishnan, M. Willing, and W. F. Anderson, Somatic Cell
Genetics, 4, 152 (1978).
~aj. Ross, Y. Ikawa, and P. Ledcr, Proc. Natl. Acad. Sci. U.S.A. 69, 3620 (1975).
508
[44]
obtained as primaries by following the procedure of Friend. l Briefly, susceptile mice are infected with Friend virus complex from which they
develop an erythroleukemia syndrome. Cells from the enlarged spleen are
then inoculated subcutaneously into another susceptible mouse and a
tumor resembling a reticulum cell sarcoma develops. Cells from this
tumor can be cultured indefinitely in suspension as MEL cells.
B. Growth Conditions. MEL cells are grown at 37 in a humidified
water-jacketed incubator equilibrated with 10% COz in air. Growth at 37
is not greatly influenced by slight variations in the CO2 concentration.
Since the CO2 content inside the incubator determines the pH of the
medium (which is buffered with NaHCOz), too much or too little COz will
lower or raise the pH and be lethal for the cells. A CO2 concentration that
lies between 5-10% maintains the pH of any one of the media described
below in a range that is conducive to good cell growth.
C. Medium. MEL cells are easily grown as a suspension culture in any
of the established media supplemented with 10% fetal calf serum
(GIBCO). Routinely these cells have been grown in BME (Basal minimal
medium, GIBCO) or in slightly richer media such as Modified Improved
Minimal Essential Medium (Modified IMEM) or F14 medium (available
from GIBCO as F12 modified medium 72004 and then supplemented with
3.7 g/liter of NaHCO3). Depending upon the medium and growth conditions, MEL cells grow with a doubling time of 10-18 hr. It is important to
maintain the cells in logarithmic growth. This is achieved by diluting the
cells 1:50 twice a week; slower growing cells can be passed at 1:100
dilution once a week. These dilutions are useful for the maintenance of
stock cultures, whereas cells to be used regularly for experiments can be
maintained at 1:10 dilution at frequent intervals.
D. Serum. Fetal calf serum is used with medium at a concentration of
10% (v/v). Several different lots of serum should be tested for their ability
to promote optimal growth as well as for hemoglobin induction of the
MEL cells. Markedly different doubling times and percent inducible cells
are obtained with different lots of serum from the same (or different)
companies.
[44]
MOUSE ERYTHROLEUKEMIA
509
510
[44]
Hemoglobin Induction
For induction experiments, cells are plated in regular growth medium
at a density of 1 105 cells/ml with 1.5-2% (v/v) DMSO. ~4 After 4 or 5
days, the cell pellet appears pink to deep red depending upon the extent of
hemoglobin production. For the quantitative estimation of the percentage
of cells producing hemoglobin, the cells are stained with benzidine as
described below.
DMSO should not be autoclaved for sterility since this inhibits induction of hemoglobin in MEL cells. Hence, a stock solution of 15-20%
DMSO in growth medium is prepared and then sterilized by filtration
through ~algene filters with 0.45-/~m pores. The stock solution can be
stored at 4 . This solution is diluted 1:10 with medium so as to bring the
final concentration of DMSO to 1.5-2.0~. In addition to DMSO, a variety
of other chemical agents bring about the induction of hemoglobin in MEL
cells, a Of these inducers, hexamethylenebisacetamide has proved to be
the most potent inducer for hemoglobin.
Benzidine Staining
A. Stock Solution. The stock solution is 0.2% (w/v) benzidine dihydrochloride in 0.5 M acetic acid. The solution is stable for many months
when stored in a b r o w n bottle and kept refrigerated. Note: ~ i s is a potential carcinogen and, therefore, should be handled using proper precautions.
B. Stain Solution. A fresh solution of 0.4% of 30% H~O2 (v/v) in the
benzidine stock solution (i.e., 10 microliters 30% H~O~ in 2.5 ml stock
solution) is prepared just prior to use. One-tenth volume of the stain
solution is added to a cell suspension that is to be tested for hemoglobin
induction. Routinely we use 4 ml of a suspension of M E L cells that have
been treated with 2% DMSO for 5-6 days; 0.4 ml of the stain solution is
added and mixed. The cells that contain hemoglobin turn blue in 5-10
min. The percentage of benzidine-positive cells is determined by brightfield light microscopy; 200-500 cells are counted for an accurate estimation. It is essential to examine cells in different areas of the dish to insure
that the distribution of benzidine-positive cells is uniform. A potential
source of error arises from the tendency of the floating population of cells
to concentrate at the center of the dish. One way of overcoming this
problem is to mix the cell population very well after the addition of the
stain solution. We have also noted that the benzidine-positive cells have a
tendency to float and the negative cells to sink. This should be taken into
account during counting.
14Obtained from Fisher, Fisher Scientific Co., Pittsburgh, Pa. Since different lots of
DMSO also differ in their ability to bring about hemoglobin induction, DMSO should be
lot tested also.
[45]
SKELETAL MYOBLASTS IN C U L T U R E
511
[45] S k e l e t a l M y o b l a s t s in C u l t u r e
By IRWIN R. KONIGSBERG
Cell culture systems have been employed for the past 17 years 1"2 to
study the differentiation of skeletal muscle fibers from myoblasts, the
embryonic stem cells from which this tissue develops in vivo. When appropriately cultured, these progenitor cells reproduce, with remarkable
fidelity, the sequence of events that have been observed during normal
embryonic development. Cell culture techniques provide a number of
advantages unattainable in the intact organism. One can obtain a uniform,
highly purified population of cells of known developmental fate. These
can be maintained under rigidly controlled conditions and subjected to a
wide range of experimental intervention. Finally one can manipulate culture conditions to impose a greater degree of synchrony than is ever
observed in the organism.
The use of skeletal muscle cell culture as an experimental system is
now widely employed. Although the same overall strategy is used, the
details of the specific techniques used vary widely from one laboratory to
another. At times, therefore, it becomes difficult to compare the results
obtained by different investigators. This article will deal largely with the
techniques and procedures currently in use in our laboratory. Where
1 L. M. Rinaldini, Exp. Cell Res. 16, 477 (1959).
2 I. R. K o n i g s b e r g , Exp. Cell Res. 2 1 , 4 1 4 - 4 2 0 (1960); I. R. K o n i g s b e r g et al., J. Biophys.
Biochem. Cytol. 8, 333 (1960).
512
[45]
[45]
513
within 1 hr. Embryos are removed sterilely, one at a time, and transferred
to a 100-mm glass petri plate. Skin and other overlying tissues are removed and discarded and the muscle cleanly dissected and transferred to
a 50-mm petri dish containing a drop or two of a phosphate-buffered balanced salt solution. The pooled muscle is then thoroughly minced with a
pair of sharp curved scissors. During this procedure the tissue is confined
to one edge of the dish by tilting the dish and using the edge of the scissors
to push the mince together. This should be done quickly to avoid desiccation. One quickly learns to gauge the proper degree of mincing (by counting scissor strokes and observing the degree of homogeneity of the
mince). If the tissue is not adequately minced this will become obvious at
the next step, which requires diluting and pipetting themince to a tube (or
flask).
Cell Dissociation (Mechanical)
514
[45]
Fig. 1. Cell filter. The device centers a cotton-plugged polypropylene test tube (Nalge
#3110-0180), with the bottom machined off, inverted in a 40-ml Pyrex centrifuge tube with a
#5 silicone rubber stopper. The centrifuge tube is vented by the insertion of a 20-gauge
hypodermic needle. Nitex or gauze filters (see text) are tightly tied to the flared lip of the
inverted tube and discarded after use.
[45]
515
516
[45]
counts are more readily obtained using enzymically dissociated cells, the
collagenase procedure previously used by us is given here:
1. Muscle tissue is dissected and minced as described above (see
"Dissection and Mincing the Tissue") using two or four embryos.
2. The mince is transferred with two 2.5-ml portions of nominally
0.1% collagenase in Puck's saline G ~8into a 25-ml Erlenmeyer flask using
a serological pipette with an orifice adequate to accept the minced fragments (usually 10 ml).
3. The suspended tissue is maintained at 37 for 5 min and subjected to
gentle pipetting with a 10-ml serological pipette to disperse the cells.
4. Enzymic digestion is stopped by adding 5 ml of complete growth
medium at about 5, and the suspension is filtered through three double
layers of cheesecloth (see Fig. 1) into a 40-ml Pyrex conical centrifuge
tube.
5. The filtered suspension is centrifuged at 800 rpm with a bench-top
centrifuge (I.E.C., Model H) and the supernatant liquid removed by
aspiration.
6. The cell pellet is dispersed in 2-5 ml of complete growth medium by
repeated pipetting with a hypodermic syringe equipped with a 20-gauge
5-inch spinal-tap needle.
7. Following a final filtration through a Nitex (Nylon monofilament)
screen with a mesh opening of 10/.~m, the suspension is counted with a
hemocytometer chamber.
8. At this point, primary cultures are started in the same manner as
with mechanically dissociated cells and are used on the following day to
prepare secondary cell suspensions that have been enriched for
myoblasts.
Other investigators prefer to establish experimental and control cultures from such primary suspensions. Under these circumstances enrichment can be achieved by exposing the suspension briefly to culture surfaces to which fibroblasts attach preferentially.19
The same protocol can be used with trypsin as well although cell
viability is somewhat poorer. During the period in which we routin/ely
employed this enzyme we used 0.05% trypsin (N.B.C. or Difco t":300)
made up in Puck's saline G.
Secondary Cell Suspension
We prefer to use cultures from secondary suspensions harvested from
briefly cultured primary cells for the following reasons: (1) We feel that we
is T. T. Puck, S. J. Cieciura, and A. Robinson,J. Exp. Med. 108, 949 (1958).
19D. Yaff,Proc. Nail. Acad. Sci. U.S.A. 61, 477 (1968).
[45]
517
get a more accurate count of viable cells (after plating such secondary
suspensions we see relatively few "floaters"). (2) W e c a n more readily
enrich for myoblasts during the harvest of primary cultures.
The protocol we adopted to prepare secondary cell suspensions from
mechanically dissociated primary cultures is described below. In this procedure we now use crystalline trypsin 2 since it has the singular advantage
of being rapidly inactivated by crystalline soybean trypsin inhibitor 2 in
stoichiometric amounts.
The minimum effective concentration of enzyme is empirically determined. Primary cultures are briefly exposed to the enzyme, and enzyme activity is stopped rapidly. We have also abandoned the practice of
centrifuging down the cells, washing, and resuspending the pellet (see
preceeding page, steps 5 and 6). This step, we find, lowers the cell yield
(either by damaging cells or by the loss of cells in undissociable cell
clumps).
1. The medium is aspirated from primary cultures incubated overnight
and each petri plate rinsed with 10 ml of saline G to remove residual
medium.
2. An iced solution of trypsin (Worthington, 3 crystallized), 5 ml
containing 13/zg/ml in saline G, is delivered into each petri plate.
3. The petri plates are maintained at room temperature and observed
periodically at a magnification of 100 under phase-contrast microscopy.
4. Cultures are gently swirled intermittently. When approximately
half of the cells have either rounded up or detached from the surface,
about 5 min, the enzyme solution is gently pipetted over the culture
surface.
5. The resultant cell suspension is quickly transferred to an iced 40 ml
Pyrex conical centrifuge tube of complete growth medium (0.5 ml for each
primary plate used) containing 0.5 mg/ml of soybean trypsin inhibitor ( 3
crystalline, Worthington).
6. The suspension is mixed well, and the cells are enumerated in a
hemocytometer chamber. Suspensions prepared as described above tend
to be dilute and are generally counted by the procedure used for white cell
counts.
Procedures for the enrichment of myoblasts in cell suspensions are
based on the observation that fibroblasts stretch out and are more tenaciously bound to the culture surface 21 (see Fig. 2). In the above protocol,
differential release of myoblasts is achieved by a controlled, brief trypsinization that leaves most of the fibroblasts bound to the petri plate.-We
2o Worthington Biochemical Co.
zl I. R. Konigsberg, Science 140, 1273 (1963).
518
[45]
Fig. 2. Morphological differences between cells in a clone of chick muscle fibroblasts (A)
and a clone of myoblasts (B) photographed on the fourth day of culture using phase-contrast
optics. Since fibroblasts attach more tenaciously they appear to be larger. The remarkable
similarity of cell shape predictably identifies the clonal type.21a
prefer to sacrifice yield for purity o f cell t y p e and can, with care, obtain
cell suspensions in which a m i n i m u m of 96% of the cells are myogenic. 22
Inoculum Size, Medium Composition, and Synchrony
One o f the chief a d v a n t a g e s of muscle cell culture as an experimental
s y s t e m is the high degree of s y n c h r o n y of differentiation that can be
achieved. S y n c h r o n y can be i m p o s e d b e c a u s e cell culture is a closed
s y s t e m in which the proliferating cells eventually deplete the m e d i u m o f
heterologous, exogenous mitogens and lag in the G~ p h a s e o f the cell
cycle. T h e protraction o f G~ is the cue for the initiation o f differentiation.
By adjusting the inoculum size and the volume and composition of the
growth medium, the timing o f differentiation can be manipulated within
limits. The limits are i m p o s e d b y the establishment, in static cultures of a
gradient o f depletion in the immediate vicinity of the cells. This can be
21a I. R. Konigsberg, Scientific American, 211, 61-66 (1964).
p. A. Buckley, and I. R. Konigsberg, Dee. Biol. 37, 186-193 (1974).
22
[45]
519
520
[45]
of cell crowding in culture can hardly be considered physiological. Indeed, our experience has been that a smaller percentage of such inocula
actually attach. Synchrony is completely lost in such cultures, myogenic
fusion being initiated during the first 24 hr in culture, and, as one might
expect, cell proliferation is diminished by cell crowding. If, as we suspect,
the use of such heavy inocula reflects an inability to obtain cell survival
and proliferation at lower cell density, the investigator should reexamine
the cell dissociation techniques, media preparation, and incubation conditions that are being used.
The criteria for the adequacy of these parameters are the plating efficiency, colony size, and differentiation of cells plated at clonal density
(200-400 cells per 5-cm dish). Hating efficiencies of about 20% (chick) and
40% (quail) are acceptable.
Media (General Considerations)
Media composition, preparation, and sterilization are discussed elsewhere in this volume [1] [2] [5]. This section will deal with just those
media requirements peculiar to freshly isolated cells and muscle cells
particularly. These cells, unlike cell lines, are neither adapted to nor selected for the culture environment. They are, therefore, more sensitive to
variations in quality of media components than are established cell lines.
I00
80
,7 60
r~
N20
DAYS IN CULTURE
[45]
521
1. We have been forced for logistic considerations to use commercially prepared media components, such as MEM, F10, and FI2. Although these products are satisfactory they are not quite up to the standard of these components as prepared by ourselves from reagent-grade
chemicals.
2. All of the sera that we use are pretested by us, using the ability of
each serum when incorporated in our "high-growth" medium to support
growth and differentiation at clonal density (see above). Generally, one of
two (rarely four) samples of horse serum proves satisfactory, and we
order and store at - 6 0 sufficient serum to last the year.
3. Unlike cell lines, freshly isolated myoblasts require, in addition to
horse serum, the incorporation of a saline extract of chick embryos. In the
absence of embryo extract, little if any proliferation occurs. The protocol
(see "Appendix") for preparing this extract is the result of exhaustive
empirical testing of the mincing procedures, extraction time, centrifugation speeds and times, as well as filtration procedures. We have employed
this protocol, with only minor changes, over the past 10 years with good
results. The extract has a useful life of only 2 weeks even when stored at
-60 and therefore is prepared on a regular schedule. Alternative methods
of preparing extract are employed by other investigators. These alternatives range from essentially the same procedures as ours, 2~to substituting
a brief (20-min) blending step (Waring Blendor) z6 for the mincing procedure we use, to forcing the embryos through a stainless-steel screen followed by freeze-thawing and centrifugation. 27
Constituents of the Medium
522
[45]
[45]
523
The standard medium used in this laboratory was developed to support the growth and differentiation of avian myoblasts at clonal density
applying the criteria of plating efficiency, colony size, and per cent differentiated clones. For that reason it is a "high-growth" medium, supporting
a rapid rate of cell proliferation. In developing this medium a number of
parameters were systematically varied, the most important of which, it
turned out, were the proportions of serum and embryo extract in the
medium. The concentration of equine serum that proved optimal was 15%
(v/v). We found also that for chick myoblasts the maximum concentration
of embryo extract was 5% (v/v) and that higher concentrations were actually inhibitory (see also Stockdale3~). In adapting this medium for quail
myoblast culture 3 we found that Eagle's MEM was as effective a nutrient
base as F10 and that the quail cells, unlike the chick, would tolerate and
do better on 10% embryo extract medium.
The high-growth ( " F M 10-15") medium is currently employed for
cells at both clonal and mass density (see above) and consists of:
Eagle's MEM (with Earle's salts) 36
Equine serum
Embryo extract
Penicillin-streptomycin stock 37
Fungizone stock 3s
740
150
100
10
2.5
ml
ml
ml
ml
ml
524
[45]
990
500
10
2.5
ml
mg
ml
ml
This defined synthetic medium does not support cell proliferation but
will support rapid, synchronous fusion (or myosin synthesis in low-Ca 2+,
fusion-blocking F12[PVP]) when cultures are switched from growth to
defined medium. ~4 This indicates that no constituent of either serum or
embryo extract is required for either fusion or cell-type specific synthesis.
It suggests, in fact, that these heterologous components of the medium,
by reason of stimulating proliferation, prevent or delay the initiation of
differentiation. This suggestion is also supported by the observation that
fusion is inhibited by continuous perfusion with high-growth medium but
not with F12(PVP). 23
Conditioned Media
Conditioning freshly prepared media by subjecting it to the metabolic
activities of cultured cells could conceivably alter media composition in a
number of ways, for example, by the depletion of media constituents or
the accumulation of cell products. Such media were employed in developing the art of cell culture (see this volume [5]) and have also proven useful
in the development of the muscle cell culture system. Specifically, conditioned media have been used: (1) to develop adequate cloning procedures, 21 (2) to study and control the time of initiation of differentiation, 3,2z'2a and (3) to identify subpopulations of myoblasts in early
embryonic development. 25,z9 Further analysis of two of these media has
led to some understanding of the nature of their alteration and eventually
to better-defined substitutes for the conditioning procedures. To a large
extent it is no longer necessary to employ conditioned media to clone
myoblasts (from embryos o f advanced stages) since a gelatin- or
39 N. K. White and S. D. Hauschka, Exp. Cell Res. 67, 479 (1971).
[45]
525
collagen-coated culture surface adequately substitutes for the altered medium. 4,41 In addition synchronous differentiation now can be achieved by
a step-down to defined medium (see above) rather than by employing a
nit rlogen-depleted medium.
H o w e v e r , should the use of any of these conditioned media seem
advantageous to the investigator's aims he would be advised to follow the
published protocols in minute detail, at least initially. All of the protocols
cited above were derived empirically. Media conditioned by each of the
three protocols elicit quite a different response from the cultured myo-.
blast. These protocols differ principally with respect to cell type, population size, and duration of conditioning. The different results achieved with
each most probably reflect a different balance of the large number of
metabolically generated changes that are occurring. Very brief exposure
to moderate numbers of cells might simply promote the accumulation in
the medium of low-molecular-weight biosynthetic products that leak into
the medium. 42 At the other extreme of population size and duration the
over riding effect may reflect the depletion of mitogenic medium
components.
Collagen P r e t r e a t m e n t of the Culture Dish
Myoblasts adhere and stretch out more satisfactorily to a collagen or
gelatin substratum than to the bare polystyrene TC dish, as supplied
by the manufacturer. Furthermore, unless provided with such an attachment surface, the percentage of clones that differentiates is low. The few
clones that do differentiate are small and contain relatively few muscle
fibers. 4'41'43 In mass cultures established from cell suspensions that are
not enriched for myoblasts, differentiation is equally normal in collagentreated and untreated petri plates. Although not strictly required, the use
of pretreated plates is advantageous since it delays the detachment of the
cell sheet that eventually occurs in long-term mass culture.
Collagen can be applied by raising the ionic strength of a concentrated
solution of the protein in 0.15 M acetic acid. A small measure volume is
then spread on the petri plate surface, which is subsequently rinsed. 4~
Solutions of gelatin in distilled water also can be spread and simply allowed to dry. 44
40I. R. Konigsberg and S. D. Hauschka, in "Reproduction: Molecular, Subceltular and
Cellular" (M. Locke, ed.), pp. 243-289. Academic Press, New York, 1965.
41 S. D. Hauschka and I. R. Konigsberg, Proc. Natl. Acad. Sci. U.S.A. 55, 119 (1966).
42H. Eagle and K. Piez, J. Exp. Med. 116, 29 (1962).
43I. R. Konigsberg, Proc. Natl. Acad. Sci. U.S.A. 47, 1868 (1961).
44S. D. Hauschka, in "Growth, Nutrition and Metabolism of Cells in Culture" (G. H.
Rothblat and V. J. Cristofalo, eds.), Vol. II, pp. 67-130. Academic Press, New York, 1972.
526
[45]
To insure more uniform distribution and to avoid the tedium of spreading we now flood TC petri plates with a larger measured volume, 1.4 ml,
of more dilute collagen or gelatin. 3,45 The protein is allowed to adsorb to
the surface overnight (about 18 hr) at room temperature in an atmosphere
equilibrated with the protein solvent used. The collagen solution applied is
made up by adding 40/zl of 3.76% NaCl to 1.0 ml of a purified collagen
stock containing 40/xg collagen per milliliter. Gelatin solutions contain 100
/zg/ml of a good bacteriological grade of gelatin (Oxoid) in distilled water.
After aspirating the excess, plates are rinsed twice with distilled water,
placed in a desiccator over silica gel, and stored at room temperature until
used.
Our procedure for preparing and purifying collagen has been published. 3 It is more exhaustive than need be, and our earlier procedure 43 is
perfectly adequate. We find it simpler to prepare and handle collagen
sterilely. However, we have sterilized collagen-treated petri plate surfaces under UV light and found them perfectly satisfactory. Since gelatin
solutions can be autoclaved the investigator may prefer this alternative.
We have found no difference between the native and denatured molecule
in routine culturing. When cultures are to be subjected to constant 23 or
intermittent 3 perturbation, however, myoblasts seem to adhere better to
the collagen substratum.
Appendix: Procedure for Preparing Embryo Extract
1. Incubate fertile chick eggs for 12 days. Candle and discard infertile
eggs and eggs containing dead embryos.
2. Cut a circular hole in the blunt end of the egg with a pair of curved
scissors and remove the embryo.
3. Cut off and discard that portion of the head anterior to the posterior
border of the eyes. Make a longitudinal cut through the body wall from
the cloaca to the base of the neck. Collect the embryos in a 150-mm petri
plate.
4. When all of the embryos have been collected, decant the blood and
empty the petri plates onto paper toweling. Transfer the embryos to a
large square of double-thickness cheesecloth. Suspend the cheesecloth
" b a g " in a beaker and wash with several changes of P-C chick 3ra (until
most of the blood is removed).
5. Place the cheesecloth on several thicknesses of paper toweling (to
drain) and transfer the embryos to a tared beaker and weigh.
45 I. R. Konigsberg, in "Chemistry and Molecular Biology of the Intercellular Matrix"
(E. A. Balazs, ed.), Vol. 3, pp. 1779-1810. Academic Press, New York, 1970.
[46]
HORMONE-PRODUCING
PITUITARY
CELLS
527
[46] C l o n a l S t r a i n s o f H o r m o n e - P r o d u c i n g
P i t u i t a r y Cells
Clonal strains of pituitary tumor cells that synthesize and secrete prolactin, growth hormone and adrenocorticotropic hormone (ACTH) have
been established and serially propagated in culture for periods of up to 15
years. The rates of biosynthesis of the specific hormonal peptides by these
clonal strains are high (up to 10% of total protein synthesis in appropriately stimulated cells), and they respond in culture to many of the same
regulatory factors as normal pituitary cells do in situ. However, because
certain of these regulatory factors, such as the hypothalamic peptides,
affect more than one cell type in the intact pituitary gland, it is not possible to perform unambiguous mechanistic studies with hemi-pituitary
fragments or mixed primary cultures of freshly dispersed pituitary cells.
Therefore, strains of homogeneous populations of functional cells serve as
useful model systems for determining the mechanisms of action of factors
that regulate the release and synthesis of prolactin, growth hormone, and
ACTH.
528
[46]
E s t a b l i s h m e n t o f P i t u i t a r y Cell S t r a i n s
T h i s s u m m a r y is n o t i n t e n d e d as a r e v i e w o f all f u n c t i o n a l p i t u i t a r y cell
s y s t e m s n o w in u s e , b u t r a t h e r a s a d e s c r i p t i o n o f cells n o w a c t i v e l y b e i n g
s t u d i e d in this l a b o r a t o r y . T h e s e i n c l u d e t h e r a t G H c e l l s a n d m o u s e
A t T 2 0 / D 16 cells.
A variety of cultures from a growth hormone- and prolactin-producing
r a t p i t u i t a r y t u m o r , M t T / W 5 , 1 w e r e e s t a b l i s h e d in 1965. 2 B e c a u s e t h e
organ-specific function originally characterized was growth hormone prod u c t i o n , t h e s e cells b e c a m e k n o w n in t h e l i t e r a t u r e as " G H c e l l s . " I t w a s
l e a r n e d s u b s e q u e n t l y t h a t c e r t a i n G H cell s t r a i n s a l s o p r o d u c e p r o l a c t i n . 3
Cells w e r e a d a p t e d t o g r o w t h in v i t r o b y a l t e r n a t e c u l t u r e a n d a n i m a l
p a s s a g e . 2 D i s p e r s e d t u m o r c e l l s w e r e a l l o w e d to a t t a c h a n d g r o w in p l a s tic c u l t u r e d i s h e s f o r p e r i o d s o f s e v e r a l ~lays to a b o u t 1 m o n t h . T h e
s u r v i v i n g a t t a c h e d cells w e r e t h e n h a r v e s t e d a n d i n j e c t e d i n t o r a t s o f t h e
W i s t a r - F u r t h s t r a i n . W h e n a n e w t u m o r d e v e l o p e d , it w a s r e m o v e d a n d a
s e c o n d g e n e r a t i o n o f c u l t u r e - d e r i v e d t u m o r cells w a s e s t a b l i s h e d in vitro.
This process of alternate passage between culture and animal was repeated
s e v e r a l t i m e s in o r d e r to s e l e c t f o r t u m o r cells t h a t w o u l d a t t a c h a n d g r o w
r e a d i l y in t h e c u l t u r e e n v i r o n m e n t . 4
S e v e r a l e p i t h e l i a l cell s t r a i n s h a v e b e e n c l o n e d , z a n d s o m e h a v e b e e n
m a i n t a i n e d in c o n t i n u o u s c u l t u r e f o r a s l o n g a s 10 y e a r s w i t h o u t loss o f
hormone production. Table I gives the characteristics of several clones of
G H cells, z'a'5-~5 T h e GH1 a n d GH3 s t r a i n s a r e a v a i l a b l e f r o m t h e A m e r i c a n
T y p e C u l t u r e C o l l e c t i o n (12301 P a r k l a w n D r i v e , R o c k v i l l e , M a r y l a n d ) ;
t h e C e l l R e p o s i t o r y N u m b e r s f o r t h e s t r a i n s a r e C C L 82 a n d C C L 82.1,
respectively.
1 H. Takemoto, K. Yokoro, J. Furth, and A. I. Cohen, Cancer Res. 22, 917 (1962).
2 A. H. Tashjian, Jr., Y. Yasumura, L. Levine, G. H. Sato, and M. L. Parker, Endrocrinology 82,342 (1968).
3 A. H. Tashjian, Jr., F. C. Bancroft, and L. Levine, J. Cell Biol. 47, 61 (1970).
4 V. Buonassisi, G. Sato, and A. I. Cohen,Proc. Natl. Acad. Sci. U.S.A. 48, 1184 (1962).
5 A. H. Tashjian, Jr., N. J. Barowsky, and D. K. Jensen,Biochem. Biophys. Res. Commun.
43, 516 (1971).
e T. F. J. Martin and A. H. Tashjian, Jr., Biochem. Actions Horrn. 4, 269 (1977).
7 A. Schonbrunn and A. H. Tashjian, Jr., J. Biol. Chem. 253, 6473 (1978).
8 H. H. Samuels, J. S. Tsai, and R. Cintron, Science 181, 1253 (1973).
9 H. H. Samuels, J. S. Tsai, J. Casanova, and F. Stanley, J. Clin. Invest. 54, 853 (1974).
10H. H. Samuels, Z. D. Horwitz, F. Stanley, J. Casanova, and L. E. Shapiro, Nature
(London) 268, 254 (1977).
H D. K. Biswas, J. Lyons, and A. H. Tashjian, Jr., Cell 11,431 (1977).
12p. S. Dannies and A. H. Tashjian, Jr., J. Biol. Chem. 248, 6174 (1973).
13p. M. Hinkle and A. H. Tashjian, Jr., J. Biol. Chem. 248, 6180 (1973).
14L.-Y. Yu, R. J. Tushinski, and F. C. Bancroft, J. Biol. Chem. 252, 3870 (1977).
15 U. I. Richardson, J. Cell. Physiol. 88, 287 (1976).
[46]
529
'~,
,~:=
~,~.
:=
%
J
,.J
I:::
a~
0
.<
'~~:
~.~.~
.~.~ ~
~o
"~
._
[-
0
Z
0
t~
0
o
h.
h,
<
.,<
(J
0
.<
Z
0
~J
Z
~0
m
530
[46]
H o r m o n e - p r o d u c i n g mouse pituitary t u m o r cells have also been established in culture. The AtT20/D16 cells are a clonal strain isolated in 1962
by Yasumura TM from a radiation-induced pituitary t u m o r in the mouse.17
These cells have continued to produce large amounts o f A C T H during
serial propagation in culture for 15 years. TM Table II gives the functional
characteristics o f this strain, la-z3 The AtT20/D16 strain is available from
the American Type Culture Collection (Cell Repository N u m b e r C C L 89).
Methods of Culture
Type o f Culture. G H cells can be grown as monolayers on plastic or
glass surfaces, 2 in roller bottles, or in suspension culture. 24
M e d i u m . G H cells are not fastidious in their medium requirements.
They are usually grown in monolayer in this laboratory in H a m ' s nutrient
mixture F1025 supplemented w i t h 1 5 % horse serum and 2.5% fetal calf
serum at 37 _ 0.5 in a humidified atmosphere o f 5% CO2 and 95% air. 2
They also can be grown in H a m ' s nutrient mixture F12, 26 Eagle's minimum essential medium, 27 and D u l b e c c o ' s modified Eagle's medium 28
supplemented with horse serum (5-15%) and 2.5% fetal calf serum. In
addition, for experiments of relatively brief duration (1-48 hr) requiring
the absence of serum, G H cells can be maintained in a functional
hormon-producing and hormone-responsive state in either N e u m a n and
Tytell's serumless medium 29 or in H a m ' s nutrient mixture F10 supplemented with lactalbumin hydrolysate (5 g/liter). Hayashi and Sato have
reported growth of GH3 cells in chemically defined medium supplemented
with certain " h o r m o n e s . ''3
For spinner cultures, modified Eagle's medium for suspension 31 supplemented with 15% horse serum and 2.5% fetal calf serum is used.
[48]
.=.
o
%
m
E
J
m
Ii
m
o
o
m
t,
t...
e-~
il ..~
O
E,
O
,-r
o
<
Z
0
;>-.
m
[..
<
H ,:.:,
r=
o
..
531
532
[46]
Characteristics of G H Cells
Stability. The rates o f growth and hormone production by G H cells
have generally remained unchanged o v e r many years o f continuous propagation. H o w e v e r , some changes have been noted: for example, GH3 cells
that initially produced about equal quantities of growth hormone and
prolactin 3 now synthesize 5-10 times more prolactin than growth hormone, and in the GH4C, strain derived from GH3 cells, 34 the ratio o f
prolactin to growth hormone is even greater. It is probable that such
changes in the ratio of hormone production reflect some unknown current
environmenthl alteration, such as medium composition, because GH3
cells that are thawed from liquid nitrogen change their ratio o f prolactin to
growth hormone synthesis o v e r a period o f several weeks from about 1 : 1
to the same high ratio (about 10 : 1) as serially propagated GH3 cells.
K a r y o t y p e . G H cells are aneuploid with a modal n u m b e r of chromosomes ranging from 69 to 76. 35 The modal c h r o m o s o m e n u m b e r for the
AtT20/D16 cells is 76. '8
H o r m o n e Production. As described in Table I, most o f the G H cell
strains, except GH,2C1 and GC, produce both growth hormone and prolactin although the ratios of hormone synthesis among different clones
varies considerably, with GH3 and GC being the highest producers of
32O. P. F. Ciausen, K. M. Gautvik, and T. Lindmo, Virchows Arch. B 23, 195 (1977).
a3T. F. J. Martin and A. H. Tashjian, Jr., J. Biol. Chem. 253, 106 (1978).
34A. H. Tashjian, Jr., P. M. Hinkl, and P. S. Dannies, in "Endocrinology" (R. O. Scow,
ed.), p. 648. Excerpta Med. Found., Amsterdam, 1973.
35C. Sonnenschein, U. I. Richardson, and A. H. Tashjian, Jr., Exp. Cell. Res. 61,121 (1970).
[46]
533
TRH
Effect
A c u t e actions (0-30 min)
Estrogens
Thyroid hormone
Somatostatin
Increases GH synthesis
Decreases PRL synthesis
Increases TRH receptor number
17/3-estradiol increases PRL and
decreases GH production
Antiestrogens increase PRL synthesis
Increases cell growth
Increases GH synthesis
Decreases TRH receptor number
Binds to specific receptors
Decreases GH release
Decreases PRL release
Bromocriptine
Prostaglandins
Cations
References
13.40--43
44
45--47
6,42
33.48
5,6.12,49.50
5.6,51
6,52,53
33,48
51,54
10,14,36,55,56
12,36
43
51,57.58
57
8
8--10
59
7
7
7
51,60
61
61
62,63
534
[46]
[46]
535
GH C e l l s as a M o d e l S y s t e m
As summarized above, GH cells produce large amounts of growth
hormone and prolactin. The biosynthesis and release of these two protein
hormones are modulated by specific factors that have been found to influence the pituitary gland in the intact animal. In fact, observations initially
made with GH cells were subsequently shown to operate in v i v o . 5 Unlike
the pituitary gland, however, GH cells are clonal populations that can be
studied in a highly controlled environment. They therefore represent a
useful tool for studies on the molecular mechanisms underlying the actions of hypothalamic peptides on growth hormone and prolactin release
and synthesis as well as how peptide hormone receptors are modulated.
Because GH cells are neoplastic, their functions and responses may not
be identical to comparable cells in the normal animal. It is recognized that
tumor cells can lose functions or express functions inappropriately, but it
is not likely that they invent new functions or novel mechanisms. Nevertheless, it is essential to perform, wherever possible, control experiments
with normal pituitary cells or explants to confirm that mechanisms deduced from tumor cells correspond to normal mechanisms.
536
S P E C I F I C CELL L I N E S
[47]
[47] L i v e r Cells
1 H.
2 H.
3 H.
4 H.
L
L.
L.
L.
TABLE I
CONSTRUCTION OF 8 LITERS OF TWICE-CONCENTRATED(X2) MEDIUM
Step
Procedure
1. Thaw:
2.
3.
4.
5.
6.
7.
8.
9.
10.
TABLE II
STOCK SOLUTIONS FOR ARGININE-FREE MEDIUM
6.00
8.40
20.96
20.96
29.24
6.00
16.80
13.20
8.40
19.04
3.20
18.72
g
g
g
g
g
g
g~
g
g
g
g
g
Vitamins (2 liters)
Choline chloride
Folic acid
Nicotinamide
Pantothenic acid
Pyridoxal HC!
Thiamine
Riboflavin
Antibiotics (330 ml)
Penicillin G
Streptomycin sulfate
Antimycotic (1 liter)
n - Butyl-p -hydroxybenzoate
0.80
0.80
0.80
0.80
0.80
0.80
0.08
g
g
g
g
g
g
g
30 106 U
6.0 g
0.1 g
Can be omitted or added from a standard sterile stock solution (332 mg/100 ml water;
add 2.5 ml/100 ml l medium).
538
[47]
TABLE III
FETAL RAT LIVEn CELL CULTURE PLATING PROCEDURE
Step
Manipulation
1. Lightly etherize pregnant rat; immobilize the animal on its back and soak entire abdomen with 95% (v/v) ETOH.
2. Make a midline incision with a sterile scissors, cutting first skin and then muscle layers;
do not perforate intestines, which lie under muscle layer.
3. Remove fetuses one at a time from embryonic sacs. Cut umbilical cord and decapitate
immediately. Place about 10-15 into a sterile 9-cm dish.
4. Dissect out intact livers sterilely and put about 15 into a 5-cm dish containing 5 ml
ornithine-minus medium at 37 supplemented with 10% (v/v) serum. Inspect each liver
to ensure absence of nonhepatic tissue.
5. Cut each liver into its four separate "lobes"; remove medium by aspiration and wash
once with 5 ml of fresh medium.
6. Pool washed, cut tissue and incubate in fresh collagenase-supplemented digestion buffer
(1-2 mi/liver), for 8-10 min at 37 in a 125-ml Erlenmeyer flask containing a magnetic
stirring bar. Set rheostat = 35-40.
7. Tilt flask to allow undigested tissue to settle and remove cell suspension, with a sterile
Pasteur pipette, placing it into a "storage" flask containing 10 ml medium (maintain
on ice).
8. Repeat tissue digestion steps 6 and 7, a total of 4-5 times, pooling each subsequent
harvest. Add an equal volume of fresh ice-cold medium to the final cell suspension and
swirl gently. Dispense 20-40 ml (by pouring) into sterile 50-ml plastic tubes.
9. Centrifuge 10 sec at full speed and apply brake immediately thereafter.
10. Remove supernatant liquid by suction and discard; wash pellet once with 10 ml of
serum-free medium (blow fluid rapidly against sidewall to disrupt pellet and then take
up the cell suspension; blow it out gently against sidewall). Repeat step 9.
11. Discard second supernatant liquid. Resuspend pellets as in step 10, pool (store on ice),
and titer a 0.5-ml aliquot.
12. Pipette cell suspensions into appropriate media and plate using a flow syringe.
P o r c i n e in-
Inorganic Reagents
T r i p l e - d i s t i l l e d w a t e r is u s e d u n l e s s o t h e r w i s e n o t e d . T r i s b a s e is f r o m
Sigma.
Equipment. T i s s u e c u i t u r e i n c u b a t o r s s u p p l i e d w i t h CO2 a n d a i r l i n e
f i l t e r s , r e g u l a t o r y v a l v e s ( s e t t i n g s a r e 0.5 a n d 4 . 0 f o r CO~ a n d a i r , r e s p e c -
[47]
LIVER CELLS
539
TABLE IV
ADULT RAT LIVER CELL CULTURE PLATING PROCEDURE
Step
Manipulation
1. Lightly etherize rat. Immobilize the animal on its back and soak entire abdomen with
95% ethanol.
2. Make a midline incision extending into the thoracic cavity. Exsanguinate the animal by
cardiac puncture, removing 5-8 ml blood, and push the intestinal contents aside
everting them to expose portal vessels.
3. Clamp portal vein (to the left of splanchnic and gastric venous inputs) together with the
celiac trunk (small pulsating arterial system dorsal to everted portal system). Clamp
inferior vena cava above the renal venous inputs but below the hepatic vein.
4. Insert a 20-ml plastic syringe equipped with a 25-gange needle (bevel up) and perfuse
20 ml hormone, purine, and collagenase-supplemented "digestion buffer" for 2-3 rain
through the liver. Complete tissue blanching; a light tan color indicates adequate
perfusion.
5. Excise perfused liver, placing it in a 9-cm plastic petri dish and removing extraneous
tissue if present. Wash liver twice with 10 ml of "complete medium." Add 10 ml of
fresh medium and dissect tissue (two livers) into 5 8 mm pieces and wash twice.
6. Incubate cut tissue at 37 with 40 ml of hormone, purine, and collagenase-supplemented
digestion buffer for 8-10 min in a 125-ml Erlenmeyer flask containing a magnetic
stirring bar. Set rheostat = 35--40.
7. Tilt flask to allow undigested tissue to settle, and remove cell suspension with a sterile
Pasteur pipette, placing it into a flask containing 10 ml of medium (maintain on ice).
8. Repeat tissue digestion (steps 6, 7) four times, pooling each subsequent harvest. Add
160 ml ice-cold "complete" medium (a total volume of about 320 ml) and dispense
40 ml into eight 50-ml sterile plastic centrifuge tubes.
9. Centrifuge 13 sec (full speed) and apply brake.
10. Remove supernatant liquid by suction and discard. Wash pellet once with 10 ml of
" b a s a l " medium; blow fluid rapidly against the sidewall to disrupt pellet and then take
up the cell suspension and blow it out gently against sidewall. Repeat step 9.
11. Remove by suction and discard clear supernatant fluid; pellets should be "white."
Resuspend pellets as in step 10 twice but blow out hard. Transfer to ice-cold sterile
bottle, swirl pooled cell suspension (about 80 ml), and resuspend seven times. Titrate a
0.5-ml aliquot.
12. Redistribute in portions of 20 ml or less to centrifuge tubes and repeat step 9.
13. Resuspend pellets in 10 ml (twice, hard) of appropriate plating media. Bring up to
needed volume and plate using a 10-ml plastic pipette.
540
[47]
TABLE V
BUFFER SOLUTIONS
Ca2Mge-freeconcentrate (2 liters)
NaCI
KC1
Na2HPO4 (anhyd.)
Tris base
1 N HCI
(Adjust pH to 7.4)
64.0
3.04
0.08
24.0
160
500
1500
25 g
1.0 liter
Steps:
1. Dissolve 8 hr at 4 with stirring; let settle.
2. Filter supernatant at 21 using Whatman
No. 1 paper.
3. Sterilize with 0.22/.Lm GS MiUipore and
AP2512450 prefilter.
4. Store 60 ml portions at - 2 0 .
ml
ml
750
2250
2
2
ml
ml
ml
ml
liter)
Trypsin concentrate
60
1% phenol red
2
(Filter through 0.45/zm Nalgene filter)
Sterile Ca2+Mg2+-free Tris buffer 540
Store 10 ml portions at - 2 0 .
ml
ml
ml
0.224
7.587
0.138
1.802
0.0012
0.010
0.010
0.010
g
g
g
g
g
g
g
g
[47]
LIVER CELLS
541
T A B L E VI
DIFFERENTIATED PROPERTIES OF PRIMARY MONOLAYER RAT
LIVER CELL CULTURES
Enzymes
Gluconeogenic
Hemopexin
Very low density lipoprotein
UItrastructure c
Bile " c a n a l i c u l i "
Desmosomes
Glycogen granules
Peroxisomes
Tight j u n c t i o n s
Fetal
Adult
+
U"
+
U
+
+
+t,
+
+
+
+
U
U
+
+~
+b
+
+
+
+
+
+
_c
+
+b
U
U
+b,c
+
+
+
+
+
+
+
+
+
+
a Unknown.
0 G r o w t h state dependent (H. L. Leffert et al., Proc. Natl. Acad. Sci. U.S.A. 75, 1834
(1978).
c U n p u b l i s h e d observations (D. Weinstein, K. D e m p o , S. Sell, and H. L. Leffert).
542
[47]
Becton Dickinson (#3054) and are fitted with autoclavable I-inch ID 4!inch OD silastic tubing from Dow Coming (#601-365).
Surgery
Sterile technique is preferred but not necessary; alcohol-washed areas
and instruments suffice. Animals are placed onto clean absorbent cotton
pads layered over a simple fiat cork board and operated on between
0900-1200 hours.
~7]
LIVER CELLS
543
containing enzyme is supplemented with 1 ml each of the insulin, hydrocortisone, and inosine stock solutions.
Cell Isolation and Plating
Detailed procedures appear in Table III (fetal) andTable IV (adult). All
steps are performed with sterile technique. The stirring speed, i.e., rheostat knob setting, for all tissue digestion is 35-40. For fetal cultures, four
animals are used and usually yield 20-40 fetuses giving rise to about
1-3 108 cells. For adult cultures, two livers are used giving rise to about
1-2 108 cells. For both'systems, the time from surgery to plating should
be about 3 hr or less. At plating, greater than 95% of isolated cells are
hepatocytes 3 with 70-95% viability as judged by exclusion of 0.05% (w/v)
trypan blue at 1 min.
Seeding densities, i.e., numbers of cells per milliliter medium, and
volume of media per dish vary with experimental design. 1-3 For the fetal
system, usual parameters for 35-, 50- and 90-mm diameter dishes are
1.25 x 105 cells/ml, 2 ml; 1.5 x 105 cells/ml, 5 ml; and 2 105 cells/ml, 10
ml, respectively. For the adult system, 5 105 cells/ml, 2 ml, are plated
per 35-mm dish. " H e a l t h y " cultures show cellular aggregates upon the
surface of the dish within 24 hr and flattening (nonrefractile nuclear and
cytoplasmic morphology under phase optics) within 48 hr. Medium
changes are performed as needed but are not necessary. In this laboratory, functional fetal cultures plated without L-ornithine have survived for
6-8 weeks without a medium change; functional adult cultures plated with
L-ornithine have survived for 2 weeks without a medium change.
Cell Counts
At plating, titrations are performed with 0.5 ml of cell suspension
added to 9.5 ml of Isoton (Scientific Products, #B3157-11) and counted
electronically (threshold at 10 ~m; attenuation and aperture settings
of 1 and 256, respectively). Hemocytometer counts should be within
___10% of corrected electronic counts using these settings.
Table V lists buffer solutions used to measure attached cell numbers.
Dishes are washed twice with Tris buffer, pH 7.4 (a volume equal to initial
medium volume). Then an equal volume of Ca2+Mg2+-free Tris buffer with
trypsin (10 ml trypsin stock solution per 180 ml Ca2+Mg2+-free Tris
chloride at pH 7.4) is added and the dishes incubated for 30-40 min at 37.
A 2 ml (#122402010) or 5 ml (#122405010) glass handpipette (Bellco)
equipped with a rubber bulb (Scientific Products, #R5000-1) is useful for
544
SPECIFIC
CELL
LINES
[48]
detaching the cells; vigorous pipetting, about 20 times, over the dish
surface usually reduces cell aggregation. The cell suspension or an aliquot
of it is brought up to 10 ml with Isoton and counted.
Functional Properties
Typical differentiated properties of primary monolayer rat liver cell
cultures are summarized in Table VI. Additional sources, including those
cited above, should be consulted, lo-19
10 S. Sell, H. L. Leffert, U. Mueller-Eberhard, S. Kida, and H. Skeily,Ann. N. Y. Acad. Sci.
259, 45 (1975).
11 H. Watabe, H. L. Leffert, and S. Sell, in "Onco-Developmental Gene Expression" (W.
Fishman and S. Sell, eds.), p. 123. Academic Press, New York, 1976.
12 D. M. Bissell, L. E. Hammaker, and U. A. Meyer,'J. Cell Biol. 59, 722 (1973).
13 R. J. Bonney, In Vitro 10, 130 (1974).
14 C. Guguen, C. Gregori, and F. Schapira, Biochimie 57, 1065 (1975).
t5 G. Michalopolous and H. C. Pitot, Exp. Cell Res. 94, 70 (1975).
~e T. H. Claus, S. J. Pilkis, and C. R. Park, Biochirn. Biophys. Acta 404, 110 (1975).
lr G. M. Williams, Am. J. Pathol. 85, 739 (1976).
18 D. Bernaert, J. C. Wanson, P. Drochmans, and A. Popowski, J. Cell Biol. 74, 878 (1977).
10 H. L. Leffert and K. S. Koch, Ciba Found. Syrnp. 55 (New Ser.), 61 (1978).
[48] L i v e r Cells ( H T C )
By E. BRAD THOMPSON
HTC (hepatoma, tissue culture) cells are a line of cells established
from the ascites form of the rat hepatoma, Morris 7288C. 1 The original
hepatoma was dissected from the liver of a male rat of the inbred Buffalo
strain that had been fed the carcinogen, N,N'-2,7-fluorenylenebis-2,2,2trifluoroacetamide. It subsequently was converted to the ascites form and
carried serially as such. The HTC line was cultured directly from the
ascitic fluid of tumor-bearing rats.
HTC cells have proved useful in studying a number of both liverspecific and general cellular processes at the cellular level. The cells have
been used especially for studies of the effect of steroid hormones on
enzyme induction. Glucocorticoids induce in these cells the hepatic
marker enzyme tyrosine aminotransferase (L-tyrosine: 2-oxoglutarate
aminotranserase, EC 2.6.1.5), as well as ornithine decarboxylase and
glutamine synthetase. In HTC cells, such steroids also increase cell adhe1 E. B. Thompson, G. M. Tomkins, and J. F. Curran,Proc. Natl. Acad. Sci. U.S.A. 56, 296
(1966).
[48]
HTC CELLS
545
siveness; reduce the activity of cyclic Y,5'-AMP(cAMP) phosphodiesterase and of plasminogen activator; induce an increase in one of the
isoaccepting forms of phenylalanyl tRNA; induce mouse mammary tumor
virus in infected cells; render cells more sensitive to cAMP analogs; and
inhibit the transport of certain amino acids. Besides studies on the action
of steroid hormones, HTC cells have been utilized in studying the control
of fatty acid and cholesterol synthesis, the viral-like A particles, RNA
viruses (C particles), growth control, phenylalanyl hydroxylase, and an
extensive list of other enzymes. The cells also have been used in somatic
cell genetics, through formation of somatic cell hybrids with various other
cell types and through the isolation of stable cell variants cloned from the
original cell line.
Propagation of HTC Cells in Monolayer Culture
HTC cells are a hardy line that grow readily as monolayer cultures on
either glass or plastic substrata. They have been successfully carded in
both commercially made special glass tissue culture flasks and in simple
medicine bottles laid on side. Plastic tissue culture flasks and dishes from
virtually all major U.S. suppliers also have been employed to grow these
cells. HTC cells in monolayer culture, being transformed cells, grow in an
irregular, trabecular pattern, display both round and polygonal cell morphology, and do not exhibit density-dependent inhibition of growth. After
being seeded so as to form a lawn of individual cells, they .grow atop one
another as well as side to side, and frequently reach stationary phase with
areas of the substratum still unoccupied by cells.
Media
HTC cells are not particularly fastidious with regard to medium. They
were originally established and maintained for years in Swim's medium
#77, (modified from 5103, with hydroxyproline omitted 2) supplemented
with 20% bovine plus 5% fetal bovine serum. The accompanying table
lists the ingredients of this medium. Later it was found that they grov~ in a
number of standard media, including Eagle's minimal essential medium, 3
Eagle's basal medium, 4 Ham's F 12, 5 and improved minimal essential medium, zinc option (IMEM-ZO). n For routine culture, these media usually
2 H. E. Swimand R. F. Parker, J. Lab. Clin. Med. 52, 309 (1958).
3 H. Eagle, Science 130, 432 (1959).
4 H. Eagle,Science 122, 501 (1955).
5R. G. Ham, Proc. Natl. Acad. Sci. U.S.A. 53, 288 (1965).
A. Richter, K. K. Sanford, and V. J. Evans,J. Natl. Cancer Inst. 49, 1705(1972).
546
$77 AS USED
[48]
HTC CELLS
Concentration
(mM)
Salts
Amino Acids
1.8
0.8
5.5
116.2
26.2
1.2
Vitamins, miscellaneous
Glucose
Choline bitartrate
Folic acid
Calcium pantothenate
Mesoinositol
Nicotinamide
Pyridoxal HCI
Riboflavin
Thiamin HCI
Phenol red (sodium salt)
Concentration
(raM)
5.6
0.01
0.005
0.005
0.01
0.005
0.005
0.001
0.005
0.02
L-Alanine
L-Aspartic acid
L-Arginine (HCi)
L-Cystine (HC1)
L-Histidine (HCL - H20)
Glycine
L-Glutamine
L-Isoleucine
L-Leucine
L-Lysine (HC1)
L-Methionine
L-Phenylalanine
L-Proline
L-Serine
L-Threonine
L-Tryptophan
L-Tyrosine
L-Valine
0.3
0.15
0.8
0.05
0.05
0.15
2.0
0.2
0,2
0,2
0.1
0. I
0.15
0.2
0.4
0.05
0.1
0.4
Routine cultures are fed 3 times a week by removing the medium from
the monolayer and replacing it with fresh medium at 37 . The cells double
about every 24 hr under these conditions, and for best viability they
should be kept in logarithmic growth phase by subculturing appropriately
into fresh culture flasks or dishes. They can be subcultured from nearconfluent flasks at a ratio o f 1 : 10 or 1 : 20 with virtually 100% viability.
After such subculturing, they should show a lag phase o f no more than 24
hr, and less if handled expeditiously. For subculturing, cells can be re-
[48]
HTC CELLS
547
moved from their substratum in any of several ways. The preferred methods is to use dilute trypsin or trypsin-EDTA as follows:
Remove spent growth medium by suction. Gently introduce 5-10 ml of
phosphate-buffered saline (PBS) (138 mM NaC1, 2.7 mM KC1, 8.1 mM
Na2HPO4, 1.47 mM KH2PO4, pH 7.4) at 25, and tip the dish to wash over
cells. Remove by suction. Introduce 1-2 ml of Puck's Saline A (containing, in grams per liter: 20 mg phenol red, 0.35 g NaHCO3, 8.0 g NaC1, 0.4 g
KC1, and 1.0 g dextrose), which contains 0.05% trypsin and 0.02%
ethylenediaminetetracetic acid (EDTA), prewarmed to 37. Tip to wash
over cells. Remove by suction all but approximately 0.5 ml. Place in a 37
incubator and examine after 5 min. Tap the dish against the heel of the
hand to dislodge cells, which should come free in 5-15 min. As soon as
cells float free, add serum-containing medium or soybean trypsin inhibitor
at 37 and reflux-pipette cells gently about 3 times to give a suspension of
single cells. Plate appropriate portions of cell suspension into fresh
dishes.
Cautions: If trypsin-EDTA is interrupted too soon or allowed to continue overlong, cell clumping will result. In addition, with overlong trypsinization, there may be loss of viability. The pH during these procedures
should not be allowed to rise above 8.0; the inclusion of phenol red in the
trypsin-EDTA solution allows visual monitoring of the approximate pH. If
a large number of cultures are to be subcultured, it is recommended that
they be carried through the procedure a few at a time, and that those
waiting to be handled be kept in the air-CO2 atmosphere of the incubator.
Alternate methods of cell removal for subculturing that can be used
successfully are: scraping with an L-shaped glass rod, either naked or
with its operative end covered by a silicone rubber tip; trypsin alone, up
to 0.1 g/100 ml in saline A or PBS; scraping with a Teflon-coated magnetic
bar inside the tissue culture flask and a stronger magnet outside moved to
manipulate the inner magnet. EDTA alone is not as satisfactory as the
other methods, although it will remove some cells.
For cells grown in serum-free medium, best results are obtained with
the trypsin-EDTA technique, using soybean trypsin inhibitor to stop proteolysis. If the introduction of even this small amount of protein is to be
avoided, scraping is the second-best choice.
Cloning
HTC cells can be cloned with high efficiency by any of several standard techniques in standard medium. If good tissue culture technique is
used, cloning efficiency should be 50-100%. Methods that have produced
good results include cloning in plastic tissue culture dishes or flasks, in
548
[48]
multiwell dishes, and in soft agar. To clone in a tissue culture dish or flask,
cells are trypsin-EDTA treated as described above (if grown monolayerstyle) or simply taken as grown if in suspension culture (see below). In
any case, for best results logarithmic-phase cells should be cloned; they
are easier to obtain in monocellular suspension and give higher cloning
efficiency. The original cell suspension should be counted carefully by
hemocytometer or electronic counter and in either case examined by
phase-contrast microscopy to be sure the closest possible approximation
to a monocellular suspension is achieved. Proper attention to procedure
should result in 90--95% of all cells existing independently, with the remainder appearing as doubles and only rare groups of three or more. It is
worth performing preliminary experiments to be sure this degree of dispersion is achieved, since most cloning experiments are aimed at obtaining truly clonal populations, and contamination with greater than 5% with
pseudoclones, derived from more than a single cell, may complicate subsequent results unnecessarily.
The dispersed cells are serially diluted with growth medium at 37 ,
taking care not to pipette too vigorously, until a convenient dilution is
reached. If the cells are to be cloned e n m a s s e in large plates or flasks, a
seeding concentration of 1-2 cells cm 2 results in a yield of colonies convenient for counting or picking for subculture. Larger numbers may be
plated, of course, if selective conditions resulting in survival of only a
fraction of the seeded cells are to be used. To cleanly pick colonies, each
colony is first, isolated in a stainless-steel ring and then trypsinized, as
described by Ham and Puck. 7 Because of the possibility that multiple
colonies obtained from a single dish may include either seed colonies from
an initial site or cross-contaminated colonies from cells detached from
other, large clones, it is recommended that attempts to obtain truly clonal
populations can be carded out either in multiwell dishes or in soft agar.
For the former, good results can be obtained with the standard 96-well
dishes at a volume of about 0.1 ml per well. If cells are diluted to average
one cell per five wells at the time of seeding, then about 95% or more of the
subsequent colonies will have risen from a single cell. Seeding at higher
numbers of cells per well results in significantly greater numbers of
nonclonal colonies. Care must be exercised to keep the parent cell suspension well mixed during the seeding. This can be done by frequent
manual mixing or by use of a sterile Teflon-coated magnet in the suspended " m o t h e r " culture, turning slowly under the control of an external
magnetic stirrer. In addition, using no more than a 2-ml pipette to seed the
0.1-ml aliquots will help avoid nonrandom cell distribution. In larger
pipettes or with very slow pipetting, cells may accumulate along the
7 R. G. Ham and T. T. Puck, This series, Vol. 5, [9] p. 107.
[48]
HTC CELLS
549
550
[48]
[48]
HTC CELLS
551
work well, although the cheapest and simplest by far is the Wheaton
bottle-stir bar combination.
Initiating a suspension culture from HTC cells previously grown exclusively as monolayer cultures requires adaptation by the cells. Considerable cell death is often seen during the adaptation period, and once the
number of viable cells falls below a critical level, the entire culture dies.
Consequently, several cell concentrations should be set out initially. Cells
in monolayer culture should be adapted to the spinner medium for several
days while still grown as monolayers. The cells that come free from the
substratum should be collected by centrifugation and returned to the culture, and not discarded during medium changes. After this initial adaptation, the monolayer-grown cells are collected, with those still adhering to
the substratum removed by trypsin-EDTA. Suspend 1-5 105 cells/ml in
fresh, suspension-culture growth medium at 37 to a total volume of at
least 100 ml. Add the cell suspension to a vessel chosen so that it will be
1 2
r-~
full, containing a stir bar whose length equals 1-1.5 times the radius of
the vessel. Place the vessel on a magnetic stirrer set at a speed to produce
gentle mixing and no turbulence. There should be a slight vortex formed at
the surface of the liquid. During the adaptation period the cells may
multiply quite slowly. Medium changes, without diluting the cells, can be
carried out by separating the cells by centrifugation (5 min at 800 g) or by
removing the flask from the magnetic stirrer and allowing the cells to
settle for 30-60 min, and then removing most of the overlying medium by
suction.
Once the cells adapt to suspension culture conditions, they double
approximately every 24 hr. If fed 3 times per week, and kept between 10~
and l0 Gcells/ml, they will remain in logarithmic growth phase indefinitely.
They have been grown successfully in volumes as low as 50 ml and as high
as 50 liters. The suspension-adapted cells can be again grown as
monolayers, although at first they attach less firmly and have a more
spherical morphology.
Cryopreservation of HTC Cells
HTC cells can be stored by freezing in a viable state, using standard
methods. Medium containing at least 10% serum is supplemented with
10% dimethylsulfoxide (DMSO) or 10-15% glycerol. Cells are harvested
and suspended in this medium at about 106 cells/ml. Aliquots of 1-1.5 ml
are placed in small screw-capped ampules and immediately frozen, lowering the temperature at 1-3/min. This can be achieved several ways: by
use of special instruments, by putting the vials in an alcohol-ice bath to
which frozen COz chips are added, or by simply standing the vials on a
552
S P E C I F I C CELL L I N E S
[49]
bed of frozen COs or, wrapped in paper towels, in a -75 freezer. For
greater security against contamination, sealed glass ampules may be substituted for the screw-capped type. After the cells are frozen, they are
transferred to a liquid N2 freezer; the lower temperature it provides is
essential for viability after long storage. HTC cells have been regrown
after more than 5 years of such preservation.
When frozen cells are to be grown, the vial is promptly brought to
ambient temperature or to 37 in a water bath and cultured without delay.
Glycerol-frozen cells can simply be diluted with fresh medium and grown.
DMSO-frozen cells should be removed from the toxic freezing medium
either by centrifugation as soon as the cells are thawed or by plating the
thawed cells with fresh medium and then replacing the medium again after
a few hours, when the cells have adhered to the substratum. Initial culture
in any case should be as monolayers, not suspension cultures. Cells grown
serum free can be frozen in serum-supplemented medium as above, or
15-20% polyvinylpyrrolidone can be substituted for serum.
[49] A n E s t a b l i s h e d b u t D i f f e r e n t i a t e d K i d n e y E p i t h e l i a l Cell
Line (MDCK)
[49]
553
and mucosal, which have distinct structures and enzyme markers. The
mechanisms by which these enzyme markers segregate to the two surfaces are not well understood. (3) What are the mechanisms by which
solutes are transported across kidney epithelia? Studies of kidney function using MDCK are not only feasible, but have the advantage over in
vivo studies that the cells in culture form a homogenous population that
can be cultured indefinitely; they can be grown readily to sizable populations for biochemical studies and are amenable to genetic analysis using
the techniques available to somatic cell geneticists. 4
Several studies have been conducted concerning the differentiated
processes in MDCK cells related to salt and water transport. That MDCK
cells transport solutes in a vectorial manner, from the mucosal to the
serosal surface, was first suggested by Leighton5 who observed the formation of multicellular blisters or domes in confluent MDCK cultures. The
blisters are groups of cells in the monolayer slightly raised from the tissue
culture dish surface. Microscopically, the cells in the blisters are out of
the focal plane of the majority of the cells in the monolayer. Blister formation can be accounted for by the transport of salt and water from the
mucosal surface of the cells (facing the culture medium), to the serosal
surface (facing the dish surface). Presumably, the solutes that have accumulated between the cell layer and the plastic surface give rise to
hydrostatic pressure, which~causes portions of the monolayer to be elevated from the dish surface, i.e., blister formation. This explanation is
supported by both morphological and electrophysiological studies.
A prerequisite for vectorial transport is that the cells have the structural polarity characteristic of transporting epithelia of the kidney; indeed, electron microscopic studies indicate that microviUi are localized
exclusively to the mucosal (upper) surface of the MDCK cells. 6 Adjacent
cells are interconnected by tight junctions. The vectorial transport of salt
and water by MDCK cells was first demonstrated by Misfeldt et al. employing a Ussing chamber. 6 They detected a spontaneous potential difference across the MDCK cell monolayer, as well as net water flux from the
mucosal to the serosal surface of the monolayer.6 More recent studies also
indicate that the cells vectorially transport certain amino acids, r
In addition to studies of differentiated functions, both the in vitro hnd
the in vivo growth characteristics of MDCK have been examined. In vitro,
MDCK cells have a saturation density typical of transformed cells, have a
4 L. H. Thompson and R. M. Baker, Methods Cell Biol. 6, 209 (1973).
5 j. L. Leighton, W. Estes, S. Mansukhani, and Z. Brada, Cancer 26, 1022 (1970).
D. S. Misfeldt, S. T. Hamamoto, and D. R. Pitelka, Proc. Natl. Acad. Sci. U,S.A. 73,
1212 (1976).
r M. White and D. S. Misfeldt, personal communication (1977).
554
[49]
[49]
555
take studies, confluent cultures are washed 3 times with the uptake buffer
[phosphate-buffered saline (PBS) or 1 mM Tris made isotonic to PBS using
sucrose], and the ceils are incubated with labeled solute at room temperature. After the uptake period, the solution is removed by aspiration, and the
monolayers are washed 3 times with 3 ml of incubation buffer. The labeled
material is solubilized by incubation for 1 hr in deionized water (for cation
transport) or in 5% trichloroacetic acid (for sugar or amino acid transport). The solubilized material is added to scintillation vials containing a
Triton-toluene based scintillation fluid, and the radioactivity is determined. Total label accumulated intracellularly is measured in duplicate
and is corrected for zero time uptake, i.e., for unincorporated material not
removed by the washing procedure.
The label present per cell population is standardized by either the
protein content per dish, determined using the Lowry protein assay as
modified for tissue culture by Oyama and Eagle, 12 or by the total cell
number per dish, determined using a Coulter counter. In order to determine the concentration of solute present intracellularly, the volume is
estimated by measuring the average MDCK cell size with a Coulter
counter. Pollen particles are used as standards, and a 75% water content
is assumed.13 The protein content is used to determine the intracellular
fluid volume per dish since a direct correlation has been made between
these two factors as a function of cell density, lz
Growth for Studies in Ussing Chamber
The transepithelial transport of salt and water by MDCK cells has
been studied with a Lucite Ussing chamber (Fig. 1) that was originally
developed for the measurement of the transepithelial electric potentials
generated by frog skin. For the Ussing chamber studies MDCK
monolayers are grown on Millipore filters, so that the monolayers resemble a continuous epithelium. Membrane filters (Millipore, HAMK 02512)
are boiled 5-10 min to remove the wetting agent and are attached to
plastic tissue culture dishes while still wet using droplets of Millipore
cement (Formulation No. 1). MDCK cells are distributed into the dishes
containing the filters and DME. When the cells on the filters have grown
to form a continuous epithelial sheet, a filter may be gently detached from
the dish and placed between Lucite Ussing chambers containing either
culture medium without serum or a balanced salt solution (for example,
Hanks's balanced salt solution). 6
The Ussing chamber (designed originally by Ussing and Zerahn 14) is
12V. I. Oyama and H. Eagle, Proc. Soc. Exp. Biol. Med. 9, 305 (1956).
13D. O. Foster and A. Pardee,J. Biol. Chem. 244, 2675 (1969).
14H. H. Ussing and K. Zerahn, Acta Physiol. Scand. 23, ll0 (1951).
556
[49]
FIc. 1. Diagrammatic sketch of Ussing chamber. Abbreviations used in the figure are as
follows: C, Celluloid or Lucite chamber containing salt solution; S, skin or membrane filter
with MDCK cells; a, air inlets; A and A ', Agar-Ringer bridges, connecting outside and inside
solutions with calomel electrodes and lx~tentiometer;B and B', Agar-Ringer bridges used for
applying outside emf; I.), battery; W, potential divider; P, tube potentiometer.
illustrated in Fig. 1. The epithelium is placed in a space b e t w e e n two
adjacent c h a m b e r s , t h e r e b y separating the solutions in these c o m p a r t ments. Saturated K C l - c a l o m e l electrodes are connected to the solutions
in the two c h a m b e r s b y means o f 1 M N a C I - 4 % agar bridges. A potent i o m e t e r is c o h n e c t e d to these half cells to determine the potential difference b e t w e e n the solutions facing either the mucosal or serosal sides of
the cell layer.
In applying this apparatus to the study of M D C K cells, a B e e k m a n
model 76 p H m e t e r is used as a p o t e n t i o m e t e r and the potential is recorded with a B e c k m a n model 1005 recorder. ~ The s y s t e m has to be
carefully balanced, i.e., the c o m p o n e n t s present on either side of the
bridges h a v e to be symmetrical, in order to m e a s u r e spontaneous potentials. Relatively high potentials induced b y external currents m a y be measured if a Simpson multimeter is placed in series with the potentiometer.
The spontaneous electrical potential of the M D C K cells has been determined w h e n identical solutions were present on either side of the Ussing chamber. 6 I m m e d i a t e l y after initiating the readings, relatively high
potentials are observed; a decline follows after 3-10 min. At this lower
steady-state level a potential of 1.42 ___ 0.26 m V with a negative serosal
surface was determined. Due to the wide range o f readings multiple determinations are n e c e s s a r y to obtain meaningful estimations of the sport-
[49]
557
558
[49]
[49]
559
filters (previously rinsed with buffer); the filters are washed with buffer to
remove unbound cAMP. To determine [all]cAMP bound, filters placed in
scintillation vials are dissolved with 1 ml of methyl cellusolve, scintillation
fluid is added (the ratio PPO/POPOP : methyl cellusolve is 3 : 1), and the
vials are counted in a scintillation counter.
Comments
MDCK has both the morphological and enzymic properties of epithelial cells from the distal tubule of the kidney.1 In addition to having high
Na+/K ATPase activity," MDCK cells possess the kidney-specific enzymes alkaline phosphatase and leucine aminopeptidase. The peptidase
has been localized primarily to the mucosal membrane by immunofiuorescence. While the cells lack proximal markers (Na*-dependent sugar
transport, p-aminohippurate transport; disaccharidase), they do have a
distinctive distal marker, responsiveness to the antidiuretic hormones.
When present at physiological concentrations, lysine and arginine vasopressin as well as oxytocin stimulate cAMP production by adenylate cyclase. TM Similarly, in the presence of PGE1, PGE2, glucagon, or cholera
toxin cAMP production is enhanced in MDCK. The stimulation of adenylate cyclase by vasopressin is apparently subject to negative cooperativity; however, these studies indicate that the cells have antidiuretic hormone (ADH) receptors specific to distal kidney cells, and that a single
kidney epithelial cell type is responsive to multiple hormonal agents.
When ADH is present in the distal portion of the kidney, increased
water flux from the kidney lumen into the mucosal surface of the cells is
observed.17 If the MDCK cells provide a good model system for the study
of kidney epithelial function one would expect that the cells would respond to treatment with vasopressin (or to dibutyryl cAMP since the
vasopressin response is cAMP mediated) by increased rates of water
transport. That cAMP levels in MDCK cause increased vectorial fluid
transport is suggested by the observation that incubation of MDCK with
dibutyryl-cAMP results in increased blister formation.l These observations indicate that the MDCK cells provide a novel model system for the
study of distal tubular cell function.
The primary function of distal cells is to transport salt and water,
thereby regulating body levels of these compounds. Misfeldt's experiments indicate that in MDCK cells, as in the kidney, salt and water
16M. J. Rindler, L. M. Chuman,and M. H. Saier, Jr., in preparation.
17E. Koushanpour,"Renal Physiology,Principlesand Functions,"p. 336. Saunders,Philadelphia, Pennsylvania,1976.
560
S P E C I F I C CELL L I N E S
[SO]
transport occur transepithelially, from the mucosal to the serosal surface. ~ The transport of Na + by the Na + channel (presumably located on
the mucosal surface) has been examined, both kinetically and genetically, is 22Na+ uptake studies indicate that a saturable Na + transport system is present in MDCK cells that is energy independent and insensitive
to ouabain inhibition. 18 Na + uptake by the channel is inhibited by a number of monovalent cations including Li +, K +, Rb +, guanidine, and
amiloride. 22Na+ uptake studies indicated that the system is Subject to two
exchange processes: (1) exchange transport with intracellular Na +, and
(2) Na+/proton antiport. Calcium is implicated as an important regulatory
molecule for the channel, inhibiting Na + uptake when present extracellularly, and stimulating Na + uptake when present intracellularly.
Variant clones of the MDCK line have been isolated by their resistance
to killing by 5 10-4 M amiloride. 18 Since the clones are stable over a
6-month growth period under nonselective conditions, and since their
frequency is increased by mutagens, they appear to have arisen by genetic
mutations. The clones exhibit reduced uptake of Na +, amiloride, and Rb +
(ouabain-insensitive uptake). Since both Rb + uptake by the Na+/K
ATPase and a-amino-isobutyrate uptakes are not affected by the mutation,
the decreased ionic uptake rates may be tentatively explained as resulting
from a mutation affecting the Na channel. The reduced uptake of
amiloride by the Na channel in the resistant cells may confer resistance
to killing by amiloride by decreasing the intracellular concentration of the
drug below the cytotoxic level. Reduced salt transport in these cells apparently results in a decreased propensity for vectorial fluid transport, as
indicated by the lack of blister formation in the variant cells.
is M. Taub, M. S. Rindler, and M. H. Saier, Jr., Cell Biol. 75, 367a (1977).
[50] L a r g e - S c a l e P r e p a r a t i o n o f C h o n d r o c y t e s
By MICHAEL KLAGSBRUN
[50]
PREPARATION OF CHONDROCYTES
561
562
[50]
Both scapular and articular cartilage are diced into pieces approximately 0.5 cm 0.5 cm and washed twice with phosphate-buffered saline
(Gibco) containing penicillin (200 U/ml) and streptomycin (200 /zg/ml).
The diced cartilage is incubated at 37 in 25 ml of 2 mg/ml clostridial
collagenase (Worthington CLS II, 140 U/mg) made up in the phosphatebuffered saline--penicillin-streptomycin mixture. The incubation is carried
out in a 60-mm petri dish that is gently rotated on a rotating apparatus
(Arthur Thomas Co.) for 12-24 hr or until the pieces of cartilage are
completely dissociated and the chondrocytes are liberated from the matrix. The suspension of chondrocytes is passed through a 153-/zm nylon
sieve (Tetko) to remove debris and undissolved cartilage fragments. The
cells are subsequently washed twice with the phosphate-buffered salinepenicillin-streptomycin mixture by means of centrifugation. After the
second wash, cells are counted in a Coulter counter (Coulter Electronics).
A typical scapular or articular chondrocyte preparation contains
1-4 108 cells per shoulder.
A source of large numbers of human chondrocytes is the costal cartilage obtained from children undergoing surgical correction of pectus excavatum, a disorder in which rib cartilage is deformed. These chondrocytes are particularly easy to isolate because the perichondrium, muscle,
and connective tissue are removed from the cartilage during surgery.
Otherwise, the isolation procedure is the same as for the bovine
chondrocytes.
For cell culture, cells (3 106 per 75 cm 2 culture dish) are plated
sparsely in the presence of 10 ml of Dulbecco's modified Eagle's medium
containing 4.5 goiter glucose (Gibco), 10% calf serum (Colorado Serum
Co.), penicillin (50 U/ml), and streptomycin (50/zg/ml) and transferred to
a 37 water-jacketed COz incubator (National Appliance Co.). Chondrocytes are round cells in situ. They gradually flatten in the culture dish over
a period of 2-4 days; growth begins after this initial lag period. In confluent cultures, two patterns of growth are observed; some cells in the dish
grow in a monolayer while others pile up and form nodules. The number
of nodules increases with repeated feeding of the cells. If the cells are
stained with 2% aqueous toluidine blue, metachromasia may be observed
in the nodules, indicating that active synthesis of proteoglycans has taken
place.
Chondrocytes cannot be maintained indefinitely in culture flasks once
they become confluent because the synthesis of the sulfated proteoglycans leads to a sharp decrease in the pH of the media. The cells are
harvested and either subcultured in new flasks by plating sparsely or are
frozen. Harvesting of chondrocytes is accomplished by detaching the
cells with 0.1% trypsin (Gibco) made up in phosphate-buffered saline
lacking calcium and magnesium (Gibco). The medium is removed from a
[50]
PREPARATION OF CHONDROCYTES
563
T-75 culture flask, and 10 ml of 0.1% trypsin solution are added. The cells
are incubated with trypsin at room temperature for about 5 min or until
they round up and are loosely attached to the flask. The trypsin is carefully removed, and 10 ml of fresh media are added. The flask is struck
several times with the palm of the hand in order to detach the cells.
Chondrocytes are frozen in a solution of Dulbecco's modified Eagle's
medium containing 10% calf serum and 7.5% dimethyl sulfoxide (Fisher
Scientific Co.). A stock solution of dimethyl sulfoxide (15%, v/v) in Dulbecco's modified Eagle's medium containing 10% calf serum is prepared.
One milliliter of the dimethyl sulfoxide solution is mixed with 1 ml of
chondrocytes (2.5 10~ cells/ml) resuspended in Dulbecco's modified
Eagle's medium containing 10% calf serum. The cells are transferred into
2-ml ampules (Wheaton Scientific) that are subsequently sealed. The ampules are gradually frozen at - 9 0 in a Revco freezer and are stored either
in the Revco freezer or in a liquid nitrogen tank (Cryogenics-East Co.).
Bovine chondrocytes will remain viable in liquid suspension culture
for at least 7 days. Maintenance in liquid suspension culture can be used
as a technique to remove contaminating fibroblasts since these cells will
not survive in suspension for more than 2 or 3 days. 8 One technique for
keeping chondrocytes in liquid suspension is to coat the bottom of a petri
dish with 0.5% agar. The chondrocytes are unable to plate out in the agar
and remain suspended. Agar plates are prepared as follows: A 1.0% solution of agar (Difco Bacto-agar) is made up in water and sterilized by
boiling for 30 min. The 1% agar is cooled to 45 and mixed with an equal
volume of warm 2 basal Eagle's medium with Earle's balanced salts
(Microbiological Associates) containing 20% calf serum, penicillin (100
U/ml), and streptomycin (100 /zg/ml). Two milliliters of the 0.5% agar
solution are poured into a 35-mm petri dish. After the agar hardens, 3 ml
of chondrocytes (2 l0 s cells/ml) are resuspended in Dulbecco's modified Eagle's medium containing 10% calf serum and plated onto the agar.
The agar plates are maintained at 37 in a water-jacketed CO2 incubator.
A major problem in chondrocyte culture is the possibility of contamination by fibroblasts arising from the perichondrium. Nevo e t al. 8 have
suggested that the fibroblasts can be eliminated by maintaining the chondrocyte preparation in suspension culture for 2 or 3 days. Fibroblasts do
not survive in suspension culture.
In general, chondrocyte cultures can be characterized by several criteria. These include a high level of ~5SO4 uptake into chondroitin sulfate, 8
metachromatic staining with toluidine blue, 9 and the ability of chondrocytes to survive in suspension culture, r-a
Cartilage in v i v o contains Type II collagen that is genetically distinct
from the Type I collagen found in skin, bone, and tendon. Thus, chondrocytes should be the only cell capable of producing Type II collagen. How-
564
[51]
[5 1] M e l a n o m a Cells
B y JOHN PAWELEK
Melanocytes synthesize melanin in a reaction controlled by the enzyme monophenyl, dihydroxyphenylalanine : oxygen oxidoreductase (EC
1.14.18.8), commonly referred to as either tyrosinase or dopa-oxidase.
Melanocytes in the skin of adult mammals divide infrequently, perhaps
once a year, and attempts at adapting these cells to culture have met with
only limited success. On the other hand, malignant melanoma cells divide
as often as once a day, and several lines have been established in culture.
Commonly used lines are the B16,1 Harding-Passey, 2 and Cloudman3
strains (mouse); the Fortner (RPM1) strain 4 (hamster); and many human
strains. 5"6 Some of these lines are available commercially. 7,s
The Cloudman $91 melanoma was established as a transplantable
tumor in DBA 2/J mice in the 1930sa and was adapted to culture in the
1960s. 3 In 1973 it was reported that the Cloudman cells responded in
culture to melanotropin (MSH) with increases in tyrosinase activity,
melanin formation, and generation time, as well as changes in morphology. ~ Since the original findings, many studies have been carried out on
factors that regulate pigmentation and proliferation of these and other
melanoma cells in culture (for reviews, see Pawelek~l'~2).
i F. Hu and R. R. Cardell, J. Invest. Dermatol. 42, 67 (1964).
H. E. Harding and R. D. Passey, J. Pathol. 33, 417 (1930).
3 y. Yasumura, A. H. Tashjian, and G. H. Sato, Science 154, 1186 (1966).
4 G. E. Moore, D. F. Lehner, Y. Kikuchi, and L. A. Less, Science 137, 986 (1962).
5 R. E. Gerner, H. Kitamura, and G. E. Moore, Oncology 31, 31 (1975).
6 L. A. Quinn, L. K. Woods, S. B. Merrick, M. Arabasz, and G. E. Moore, J. Natl. Cancer
Inst. 59, 301 (1977).
7 The American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland.
a The Jackson Laboratory, Bar Harbor, Maine.
g A. M. Cloudman, Science 93, 380 (1941).
lo j. Pawelek, G. Wong, M. Sansone, and J. Morowitz, Yale J. Biol. Med. 46, 430 (1973).
11 j. Pawelek, J. Invest Dermatol. 66, 201 (1976).
12 j. Pawelek, Pigm. Cell 4 (in press).
M~THODS
IN F_MZYMOLOOY,VOL.LVIII
Copyright1979by AcademicPress,Inc.
All rights of reproductionin any form reserved.
ISBN 0-12-181958-2
[51]
MELANOMA CELLS
565
The purpose of this article is to describe techniques for culturing Cloudman $91 in monolayers and clones, and to outline some of the problems
unique to melanin-producing cells that are encountered in normal culturing procedures and in some biochemical analyses.
Cell Culture
Culture procedures are straightforward. For monolayers, cells are
placed into plastic flasks or petri dishes that have been coated for cell
culture (available commercially). The culture medium consists of H a m ' s
F10 medium 13 supplemented with 2% fetal calf serum and 10% horse
serum. Antibiotics can be added if desired, but it is important to first
determine whether a specific antibiotic has an effect on pigmentation. For
example, tetracycline and bacitracin both increase pigment formation
whereas streptomycin and penicillin do not. Fresh culture medium is
added 3 times per week. Subculturing of cells involves removal of the
medium and addition of ethylenediaminetetracetic acid (EDTA) (1 m M or
less) in a Ca- and Mg-free balanced salt solution. After about 10 min at
room temperature, cells detach from the culture vessel and are collected
by centrifugation. The use of trypsin or other proteases should be avoided
in the harvest procedure, particularly if regulatory processes involving
the outer cell surface are being studied.
For culturing cells in clones, we use the "dilute agar colony m e t h o d "
of Chu and Fischer. TM This involves culturing cells in medium supplemented with 0.12% agar; the concentration of horse serum is increased to
15%. Culture vessels can be in a variety of sizes and shapes, but for
convenience we use plastic screw-cap test tubes. The clones grow in
suspension, and it is therefore unnecessary to use vessels that have been
coated for cell culture. Clones are generally visible to the naked eye after
1 week and can be picked with a Pasteur pipette after 2-4 weeks.
566
[51]
Total
clones
Amelanotic
variants
Relative
survival (%)
0
0.02
0.05
0.10
0.20
0.50
1.0
6110
6400
2292
1846
1438
100
0
0
0
11 (0.5%)
51 (2.7%)
27 (1.9%)
5 (5 %)
0
100
i05
38
31
24
1.7
0
a Cells in monolayer were incubated with ethylmethanesulfonate (EMS) at the concentrations indicated. After 2 hr the EMS was removed, the cells were rinsed several
times with fresh culture medium, and allowed to pass through three generations (6
days). Cells (3 x 104) from each category were then cultured in petri dishes (100-mm
diameter) by the method of Chu and Fischer.~3 After 4 weeks the total number of clones
and the number of amelanotic variants were determined by counting with the aid of a
dissecting microscope.
[51]
MELANOMA CELLS
567
Several years ago, Lerner suggested that in the process of melanization pigment cells produce substances that are potentially autotoxic. TM
Later it was demonstrated that pigmented cells are killed when exposed to
excess tyrosine or dopa in the culture medium.l.1. ~ It has been shown
recently that among all the naturally occurring amino acids, only tyrosine,
dopa, and trytophan are toxic to pigmented cells. The toxicity of these
agents is proportional to the amount of tyrosinase activity within the cells.
5,6-Dihydroxyindole is far more toxic than tyrosine, dopa, or tryptophan,
and it is likely that this ~elanin precursor is responsible for cell death. 2
Although these findings may be potentially useful for chemotherapy of
melanoma, they present a problem when culturing melanoma cells. It is
important to choose a culture medium that is low in tyrosine, and it is for
this reason that Ham's F10 medium (tyrosine concentration is 2 rag/l)
was used for the Cloudman cells. Most other media have tyrosine
concentrations that are orders of magnitude, higher than that in Ham's
F10, and cells with high tyrosinase activity usually do poorly in these
media.
568
[SI]
Tyrosinase Activity
A simple and convenient assay for tyrosinase activity was developed
by Pomerantz. 21 Tritiated 3,5-tyrosine is available commercially. When
this molecule is oxidized to dopa by tyrosinase, [ZH]H20 is formed and is
separated from unreacted [ZH]tyrosine by the addition of activated charcoal. A second purification of [ZH]H~O consists of passing the material
that does not bind to charcoal through a Dowex-50 column. 22 Tyrosinase
can be assayed in living cells by adding [3H]tyrosine to the culture medium. The cells actively take up the tyrosine and release [ZH]H~O to the
culture medium. Thus by removing small amounts of medium and measuring [3H]H20, one can measure tyrosinase activity in the cells. Experiments of this sort must be interpreted with caution, however, because
[3H]H~O released to the culture medium is not.only a function of tyrosinase
activity but also of the rate of transport of [ZH]tyrosine into the cells, the
preexisting pool of unlabeled tyrosine within the cells, and the amount of
[ZH]tyrosine being diverted from melanin synthesis to protein synthesis.
Nonetheless, it is frequently useful to measure [ZH]H~O produced by
living cells as an assay of tyrosinase activity. To avoid the problems
mentioned above, one should then compare the results in living cells with
direct measurements of tyrosinase activity in cell homogenates. A typical
assay in cell homogenates has been described in detail. 2~
Normalization of Data
The colorimetric method of Lowry e t al. ~ for measuring protein is
used widely for the normalization of data. Unfortunately this technique
should not be employed with crude extracts of melanin-producing cells
because melanin reacts 2-3 times more strongly than bovine serum albumin (fraction V) with the Folin reagent that. is used. It is difficult and
time-consuming to quantitatively separate melanin from protein, and for
purposes of normalizing data we have found it more convenient to estimate DNA content by using Button's modification of the diphenylamine
reaction, 24 or to measure cell number in a Coulter counter.
Isolation of RNA
Melanin interferes with the isolation of RNA because it binds tightly
to RNA molecules. We found this to be a particularly vexing problem
zl S. Pomerantz, Science 164, 838 (1969).
22 A. Korner and J. Pawclck, Nature (London) 267, 444 0977).
2z O. H. Lowry, A. L. Rosenbrough, and R. J. Randall, J. Biol. Chem. 248, 190 (1951).
24 K. Burton, Biochem. J. 62, 315 0956).
[51]
MELANOMA CELLS
569
570
[52]
Melanin in the cells can be detected visually and thus provides an easy
assay for the expression of a differentiated function. Isogenic pigmented
cell lines can be established by clonal techniques, and from these lines
amelanotic variants can be isolated. Since at least some of these variants
should have lesions in the genes controlling melanin biosynthesis, a genetic analysis of this differentiated function should be possible. The
Cloudman melanoma line has been found to respond dramatically to
MSH, and studies with these cells promise to provide much information
concerning the mechanism of action of this hormone. It is obvious that the
factors regulating pigmentation and proliferation of melanoma cells are
complex. The molecules regulating these functions include hormones affecting cyclic AMP levels, the corresponding hormone receptors, adenylate cyclases, phosphodiesterases, cyclic AMP-dependent protein kinases
and their phosphoprotein Substrates, and possibly phosphoprotein phosphatases. Also to be considered are structural and regulatory genes that
control the synthesis of these molecules. Certainly this is an abbreviated
list that should expand rapidly as research continues.
[52] A d r e n o c o r t i c a l Y1 C e l l s
By BERNARD P. SCHIMMER
The Y1 adrenal cell line was developed by Sato and his colleagues I as
one of a series of endocrine cell cultures that retain many of their tissuespecific characteristics. 2 This cell line originated from a transplantable,
adrenocortical tumor of an LAF1 mouse; a it was cloned in 1964 after
adaptation of the tumor to culture conditions by the technique of alternate
passage between culture and animal. 4 The cell line currently is available
from the American Type Culture Collection (No. CCL 79).
The Phenotype and Its Stability
The Y1 cell line has an average doubling time of 30-40 hr, has a plating
efficiency5 of 4-10%, and reaches saturation density at approximately
2.7 l05 cells/era 2. Its morphology is epithelial-like in monolayer culture,
and its karyotype is nearly diploid with a modal number of 39 acrocentric
Y. Y a s u m u r a , V. Buonassisi, and G. Sato, Cancer Res. 26, 529 (1966).
z y. Y a s u m u r a , Am. Zool. 8, 285 (1968).
A. I. Cohen, E. Bloch, and E. Cellozi, Proc. Soc. Exp. Biol. Meal. 95, 304 (1957).
4 V. Buonassissi, G. Sato, and A. I. Cohen, Proc. Natl. Acad. ScL U.S.A. 48, 1184 0962).
5 Plating efficiency is defined as the percentage o f single cells plated which grow into
colonies.
METHODS IN ENZYMOLOGY,VOL. LVIII
[52]
ADRENOCORTICAL Y 1 CELLS
571
572
IS2]
Methods
All operations are carried out using strict aseptic bacteriological procedures. Growth medium and other reagents are sterilized before use by
filtration through sterile membrane filters (0.2 /zm pore size). Cultures
should be screened regularly for aerobic and anaerobic contaminants ineluding pleuropneumonia-like organisms (PPLO).
Initiation of Stock Cultures. Stock cultures of YI cells are grown as
monolayers in plastic tissue culture flasks (75 cm ~ area). Dispersed cells
(approximately 2 x 106) are added to 15 ml of growth medium, and the
suspension is gently rocked over the growing surface of the culture flask
to effect a uniform distribution of cells. The inoculated flasks are capped
loosely and maintained at 36.5 in a humidified atmosphere of 5% CO2 in
air. The cells attach and begin to spread out on the plastic surface within
2-3 hr. Culture medium is changed every third or fourth day, and the
progress of the culture is monitored visually at these intervals with an
inverted, phase~contrast microscope. Spent growth medium is removed
by aspiration, and fresh medium is added immediately, with care taken to
ensure that the cell monolayer is not disturbed. At the second medium
change, the volume of growth medium is increased to 30 ml per flask.
Cells reach saturation densities of 2 10r cells per bottle after 10-12
days. Cells can be maintained at saturation density for at least I week if
the growth medium is replenished regularly.
Subculture. When the stock culture reaches saturation density, the cells
are subcultured by treatment with a solution of Viokase (Viobin Corp.,
Monticello, Illinois). Growth medium is removed, and the monolayer is
rinsed 14 twice with 2-ml portions of the Viokase solution (0.1% in
phosphate-buffered saline 1~ without Ca 2+ or Mg2+ salts) and incubated at
10 D. Gospodarowicz and H. H. Handley, Endocrinology 97, 102 (1975).
~4 Rinsing is effected by gently rocking the solution over the monolayer surface and then
aspirating the liquid.
15 R. Dulbecco and M. Vogt, J. Exp. Med. 99, 167 (1954).
[52]
ADRENOCORTICAL Y1 CELLS
573
36.5 for 10 min. The residual Viokase solution coating the cells is sufficient to detach the cells from the monolayer. Treatment with Viokase is
complete when gentle tapping of the flask dislodges the cells. The cells are
washed from the flask wall with 10 ml of growth medium and are dispersed by pipetting repeatedly. One milliliter of fully dispersed cells, i.e.,
2 106 cells, is used to inoculate each new stock flask. Since the cells
settle rapidly, they must be resuspended immediately before each transfer. An Erlenmeyer flask and magnetic spin bar can be used to suspend
cells where a large number of uniform transfers are required. The cells are
stirred at a moderate speed over a nonheating stirrer.
The size of the culture vessel and the size of the inoculum can be
altered over a wide range as required by the investigator. From the data
given for plating efficiency, growth rate, and saturation density of the cell
line, the state of the culture can be approximated at any time after plating.
Storage. Y1 cells can be stored frozen in liquid N2 for at least 8 years in
growth medium supplemented with 10% glycerol. Cells, 2 106 in 1 ml of
growth medium containing glycerol, are placed in sealed ampules, packed
in a cardboard container, kept at - 7 0 for 3.5 hr, and transferred quickly
to liquid N2. To recover cells from the frozen state, one vial is thawed
rapidly in a 37 water bath, suspended by pipetting, and divided between
two tissue culture flasks (75 cm 2) containing 15 ml of growth medium.
Cells are ready for subculture after approximately 3 weeks of growth.
Plating of Adrenal Tumors. YI cells also can be grown as tumors and
studied in primary cell culture. Cells, 105--106 in 0.1 ml saline, injected into
the hind leg muscle of an LAF1 mouse will give rise to a tumor in approximately 6-8 weeks. To harvest the tumor, the mouse is killed by cervical
dislocation and cleaned with 70% ethanol. The tumor and associated leg
tissues are dissected free, transferred to a Petri dish, and washed several
times with saline solution. The tumor capsule is cut, and the tumor tissue
is removed and minced with scissors. The fine mince is triturated by
pipetting with 25 ml of Viokase (0.2% solution in nutrient mixture F10) in
5-ml portions, transferred to an Erlenmeyer flask with spin bar, stirred
gently for 20 min at room temperature, and centrifuged in conical centrifuge tubes at about 200 g for 10 min. The pellet is washed once and
resuspended in complete growth medium. Residual large tissue fragments
are allowed to settle, and the dispersed cells are collected for plating.
Cells from one tumor distributed among 40-60 tissue culture plates
(60-mm diameter) will approach confluence after 2 weeks in culture.
Propagation of Y1 cells as tumors minimizes the requirements for maintaining stocks in cell culture and also provides a convenient means of
generating cells in large yields. These advantages, however, are offset to
some degree by the heterogeneity of the tumor material.
574
SPECIFIC
CELL
LINES
[53]
Caution
Although experience to date suggests that there is no biohazard associated with the culture of these mouse adrenal tumor cells, it seems prudent to adopt containment procedures similar to those of the Medical
Research Council of Canada 16 when handling them.
16 "Guidelines for the Handling of Recombinant DNA Molecules and Animal Viruses and
Cells," Cat. No. MR21-1/1977. Minister of Supply Services, Ottawa, Canada, 1977.
[53]
DISSOCIATED S Y M P A T H E T I C N E U R O N S
575
576
[53]
low) and cleaned free of all surrounding tissue. The ganglion cells are then
dissociated mechanically using very fine watchmaker's forceps (#5).
Usually the ganglion sheath is first peeled away with forceps, and the
ganglion cells are then gently teased apart into smaller groups of cells. The
chunks and cells are triturated by several passages through a stainlesssteel hypodermic needle (22 or 23G, 1.5 inch) using a 5-ml disposable
plastic syringe. The resulting suspension is further agitated with a Vortex
mixer for 3-5 min (avoiding any swirling action) and undissociated pieces
removed by sedimentation or filtration. The suspension is either allowed
to settle undisturbed for 5-10 min or filtered, e.g., with a 10/xm Nuclepore filter (Nuclepore Corp., Pleasonton, California), to remove undissociated fragments. The dissociated cells are centrifuged (3-5 min at
800 g), resuspended, and plated in modified culture dishes (see below) by
adding 1 or 2 drops per dish of cells, suspended in plating medium (see
below). In general, the overall yield of neurons in culture ranges from
1-10% based on the number of neurons found in adult rat superior cervical ganglion (approximately 25,000). Normally, 40 to 50 dishes of about
3000 to 5000 viable neurons each are prepared from 40 pups. In addition to
the mechanical procedures described here, sympathetic neurons have
also been dissociated with trypsin treatment) '6
Culture Dishes
Falcon petri dishes (35 ram, #1008) are modified by boring a 1-cm hole
in the bottom of the dish and affixing a polystyrene (Lux Scientific Corp.,
Newbury Park, California; #5407, 25 mm) tissue culture cover slip with
paraffin to form a shallow well. This well protects the cells from movement of the medium, is convenient for isotopic incubations, provides better optics for inverted phase microscopy, and facilitates preparation for
electron microscopy. Prior to plating, a glass ring (1.5-cm diameter microslide ring, #6705-R12, A. H. Thomas, Philadelphia, Pennsylvania) is
placed in each dish surrounding the center well. The ring is added after
the growth medium (see below) and serves to restrict the area to which
the cells can settle to that of the coverslip. After 2 days of culture these
rings are removed.
Culture Surfaces
Several substrata are suitable for attachment and extension of
neuronal processes. Most commonly, a thin, clear film of collagen is used.
S. Varon and C. Raiborn, J. Neurocytol. 1,211 (1972).
6 K. D. McCarthy and L. M. Partlow, Brain Res. 114, 391 (1976).
[53]
577
578
[53]
[53]
579
glass bottles. (Glassware is never cleaned with detergent; if used for proteinaceous solutions, it is discarded after use. Bottles are cleaned by
autoclaving them full of water followed by repeated rinsing while still
hot.)
2. L-15-C02. The medium used both for sympathetic neurons and
nonneuronal cells is prepared by adding 170 ml of 150 mM NaHCOa for
every 850 ml of basal L-15-Air (see above). (The sodium bicarbonate is
sterilized by filtration either before addition to sterile L-15-Air or in combination with the L-I5 medium). The pH of this medium is adjusted by
blowing COz over the solution. In general, most solutions are sterilized by
passage through autoclaved Nuclepore filters (0.2 /zm) washed as described above.
3. Plating Medium. The medium used for ganglion dissection and cell
dissociation consists of 100 ml of basal L-15-Air to which is added glucose
(4 ml of a 30% w/v solution), glutamine (1 ml of a 200 mM solution;
Microbiological Associates, Inc., Walkersville, Maryland), penicillin, and
streptomycin (1 ml of a solution containing 10,000 U of penicillin and 10
mg of streptomycin per milliliter; obtained as lyophilized powder from
Grand Island Biological Co., Grand Island, New York). Plating medium
contains twice the normal glucose concentration so as to increase the
density of the suspending medium and facilitate cell settling during
plating.
4. Complete Growth Media. For routine growth of cells in either
L-15-Air or L-15-CO2, the following additives are combined with 100 ml of
the basic formulation: glutamine, penicillin, and streptomycin (same as
for "Plating Medium"), glucose (2 ml of a 30% w/v solution), fresh vitamin mix (1 ml, see below), adult rat serum (5 ml, see below), Methocel
(0.6 g; see below), and nerve growth factor (NGF; final concentration of
1/~g/ml 7S, see below). Complete media are stored for no longer than 10
days at 4. L-15-Air cultures are incubated in a humidified air atmosphere
at 36 whereas L-15-CO2 cultures are incubated in a 5% CO2 in air
humidified atmosphere, also at 36. The growth medium (2 ml) is changed
every 4 days except for cultures receiving conditioned medium (see below) which receive fresh medium every 2 days. Rat sympathetic neurons
also have been grown in media based on commercial preparations other
than L-15 and with additives different from those described here. 18-2
5. Growth Medium Additfi, es. The fresh vitamin mix is prepared as a
100-fold concentrate and contains I mg of 6,7-dimethyl-5,6,7,8-tetrahydropterine (Calbiochem, La Jolla, California), I00 mg ascorbic acid
18 R. P. Bunge, R. Rees, P. Wood, H. Burton, and C.-P. Ko, Brain Res. 66, 401 (1974).
19 C.-P. Ko, H. Burton, and R. P. Bunge, Brain Res. 117, 437 (1976).
20 K. J. Lazarus, R. A. Bradshaw, N. R. West, and R. P. Bunge,Brain Res. 113, 159 (1976).
580
[53]
[53]
581
only the neurons survive; less than 10% of the cells seen by phase microscopy are nonneuronal even after several weeks in culture. 4
When dissociated ganglion cells are plated into L-15-CO2, both
neurons and nonneuronal cells become established in culture. The nonneuronal cells proliferate and form a monolayer within 2-3 weeks. Growth
of nonneuronal cells in L-15-CO2 cultures can be prevented by the use of
antimitotic poisons such as cytosine arabinoside or fluorodeoxyuridine.
Both of these inhibitors reduce the proliferation of ganglionic nonneuronal
cells without producing any reduction in neuronal catecholamine synthesis. Sympathetic cultures are incubated in growth medium containing 10
/zM cytosine arabinoside (Sigma) for 2 periods of 48 hr each, beginning on
days 2 and 6 in culture. Fluorodeoxyuridine and uridine (Sigma) are also
antimitotic when added at 10/~M on the same schedule.
In addition to these methods, nonneuronal cells may be removed at a
stage prior to plating by cell fractionation procedures. One method successfully employed for this propose in cultures of chick sympathetic
neurons involves differential cell adhesiveness and takes advantage of the
fact that, under appropriate conditions, chick ganglionic nonneuronal cells
attach to the substratum sooner and adhere tighter than the neuronal cell
population. ~,6
Co-Culture of Neurons with Nonneuronal Cells
582
[53]
[53]
583
584
SPECIFIC
CELL
[54]
LINES
and identified by high-voltage paper electrophoresis.'7 Furthermore, uptake, storage, and evoked release of neurotransmitters can be assayed, z7
The biosynthetic enzyme, choline acetyltransferase, also can be measured in extracts of sympathetic cultures, and levels of this enzyme are
elevated in cultures induced for cholinergic function. 4
Acknowledgments
Part of the work described here was supported by the American and Massachusetts
Heart Associations, the Helen Hay Whitney Foundation, and the National Institute of
Neurological and CommunicableDiseases and Stroke.
27p. H. Patterson, L. F. Reichardt, and L. L. Y. Chun, Cold Spring Harbor Symp. Quant.
Biol. 40, 389 (1975).'
[54] N e u r o n a l C e l l s f r o m R o d e n t N e o p l a s m s
By DAVID
SCHUBERT
and
WILLIAM
CARLISLE
[54]
585
586
[54]
Reagents
Tissue Culture Medium
For both historical and practical reasons all of the culture work on
cell lines derived from NEU-induced CNS brain tumors has employed
modified Eagle's medium 9 containing 10% fetal calf serum. With one exception, the PC12 sympathetic neuron-like cell line, 4 all cell lines that we
have tried will grow in this medium, even if they have been originally
adapted to other types of culture media and serum. This standardization
of culture conditions simplifies medium preparation and serum testing
and, in addition, provides a common culture regime by which the various
cell lines can be compared. Modified Eagle's medium can be purchased in
powdered or liquid form, but we have found that preparing it ourselves is
necessary both to maintain high quality and to provide the flexibility
needed in altering its composition for experimental purposes. (The prepr H. Druckrey, R. Preussmann, S. Ivankovi~, and D. Schmahi, Z. Krebsforsch. 69, 103
(1967).
a A. Hirano, J. Hasson, and H. M. Zimmerman, Lab. Invest. 27, 555 (1972).
a M. Vogt and R. Dulbecco, Proc. Natl. Acad. Sci. U.S.A. 49, 171 (1963).
[54]
587
Serum
There is a tremendous variation in the fetal, calf serum obtained from
different commercial sources and between lots from the same dealer.
Therefore, samples from several lots are tested for their ability to support
the growth and differentiation of different cell types. Once a lot is decided
upon; sufficient serum from that lot is purchased to last from 6-12 months.
It is stored frozen ( - 2 0 ) and dispensed in 10-ml aliquots for use over a
2-week period. The dispensed serum is stored at - 2 0 and only thawed
once immediately before use.
Culture Dishes
Many cell types are anchorage dependent for growth and require a
tissue-culture dish surface. Although most of the nerve cell lines will grow
in plastic petri dishes, where cells attach very poorly, all of the nerve cell
lines are routinely passaged on tissue culture dishes; 60-ram plastic dishes
are used for most purposes.
588
[54]
[54]
589
less desirable cells that will eventually overgrow the primary explant cultures. Many "ringed" cultures should be made, for all do not grow.
Once these "ringed" cultures begin to divide and appear somewhat
homogeneous, they should be transferred at high cell densities (1 : 5 split
of a confluent dish) along with conditioned medium for several transfers.
Assuming the cells are growing, they are ready for cloning.
Cloning
There are several methods for cloning cells (see this volume [12]).
Since the CNS-derived nerve and glial cells tend to grow attached to the
tissue culture dish, the method most frequently employed is that in which
cloning rings are used. Exponentially dividing cultures are dissociated to
single cells with a 1 : 4 dilution of Viokase for 1 hr at 37, and plated
between 102 and 104 cells per 100-mm tissue culture dish. If the plating
efficiency is low, the use of 50% conditioned medium may be helpful.
After 24 hr, well-isolated single cells are located on an inverted microscope and marked from below with a felt-point pen. Between 1 and 3 weeks
later, the plates are examined again for cell growth in the marked areas. If
a small colony has formed and remains isolated from other cells, then it
should be removed with a cloning ring and placed in 35-mm tissue culture
dishes. To be sure of the clonal origin of a cell line this procedure should
be repeated once. At this stage, the clones are grown in a dozen 60-mm
dishes, and the cells are stored in liquid nitrogen to preserve the lines.
Handling of Clonal Cell Lines
590
[54]
Shipment of Cells
If cells are to be shipped in the mail, they are placed in sealed tissue
culture bottles filled to the top with medium so that no air bubbles are
present and shipped via regular air mail.
AUTHOR INDEX
591
Author I n d e x
Numbers in parentheses are footnote reference numbers and indicate that an author's
work is referred to although his name is not cited in the text.
A
592
AUTHOR INDEX
AUTHOR INDEX
Brown, B. L., 60
Brown, J. M., 126
Brown, M. S., 444
Bruce, W. R., 151
Bruchovsky, N., 256
Brunsting, A., 235
Bryant, J. C., 14, 60, 142, 203, 211
Buckingham, M. E., 512
Buckler, C., 296
Buckley, P. A., 518, 519(22), 524(22)
Buell, D. N., 257
Biittner, W., 425, 426(3)
Bui-Dang-Ha, D., 367
Bullard, J. C., 130, 513
Bulloch, K., 585
Bunge, R. P., 579
Buonassisi, V., 97, 528, 570
Burger, M. M., 368
Burges, J., 364
Burk, R. D., 346
Burk, R. R., 265
Burke, D. C., 294
Burstin, S. J., 254
Burton, H., 579
Burton, K., 568
Butcher, G. W., 349, 351(26)
C
Caffier, H., 425, 426(2)
Cahn, M. B., 132
Cahn, R. D., 132, 198
Cailleau, R. M., 264, 266(4)
Caldwell, J. M., 451,457(1)
Calenoff, M. A., 184
Cailahan, R., 417, 421,424(28)
Camier, M., 585
Campbell, R. L., 162, 163(33)
Campenot, R. B., 306, 307
Canary, P., 377
Caplan, A. I., 130, 513
CarboneU, A. W., 252
Cardell, R. R., 564
Cardella, C. J., 265, 271(21)
Carlisle, W., 96
Carlson, P. S., 359, 360, 362(2), 365, 366(2),
367(2)
Carnright, D. V., 309
Caro, L. G., 280
593
594
AUTHOR INDEX
D
Daesch, G. E., 425
Dalton, A. J., 417
Daly, W. F., 149
D'Amato, F., 484
Danna, K. J., 412
Dannies, P. S., 528, 529(12), 532, 533(12, 42,
AUTHOR INDEX
DeIGiudice, R. A., 22, 24, 25(II),27
DeIl'Orco, R. T., 145
Dempo, K., 541
Denis, K., 351
Deshmukh, K., 561,563(7), 564(7)
Desmond, W., 160, 375(14, 18),376(14, 18),
377(18), 378,554
Dev, V. G., 174, 342
De Vitry, F., 585
Devlin, T., 267
deVries, F. A. J., 426, 434(11)
DeWitt, C. W., 149
Dhanda, V., 463
Dhar, R., 405
Dhermgrongartama, B., 351
Dieckmann, M., 411
Diez, N., 33
di Mayorca, G., 297
Dimmick, R. L., 38
Di Paolo, J. A., 343, 375(21), 377(21), 378
Dirston, W. E., 302
Dittrich, W., 240
Dixon, M. A., 296
Dolbeare, F. A., 243
Donfini, S. F., 343, 455
Donner, L., 355
Donta, S. T., 60
Dorfmann, A., 561,563(8, 9)
Dosik, H., 264
DougaII, P. K., 154
Dougherty, E. P., 140
Douglas, G. R., 343
Douglas, W. H. J., 140
Drake, W. P., 149, 151(31)
Drochmans, P., 267, 544
Druckrey, H., 586
Dubois, P., 97, 105(22)
Dudits, D., 366
Duesberg, P. H., 380
Duff, C., 309, 310(9), 343
Dulak, N. C., 107, 162, 163(31)
Dulbecco, R., 95, 120, 121, 142, 156, 298,
406, 530, 572, 586
Dupr6e, L., 153
Durie, B. G. M., 288
Dutrillaux, B., 325
E
595
596
AUTHOR INDEX
F
AUTHOR INDEX
Gey, M. K., 121, 142, 203, 211
GfeUer, M. D., 265
Giagnoni, G., 530, 531(21)
Giambrone, M. A., 271
Giard, D. J., 192, 264
Giardinello, F., 203
Giardinello, F. E., 203,204(23)
Gibbs, J. E., 288
Gifford, G. E., 198
Gilden, R. V., 405,418
Giles, R. E., 354, 359(51)
Gilman, A. G., 557, 558(15)
Gimbrone, M. A., Jr., 140
Ginsberg, H. S., 425
Giordano, R., 297
Giovanella, B., 373
Giovanella, B. C., 375(15), 376(15), 378
Girardi, A. J., 405
Giuffre, N. A., 115
Glaser, D. A., 310
Gledhill, B. L., 236
Glimelius, K., 360, 361(9), 362, 364
Glover, F. L., 203
Glover, J. S., 418
Godawska, E., 566
GiShde, W., 240
Goetz, I., 33
Goldblatt, H. L., 264
Golde, D. W., 497, 498(7)
Goldman, D., 267
Goidsby, R., 350
Goldschmidt, E., 478
Goldstein, J. L., 444
Goldstein, S., 342, 445, 446(5)
Gonzalez, E., 272
Good, N. E., 136
Good, R. A., 371
Goodhead, D. T., 301
Goodman, H. M., 534
Goodpasture, C., 326
Goodwin, R. H., 451, 460, 461
Gopalakrishnan, T. V., 507
Gordon, S., 496, 503(6)
Gorer, P., 219
Gorham, L. W., 60
Gosch, G., 365, 366, 367
Gospodarowicz, D., 78, 102, 107, 572
Grace, J., 153, 155(10)
Grace, T. D. C., 457, 458(27)
Graessman, A., 412
Graf, T., 396, 403
597
H
Haapala, D. K., 423
Habeshaw, J., 495, 496(5)
Haegeman, G., 405
Haguenau, F., 417
Halaban, R., 566, 567(18)
Hales, A., 356
Hall, D. A., 272
Hall, R. F., 30
Halle, W., 60
Ham, R., 153
598
AUTHOR INDEX
AUTHOR INDEX
Hoogasion, A., 30
Hopps, H. E., 22, 24, 25(11), 197, 405
Horine, R. K., 361
Horiwitz, S. B., 289
Horng, C. B., 184, 186(5), 207
Horwitz, A., 561,563(8)
Horwitz, A. L., 561,563(9)
Horwitz, Z. D., 528, 529(10), 533(10),
534(10)
Hosick, H. L., 147
Hoskins, J. M., 151
House, W., 14
Howard, J. C., 349, 351(26)
Howe, S. C., 349, 351(26)
Hoyt, R. F. Jr., 533(51, 54, 60), 534(51, 54,
60), 535
Hozumi, N., 350
Hsie, A. W., 288
Hsu, S. H., 454
Hsu, T. C., 173,220, 325,439
Hu, F., 564
Huang, L. C., 485
Hudson, B. G., 271
Hudson, L., 132
Huebner, R., 417
Huebner, R. J., 413
Hull, R. N., 37, 203
Humason, G. L., 287
Humphrey, R. M., 308
Humphreys, S., 583
Humphries, E. H., 389
Hungerford, D. A., 148
Hunter, W. H., 421
Hunter, W. M., 108, 418
Hutching, S. E., 94
599
J
Jacob, F., 97, 125(21, 22)
Jacobson, B., 268
Jaffe, E. S., 499
Jagannathan, V., 462
Jainchill, J. L., 413, 422(5)
Jakob, H., 97, 105(21)
Jameson, P., 296
Jamieson, C. W., 149
Jarrett, O., 297
Jenkin, H. M., 60, 454
Jensen, D. K., 528, 529(5), 533(5), 534(5),
535(5)
Jensen, F. C., 405
Jensen, M. D., 210
Jett, J., 241
Joftes, D. L., 287
Johansson, C., 174
John, R., 495
Johnsen,'J., 493
Johnson, K. M., 405
Johnson, M. J., 52, 60
Johnson, R., 352
Jones, C., 372
Jones, C. W., 364, 365, 366(30)
Jones, F. A., 124
Jones, G. E., 314
Jones, K. L., 94
Jones, V. E., 223
Jouanneau, J.-P.,482
Jovin, T. M., 246
Joworska, H., 287
Junghans, R., 377
600
AUTHOR INDEX
L
Labib, G., 363, 367(19)
Lake, S., 236
Laland, S., 493
Lambert, W. C., 261
AUTHOR INDEX
Landgraf-Leurs, I., 425, 426(2)
Landis, S. C., 583
Lang, W., 286
Langer, R., 561
Lapiere, C. M., 274
Lamer, J., 60, 79, 88(23), 99
Landau, T., 162, 163(36)
Latt, S. A., 317, 326
Lauris, V., 183, 184
Law, L. W., 369
Lawley, K. R., 271
Lazarus, K. J., 579
Leake, C. J., 463
Leblond, C. P., 287
Leder, P., 507
Lee, F. D., 302
Lee, G. F.-Y., 149
Lee, J. C., 467
Lefevre, P. A., 297
Leffert, H. L., 536, 541,542, 543(1-3), 544
Lehner, D. F., 564
Lehr, H., 31
Leibovitz, A., 59, 264, 463
Leibovitz, J., 356
Leighton, J., 264
Leighton, J. L., 553, 560(5)
Leighton, S., 554, 557(10)
Leinerman, J., 266
Leis, J., 401
Leiter, E. H., 60
LeJeune, J., 325
Lengyel, J., 454, 455(8, 13)
Lerner, A. B., 566, 567
Lerner, R. A., 218
Less, L. A., 564
Letourneau, P. C., 577
Levenson, R., 561
Levine, E. M., 22, 25(13), 27(16, 22)
Levine, D. W., 96, 185, 186(7, 8), 192,
193(9), 208
Levine, L., 97, 528, 529(2, 3), 530(2), 532(2,
3), 533(36), 534(3)
Ley, G. O., 60
Ley, K. D., 258
Leyva, A., Jr., 146, 148
Li, C. Y., 498
Li, S. Y., 454
Lieber, M. M., 416, 421
Lieberman, M. W., 297
Liebo, S. P., 31, 33(9)
Lightbody, J., 97
601
602
AUTHOR INDEX
AUTHOR INDEX
Mezger-Freed, L., 467
Michalopoulos, G., 266, 269(31), 270,
271(31), 544
Michaud, T., 567
Michayluk, M. R., 362, 363, 364(21), 365,
367
Michl, J., 499
Mickelson, M. M., 493
Miedeman, E., 145
Migeon, B. R., 315
Mijazawa, M., 219
Miller, D. A., 174, 325, 341,342, 343
Miller, E. J., 271,273(68)
Miller, O. J., 174, 325, 326, 341, 342, 343,
348, 439
Miller, O. L., Jr., 289
Miller, R. A., 367
Mills, S., 264, 278(7)
Milstein, C., 349, 351(25, 26), 357(26)
Minna, J., 346
Mironescu, S., 260, 261(27)
Misfeldt, D. S., 553,555(6), 556(6), 557(6)
Mishra, N., 297
Mitchell, B. F., 257, 258
Mitsuhashi, J., 452, 456, 462
Mix, T. W., 184
Miyake, T., 455
Miyamoto, T., 387
Mizrahi, A., 136
Mobley, W. C., 580
Moehring, J. M., 315
Moehring, T. J., 315
Moellmann, G., 566
Mohandas, T., 341, 343
Mohberg, J., 60
Moklebust, R., 33
Molander, C. W., 28, 29(29)
Montagnier, L., 160, 297
Montes De Oca, F., 166
Moore, G. E., 59, 136, 197, 204, 205, 211,
264, 564
Moran, J. S., 78, 102, 107
Moran, T., 536, 543(3)
Morgan, J. F., 59, 153, 155(12), 382
Morecki, R., 271
Morimoto, H., 148
Morley, C. G. D., 146, 147(17)
Morowitz, J., 564, 566, 567(10)
Morris, P. W., 352
Morrison, S. J., 60
Morrison, S. L., 351
603
N
Nadal-Ginard, B., 377
Nagai, K., 371
Nagington, J., 30
Nagle, S. C., Jr., 60
Nairn, R,, 434
Nakano, R., 482
Natali, P. G., 218
Nathan, C., 501
Nathans, D., 370, 405, 406(8), 412
Nathanson, M. A., 130
Nebes, S., 401
Neff, J. M., 354
Neill, W. A., 288
Nell, M. B., 354
Nelson-Rees, W. A., 29, 165, 166, 174, 323,
342, 343,439, 440
Nemeroff, K., 286
Nettleship, A., 296
Neufed, E. F., 444
604
AUTHOR INDEX
O
Obata, K., 575
O'Brien, S. J., 170
Obrink, B., 267
Oehlenschlager, V., 59
O'Farrell, M. K., 251
O'Gorman, P., 219
Ohasa, S., 96
Ohishi, M., 124, 130(9)
Ohlhaum, D. J., 499
Oie, H. K., 296
Oka, M. S., 149
Okada, Y., 355, 356(53), 359
Okayama, M., 560
O'Lague, P. H., 575, 578, 583(14)
Old, L. J., 219
Oldberg, A., 267
Oldstone, M. B. S., 218
Oliveros, R., 200
Olson, R., 170
O'Malley, K., 356
O'Neill, M. C., 521
Onoue, K., 124, 130(9)
Oroszlan, S., 418
P
Padilla, S. R., 564
Pagano, J. M., 412
Pahl, K., 365, 366
Palser, H. R., 343
Pan, J., 405
Panayi, G. S., 288
Pannett, C. A., 523, 526(37a)
Panol, G., 184
Pant, U., 462
AUTHOR INDEX
Paul, J., 4, 151
Pawelek, J., 564, 566, 567(10, 11, 18), 568
Paz, M. A., 271,272(62)
Pearlstein, E., 273
Pearse, A. G., 287
Pearson, P., 325
Pearson, P. L., 343
Peaud-Lenoel, C., 482
Peebles, P. T., 423,424(34)
Peehl, D. M., 90
Pellegrino, A., 218, 219
Pellegrino, M. A., 218, 219
Pelletier, G., 360
Penman, S., 454, 455(8, 13)
Penso, G., 4, 5(6)
Peraino, C., 150
Peralta, P. H., 405
Perdue, S. W., 288
Perez, A. G., 22, 27(15)
Perlman, D., 115, 211
Perper, R. J., 493
Perrone, M. H., 533(59), 534(59), 535
Perry, V. P., 33, 150
Pessac, B., 268
Pestka, S., 292
Petersen, D. F., 255, 343
Peterson, E. R., 577
Peterson, J. A., 347
Peterson, W. D., Jr., 165, 166, 174, 178
Peterson, W. P., 33
Petterson, U., 425, 426(6)
Pham, T. D., 326
Philip, J., 166
Phillips, D. M., 22, 28, 375
Phillips, G. B., 38
Phillips, G. W., 140
Phillips, H. J., 139, 151,218
Phillips, H. M., 267
Phillips, L. A., 423
Pictet, R. L., 265, 352
Pierce, S. L., 287
Piez, K., 525
Pierson, R. W., Jr., 571
Pike, R. M., 38
Pilkis, S. J., 544
Pina, M., 425, 426(2), 431,432, 433(14, 17),
434(4)
Pincus, T., 413
Pirt, S. J., 60
Pitelka, D. R., 126, 266, 269(32), 271(32),
272(32), 553, 555(6), 556(6), 557(6)
605
Q
Quade, K., 401
Quevedo, W. C., 565
606
AUTHOR INDEX
AUTHOR INDEX
Rugaard, J., 348
Runyan, C., 211, 212(7), 213(7), 215(7),
218(7), 219(7)
Rushton, A. R., 343
Russell, W. C., 433, 434
Russin, D., 149
Russlahti, E., 267, 273(34)
Rutter, W. J., 265, 352
Rutzky, L. P., 184
Rygaard, J., 276, 370
Rymbrandt, D., 266
S
Sabol, S. L., 530, 531(21)
Sachs, L., 162, 163(36), 296, 368
Saier, M. H., Jr., 160, 375(14, 18~, 376(14,
18), 377(18), 378,552, 554, 559, 560
Saigo, K., 455
Sakakura, T., 266
Salmon, S. E., 288
Sambrook, J., 411,425, 426(6)
Samuels, H. H., 98, 528, 529(8-10), 532(810), 533(8-10), 534(8-10)
Sanders, F. K., 151
Sanford, K. K., 5, 60, 153, 154, 156(13, 14),
188, 197, 209, 264, 369, 545
Sansone, J., 566
Sansone, M., 564, 566, 567(10)
Sarama, J., 18, 19(4), 29(4)
Sarangi, F., 315
Sargent, P. A., 314
Sarkar, N. H., 418
Sartorelli, A. C., 59
Sato, G. H., 60, 79, 88(23), 94, 96, 97,
98(14), 99, 102(6, 18), 105, 107, 109(6),
155, 160, 162(17), 264, 267(8), 278(7),
375(14, 18), 376(14, 18), 377(18), 378,
528, 529(2), 530(2), 532(2), 554, 564,
570, 571,585
Sato, K., 359
Sato, T., 271,507
Saunders, J. W., Jr., 265
Savage, R. E., 347
Sawicki, W., 151, 152(53), 288, 289(26)
Sawyer, B. D., 561,563(7), 564(7)
Scaramuzzino, D., 60, 128
Schafer, I. A., 125
Schapira, F., 544
Scharff, M. D., 350, 351, 354, 356(28)
607
608
AUTHOR INDEX
AUTHOR INDEX
Stuhblefield, E., 251,255
Studzinski, G. P., 261
Stulberg, C. S., 33, 165, 166, 174, 175, 178
Stumpf, W. E., 289
Styles, J. A., 297, 302(10, 11)
Subramanian, K. N., 405
Sugano, H., 507
Sugg, M. T., 203
Sulkin, S. E., 38
Sullivan, J. C., 125
Sullivan, P. M., 90
Summers, J., 434
Sumner, M., 162, 163(34)
Sun, A. M., 184
Sun, T., 265
Sun Lin, P., 221
Sussman, P. M., 533(56), 534(56), 535
Sutherland, Win. M., 520
Swanborg, N. K., 166
Sweet, B. H., 457
Sweet, R. G., 235
Sweet, W., 97
Swim, H. E., 30, 545, 548(2)
Sykes, J. A., 140
Syverton, J. T., 166, 178
Szegedi, M., 367
Szybalski, E., 352
Szybalski, W., 351,352
T
Tadokoro, J., 355, 356(53)
Taft, E. B., 251
Takaoka, T., 52, 60, 82, 85
Takebe, I., 363, 367(19)
Takemoto, H., 528
Talavera, A., 348
Tan, Y. H., 292
Tarikas, H., 96
Tashjian, A. H., Jr., 96, 98(14), 264, 267(8),
528,529(2, 3, 5-7, 11-13), 530(2), 532(2,
3), 533(5-7, 12-14, 33, 36, 40-45,
47-49, 51-54, 57, 60, 62, 63), 534(3,
5-7, 12-14, 33, 47--49, 51-54, 56, 57, 60,
62, 63), 535(5), 564
Taub, M., 552, 560
Taylor, J., 265
Tchao, R., 264, 554, 557(10)
Teague, J., 147, 148
Teleki, S., 60
609
610
AUTHOR INDEX
U
Uchida, T., 359
Uchimiya, H., 484
Uecker, W., 146
Ungaro, E C., 149, 151(31)
Ussing, H. H., 555, 557(14)
V
Vagi, Y., 136
Vaheri, A. J., 267, 273(43), 278
Vail, W. J., 55, 359
Valentich, J. D., 554, 557(10)
Vanaman, V., 18, 19(4), 29(4)
van der Eb, A. J., 412, 426, 434(11)
Van de Voorde, A., 405
Van Diggelen, O. P., 28, 375
Van Dilla, M. A., 235, 252
Vanha-Perttula, T., 533(553,534(553,535
van Hemert, D., 184, 186(4), 207
Van Herreweghe, J., 405
Van Heuverswyn, H., 405
Vanucchi, S., 269
van Wezel, A. L., 184, 186(1-4), 207
Varga, J. M., 566
Varma, M. G. R., 463
Varnum, D., 31
Varon, S., 576, 580, 581(53
Vas, M. R., 487
Vaughan, F. L., 265
Vaughn, J. L., 135, 451,455, 460, 461(38a)
Vaughn, V. L., 356
Velez, R., 346
Venuta, S., 368
Veri, A., 267
Vesell, E. S., 166
Vi~ek, J., 292, 293, 295(3), 296(3)
Vinograd, J., 407
Virolainen, M., 502
W
Walaas, E., 533(46), 534
Walaas, O., 533(46), 534
Walcott, R. M., 213
Waldren, C. A,, 321
Walicke, P. A., 307
Wallace, J. H., 149, 151
Wallace, R. E., 120, 121
Wallach, D. F. H., 210, 221
Wallin, A., 360, 361(9), 362, 364
Walton, M. J., 60
Waltz, H. K., 209
Waltz, J. K., 188
Wang, D. I. C., 96, 185, 186(7, 8), 192,
193(9), 208
Wang, L.-H., 380
Wang, R. J., 75, 136
Wanson, J. C., 267, 544
Wantanabe, H., 534(64), 535
Warburg, B., 368
Warburton, D., 325, 343
Wamaar, S. O., 426, 434(11)
Warren, R. J., 446
Wartiovaara, J., 267, 273(43)
Wasley, G. D., 495
Wasmuth, J. J., 317
Watabe, H., 544
Watts, R. W. E., 147
Waymouth, C., 31, 44, 60, 78(3), 141, 142,
149, 264, 500
Weber, G., 361,365, 366
Weber, M., 368
Wedum, A. G., 38
Weed-Kastelein, E., 347
Weinstein, D., 125, 146, 541
Weinstein, D. B., 542
Weinstein, R. S., 265
Weiss, M., 347, 348(16)
AUTHOR INDEX
Weiss, M. C., 347
Weiss, R., 210
Weiss, R. A., 370, 387
Weissman, S. M., 405
Wensink, P. C., 425,434(4)
Werb, Z., 496, 503(6)
Werdelin, O., 124, 130(8)
West, N. R., 579
Westermark, B., 273
Westwood, F. R., 297
Wetter, L. R., 3, 366
Weyns, J. C., 149
Whatley, S., 147, 148
Wheeler, T. B., 356
White, M., 553
White, N. K., 521,524(25)
White, P. R., 59
Whiteside, J. P., 184, 186(6), 190(6)
Whitfield, 262
Whittle, W. L., 13, 137
Widholm, J. M., 361
Wiebel, F., 248
Wiepjes, G. J., 129
Wife, R. L., 506, 510(3)
Wigglesworth, N. M., 252
Wigley, C. B., 297
Wigzell, H., 219
Wilkie, N. M., 434
Williams, G. M., 60, 128, 129, 544
Williams, J., 425,426(6)
Williams, J. F., 267, 425
Williams, J. L., 213
Williams, L. J., 375(15), 376(15), 378
Williams, R. H., 147, 148
Williamson, F., 361
Willing, M., 507
Willingham, M., 369
Willis, G. E., 365
Wilson, J. D., 445
Winget, G. D., 136
Winter, W., 136
Wise, K. S., 211,216(6), 223,228
Wiseman, L. L., 125
Wither, L. A., 363
Wittinghorn, D. G., 31, 33(9)
Wittmarm,., 512
Wold, W. S. M., 425, 426(3), 435
Wolf, C. F. W., 182, 184(4)
Wolf, K., 135, 467, 475, 477
Wolfe, S. W., 60
Wollenberger, A., 60, 147, 148
Wong, F. C., 184
611
Y
Yaffe, D., 516
Yaffe, I. R., 512
Yahara, I., 492
Yam, L. T., 498
Yamada, K. M., 267, 274
Yamada, S. S., 267, 274(38, 39)
Yamaizumi, M., 359
Yamamoto, K., 346
Yamane, I., 60
Yamasaki, E., 302
Yang, T. K., 454
Yanishevsky, R., 146
Yasumura, Y., 96, 98(14), 264, 267(8), 528,
529(2), 530(2), 532(2), 564, 570
Yavin, E., 577
Yavin, Z., 577
Yen, P. M., 533(57), 534(57), 535
Yergarian, G., 354
Yip, D. K., 151
Yokoro, K., 528
Yokota, H., 248
Yoshida, F., 146
Yoshimura, M., 560
Young, C. S. H., 425
Ysekaert, M., 405
Yu, L.-Y., 528, 529(14), 533(14, 56), 534(14,
56), 535
Yunker, C. E., 460, 461(38a)
Z
Zain, B. S., 405
Zapol, W., 181
Zaroff, L., 264, 278(7)
612
Zech, L., 174
Zee, T. W., 493
Zeigler, C. J., 356, 357(63)
Zenk, M. H., 478
Zerahn, K., 555, 557(14)
Zerthen, J., 347, 359(18)
AUTHOR INDEX
Ziegler, D. W., 203, 204(24)
Zimmerman, H. M., 586
Zimmerman, M., 267
Zimring, A., 152
Zwemer, R. K., 211, 212(7), 213(7), 215(7),
216(6), 218(7), 219(7, 14), 223, 224
SUBJECT INDEX
613
Subject Index
A
A2 APG medium, 57
Abortive infection, see Infection, abortive
Acetate, in media, 66
Acetoorcein, autoradiography prestain, 286
Acid cleaning, 5
Acridine orange, cell viability, 152
ACTH, see Adrenocorticotropic hormone
Action potential, in culture, 307
Adenine, in media, 54, 66
Adenocarcinoma
cervical, monolayer culture, 133
renal, 376
Adenosine, resistant strains, 314
Adenosine monophosphate, in media, 66
Adenosine triphosphate, in media, 66
Adenovirus
availability, 426
human, 425--435
isolation of DNA, 426
large-scale growth, 431--433
media, 427-431
monolayer culture, 426, 430
purification, 431-433
serotypes, 425
suspension culture, 426, 430
transformation, 370, 426
tumorigenicity, 425
Adenovirus DNA, 433,434
Adhesion
cellular, 369
to vessels, 137
Adhesion protein, 267
preparation, 273,274
Adipocyte, differentiation, 105
Adrenal cell Y1, see Y1 adrenal cell
line
Adrenal cortex carcinoma, 376
Adrenal tumor, plating, 573
Adrenergic neuron, 575
Adrenocortical Y1 cell, see Y1 adrenal cell
line
Adrenocorticotropic hormone
effect on Y~ cell line, 571
pituitary tumor cell, 527
release in GH cells, 534
Aedes aegypti
media, 462
suspension culture, 454
Aedes novalbopictus, media, 462
Aedes taeniorhynchus, media, 462
Aedes vexans, media, 457
Aedes w-albus, media, 462
Aerosol, biohazard, 39, 40
African green monkey kidney cell
microcarrier culture, 190
SV40, 404
Agallia constricta, media, 462
Agar assay, soft, 297, 299, 300
Agglutinability, transformed cells, 368, 369
Aging, cell, 30
Aging Cultured Cell Repository, 442
Air circulation, 40, 43
Alanine, in media, 63
Albumin, in media, 88, 500
Alpha-MEM medium, 56
Alveolar macrophage, see Macrophage, alveolar
AMBD 647/3 medium, 59
Amelanotic mutant, see Mutant, amelanotic
a-Amanitin~ resistance, 309, 314
American Type Culture Collection, 119,440,
441
Ames test, 302
Amino acids
essential, in media, 53, 54, 62
in insect media, 455
nonessential, in media, 63
Amino acid derivative, in media, 63
Aminoacyl-tRNA synthetase, mutants, 319
p-Aminobenzoic acid, in media, 64
a-Aminobutyrate, in media, 63
Amoxicillin, in media, 112, 114
AMP, see Adenosine monophosphate
Amphibia
balanced salt solution, osmolarity adjustment, 469
incubation temperature, 471
media, 469
skin culture, 473
614
SUBJECT INDEX
B
Bacitracin, in media, 565
Bacterial contamination,see Contamination,
bacterial
Balanced salt solution, see Salt solution,
balanced
Balb/c cell, effect of hormone, 101, 102
Balb 3T3 cell, 96
Banding
chromosomal, 324
techniques, problems, 337
isopycnic viral, 415
615
SUBJECT INDEX
Barley mesophyll x soybean culture, 365
Basement membrane, 265
collagen, 266
components, in media, 269
in malignancy, 266
reconstituted, rafts, 263-278
Batch growth, cost, 206
B cell, 178, 493
B1e cellline, 564
B1e melanoma M 2 R cell,96
Benzidinc, staining, 510
N6-Benzyladenine, as cytokinin, 478
Benzylpenicillin,see PenicillinG
B H K cell,trypsinization,298
B H K 21 cellline, 377
Bicarbonate
as buffer, 199, 200
in media, 70
Bilirubin, conjugation, 184
Biohazard, 36--43
aerosols, 39, 40
infectious virus, 37, 42, 43
lymphoid cells, 37, 42
Biomphalaria glabrata, media, 466
Biopsy, skin, see Skin biopsy
Biotin, in media, 54, 64
Birch and Pirt's medium, 58
14-Bis-2-(4-methyl-5-phenyloxazolyl)benzene, as scintillator, 288, 289
Biuret, 176
Blister formation, MDCK cells, 553
BME, see Eagle's basal medium
B o m b y x rnori, media, 457
Bone marrow cell
cloning, 163
human, growth factor, 163
mouse, growth factor, 163
Bovine herpes virus, 28
Bovine serum
fetal
freezing, 31
preservation, 31
growth promotion, 20, 21
mycoplasmal testing, 22-27
sterility, 19-21
tests, 20, 21
toxicity, 20
viral contamination, 28, 29
virus testing, 28, 29
Brain cell
cloning, 163
dispersion, 123
616
SUBJECT INDEX
Carcinogenicity ( c o n t ' d . )
testing, 301
survival curve, 301
transformant induction, 301
Carcinoma
adrenal cortex, 376
cervical, 376
colon, 184
embryonal
effect of hormone, 105
mouse, 97
Lewis lung, 376
lung, 376
nasopharyngeal, 376
transplantable, 276, 277
Carrot, from protoplast, 367
Carrot x petunia, 365
Cartilage, 560
Cat kidney cell, media for, 58
C-banding, s e e Centromere banding
Ce cell, effect of hormone, 103
Cell, s e e a l s o specific types
anchorage dependence, 44, 45, 48, 81,
133, 137, 152
characteristics, monitoring, 164-178
differentiated, 80, 263-278
culture, 264
media for, 58
DNA content, 148
enucleated, 359
hard-to-grow, media for, 56
intended use, 81
lifespan, 134
metaphase, s e e Metaphase cell
mitotic, s e e Mitotic cell
nontransformed, 45
media for, 56, 58
normal, 45
media for, 85, 90
protein content, 145
pulse-labeled, 244
transformed, 80, 81, 134
effect of lectins, 368
properties, 368
virus-infected
cloning, 155
microcarrier culture, 191, 192
Cell adhesion, s e e Adhesion, cellular
Cell classification, criteria, 44
Cell counting, 143
data expression, 150
direct, 143
liver cells, 543, 544
microcarrier, 188, 189
Cell culture, s e e a l s o Media; specific cell
type
characterization, 164-178
cost, 195,204-206
defined, 263
equipment, s e e Laboratory equipment
effect of buffer, 72, 73
of carbon dioxide, 72, 135
of humidity, 73, 74, 136
of light, 136
of osmolality, 73, 136, 137
of oxygen, 74, 136
of pH, 72, 135
of temperature, 55, 135, 450
facility, s e e Laboratory facility
large-scale, s e e Large-scale culture
management, 116-119
materials, sources, 196
monolayer, 46
packaging, 36
preparative operations, 195-202
shipment, 29-36
regulations, 34-36
sources, 119
sterility testing, 21
Cell cycle
analysis, 233-247
DNA content, 243-246
parameters, 241
Cell cycle phase, 248-262
quantitation, 241
Cell density, 47, 218, 219
measurement, 225
Cell dispersion, 239
fluorescence, 246, 247
mechanical, 130, 131
protocol, 246, 247
shearing, 130, 131
Cell disruption, 223
Cell dissociation
with collagenase, 128, 129
after culture, 129, 130
epithelial cell, 129
of explants, 129, 130
muscle, 513-516
by organ perfusion, 128, 129
with pronase, 129
sorting, 239
SUBJECT INDEX
Cell filter, 514
Cell fixation, 140
Cell fractionation, 222
Cell fusion, 324, 345-359
polyethylene glycol, 353, 356-359
protocol, 353, 354
Sendai virus, 353-356
suppression of function, 347
virus detection, 348, 349
Cell growth
analysis, 223
curves, 75, 76, 215
data evaluation, 150
interactions, 78, 79
kinetics, 133, 214-217, 241, 245
large-scale, 13
measurements, 142-150
capillary culture unit, 183
chemical, 144-149
electronic, 149
hematocrit, 149
microscopy, 149
visual, 142-144
on microcarrier, 187-190
parameters, 214-217
plateau region, 75, 76
population-dependent, 47
quantitation, 141-152
requirements, 51-79
synchronous, 218
Cell harvest, 221-229
Cell hybrid, s e e Hybrid cell
Cell identification, 164-178
karyotyping, 323, 324
Cell line, s e e a l s o specific types
cloning, 155
heteroploid, 323
mammalian, 371
invertebrates, 450-466
mixed, 117
nontumorigenic, 378
permanent, 45
quality control, 439
sources, 132, 439-444
stable, 439-444
suspension culture, 202-206
tumorigenic, 371, 376-378
human, 376
mouse, 377
various species, 377
Cell monitoring, s e e Monitoring
617
618
SUBJECT INDEX
SUBJECT INDEX
protocol, 154, 155
Rous sarcoma virus, 390-392
Cloth, for plating, 320
Cloudman $91 melanoma, 564
CMRL 1066 medium, 57, 88, 155
composition, 62-70
CMRL 1415 medium, 58, 87, 90, 91
composition, 62-70
CMRL 1415-ATMmedium, 58, 87
CMRL 1969 medium, 59, 87, 91
composition, 62-70
Cobalt, in media, 69
Codling moth, media, 457
Coenzyme A, in media, 64
Colcemid
mitotic cells, 255
mitotic spindle inhibitor, 327
slide preparation, 329, 331,343
synchrony, 261
Colchicine
mitotic inhibitor, 256, 327
resistance, 309, 314
Colchium mesophyU soybean culture, 365
Collagen
as culture substrate, 266
floating substrates, 266
preparation, 270-273
purification, 270, 526
as raft, 271
cell separation, 275
preparation, 270
treatment of culture dish, 525, 526
Coilagenase
cell dissociation, 128, 129
crude, 126
in media, 139
tissue culture, 125
types, 126
Colon carcinoma, 184
Colony stimulating activity, 162, 163
Complementation, genetic, 347
protoplast fusion, 367
Concanavallin A, as mitogen, 487, 492
Conditioned medium, 47, 153, 524, 581
preparation, 161-164
Conditioning, mechanism, 47
Confluence, 133
Connective tissue, transformed, 376
Contact feeding, 315
Contact inhibition, 134
Contaminant, see Contamination; specific
type
619
Contamination
bacterial, 18, 19
tests, 20
carry-over, 83
culture, 201
detection, 18-29, 116
evaluation, large-scale, 219, 220
by extraneous cells, 165
fungal, 18, 19
test, 20
microbial, 110
mycoplasmal, 18, 21-28,219, 220, 439
testing, 21-28
toxic compound, 5
viral, 28, 29
Continuous culture, 216
Continuous growth, cost, 206
Coon's medium F12, 86
Copper, in media, 52, 69
Corneal cell, rabbit, 414
Corneal tissue culture, fish, 473
Corn mesophyll x soybean culture, 365
Cotton bollworm, media, 457
Cross feeding, 315
Cross-reacting protein, 177
Cross reaction, immunological, 178
Cyrogenic storage, 31
Cryogenic supplies, 17
Cryopreservation, HTC cells, 551,552
Cryoprotective agent, 12
Crystal violet, microcarrier culture, 189
Culex quinquefasciatus, suspension culture,
454
Culex tritaeniorhynchus, suspension culture,
454
Culiseta inornata, media, 457
Culture flask, 13
Culture media, see Media
Culture plate, see also Plating
collagen-coated, 158
fibrin-coated, 158
microtest, 156
Culture system, 12-15, see also specific
system
Culture vessel, 12-15
Cyclic adenosine monophosphate
measurement, MDCK, 557-559
melanoma cell cycle, 567
Cycloheximide resistant strains, 314
Cyclostome
balanced salt solution osmolarity.adjustment, 469
620
SUBJECT INDEX
Cyclostome (cont'd.)
incubation temperature, 471
Cynthia moth, media, 457
Cysteine, in media, 53, 62, 87
Cystine, in media, 53, 62
Cytogenetics, 322
Cytokinin, in media, 478
Cytological hybridization, see Hybridization, cytological
Cytopathogenicity, mixed culture, 421-423
Cytosine arabinoside, as mutagen, 316
D
Darkroom
for autoradiography, 280, 281
safelight, 281
N - D e a c e t y l - N - m e t h y l c o l c h i c i n e , metaphase
preparations, 170
DEAE-cellulose
as microcarrier, 186
preparation, 186
DEAE-Sephadex, as microcarrier, 186, 209
Decontamination, 39
6-Demethyl-7-chlortetracyline, in media,
112, 114
Deoxyadenosine, in media, 66
Deoxycytidin, in media, 67
Deoxyguano[ine, in media, 67
Deoxyribonuclease, tissue culture, 125, 126
Deoxyribonucleic acid
cell content, 148
in cell cycle, 243-246
determination, protocol, 146-149
extraction, 406-409
Hirt procedure, 406--409
injection into cells, 412
isolation, from human adenovirus, 426
linear duplexes, 411
relaxed circles, 411
separation, 411
supercoiled, 411
thymidine incorporation, 251,252
viral, see Viral DNA; specific types
Deoxyribose, in media, 66
Dermatan sulfate, 268
Detergent, in media, 70
Diarrhea virus, 28
Dibutyl cAMP, effect on growth, 245
2,4-Dichlorophenoxyacetic acid, as auxin,
478
Digestion buffer, liver cells, 542, 543
karyotype, 342
media, 464
suspension culture, 454, 455
Drosophila virilis, media, 464
Drug-resistance mutant, see Mutant, drugresistance
Dual-rotary circumfusion, 14
SUBJECT INDEX
621
622
SUBJECT INDEX
SUBJECT INDEX
G
623
624
SUBJECT INDEX
storage, 5 5 1 , 5 5 2
suspension culture, 550, 551
HEPES buffer, in media, 70, 72, 73
H e r p e s v i r u s s i m i a e , 37, 43
Heterokaryon, 345
Heteroploid cell line, see Cell line,
heteroploid
Higuchi's medium, 57
composition, 62-70
Hink medium, 461
Hirt procedure, extraction, 406-409
Histidine, in media, 53, 62
Histidinol, resistant strains, 314
Histone, 49
33258 H/Sechst dye, as sensitizer, 317
Hormone, see also specific substance
effect on embryonal carcinoma cells, 105
on GHa cells, 98-100
on growth, 94-109
on HeLa cells, 100
on HeLa-S cells, 107-109
on melanoma ceils, 102
on mouse fibroblasts, 101, 102
Swiss 3T3, 105, 106
on neuroblastoma, 97, 98
on rabbit intimal cells, 106, 107
on rat follicular cells, 100, 101
on rat glial C6 cells, 103-105
on testicular cells, 103
in serum, 78, 83
Hormone growth factor, 78
Horse serum, 95, 213
in media, 523, 530
HTC cell, see Hepatoma tissue culture cell
Human
adenovirus, see Adenovirus, human
cell
media for, 56, 57
nontransformed, media for, 56
tumorigenic, 376
epidermal keratinocyte, media for, 90
embryonic lung fibroblast, microcarrier
culture, 190
fetal lung cell, protein content, 145
fibroblast
DNA content, 148
as feeder cell, 358
foreskin, microcarrier culture, 190
interferon production, 295
isolation, 444--450
media for, 56, 58, 59
SUBJECT INDEX
monolayer culture, 132
preservation, 31
propagation, 444
karyotype, 340-342
leukemia cell, protein content, 145
leukocyte, cloning, 163
lymphoblast, slide preparation, 328-330
lymphocyte, 486-494
culture conditions, 491,492
DNA content, 148
marrow cell, growth factor, 163
RSTC-2 cell, interferon, 293
Human Genetic Mutant Cell Repository,
441,442
Humidity, effect on culture, 73, 74, 136
Hyaluronate, 268
aggregation, 268
Hyaluronidase, tissue culture, 125
Hybrid
mouse human, 347, 357
somatic, 359, 360
Hybrid cell, 324, 345
Hybrid fusion, plants, 364-366
Hybridization, cytological, 326
Hybrid plant, cell fusion, 364-366
Hybrid selection, procedures, 351-353
Hydrocortisone, in media, 100, 101, 109
Hydroxymethyl cellulose, 580
20ct-Hydroxyprogesterone, synthesis, 571
Hydroxyproline, in media, 53, 63
5-Hydroxytetracycline, in media, 112, 114
Hydroxyurea
resistant strains, 314
synchrony, 259-261
Hygienic practice, 39
Hypercholesterolemia, familial, 444
Hypothalamic peptide, regulatory effects,
527
Hypoxanthine, in media, 54, 67
Hypoxanthine-guaninephosphoribosyl
transferase, hybrid selection, 352
I
IMEM-ZO medium, 58, 87
composition, 62-70
Immunological cell monitoring, 174-178
Immunological cross reaction, 178
IMR-91 cell, protein content, 145
Incubation, mutant isolation, 315, 316
Incubation temperature
poikilotherm vertebrate cells, 470-472
625
626
SUBJECT INDEX
Isozyme
cell monitoring, 166-170
extract preparation, 167
staining, 168-170
J
Japanese quail
feeder cell, 392
Rous sarcoma virus, 395
Jensen sarcoma cell, protein content, 145
K
Kanamycin, in media, 112, 114, 116
Karyotyping, 170-174, 220, 322-344
characteristics, 342
CHO cells, 340-342
Drosophila melanogaster, 342
GH cells, 532
human, 340-342
modifications, 333, 334
mouse, 341,342
mutants, 309
problems in, 330
rat, 342
standardization, 339
Y1 adrenal cell line, 571
Katsuta medium (DM, 57, 62-70, 88
KB cell line
infection, 425; 431
monolayer culture, 430
suspension culture, 430
Keratinocyte, human epidermal, media for,
9O
A4-3-Keto-C~z-steroid, synthesis, 571
Kidney, dispersion, 123
Kidney cell, 377
cat, media for, 58
monkey
dissociation, 127
microcarrier culture, 190
suspension culture, 203
SV40, 404
Kidney epithelial cell, 552-560
Kinetics, of growth, 133,214-217, 241, 245
Kinetin, as cytokinin, 478
K12 cell, synchronization, 252-256
L
Laboratory equipment, suppliers, 15-17,
196
Laboratory facility
cleaning, 3-7
design, 4-7
sterilization, 3-7
storage, 3, 7-9
Laboratory operation, 3
Laboratory safety, 36--43
Lactate dehydrogenase
assay, 169, 170
cell monitoring, 168-170
isoenzyme, 169, 170
Lactate dehydrogenase stain, 169
Lamelopodia, transformed cells, 368
Laminar flow system, 10, 40, 154
Large external transformation substance,
see Fibronectin
Large-scale culture, 211-221
adenoviruses, 431-433
chondrocytes, 560-564
cost, 205
equipment, 221-229
growth methods, 194-210
microcarrier, 193
Rous sarcoma virus, 395-403
Large-scale growth, see Large-scale culture
L cell
large-scale growth, 211
mouse, 77, 84
cloning, 163
interferon production, 295
medium, 84, 85
Leafhopper, media, 462
Leaf mesophyll cell, 360
Lecithin, in media, 68, 500
Lectin
cell transformation, 368
resistance, 309, 314
Leibovitz's medium L-15, 56, 87, 91
composition, 62-70, 463
preparation, 463, 464
Lesch-Nyhan syndrome fibroblast, 444
Leucine, in media, 53, 62
Leukemia antigen
murine thymus, 219
Leukemia cell
granuloeytic, DNA content, 148
SUBJECT INDEX
human, protein content, 145
media for, 56
Leukemia virus, see Murine leukemia virus
Leukocyte
cloning, 155
human, 163
isolation, 487-489
poikilotherm, 472
Leukopheresis, 493, 494
Lewis lung carcinoma, 376
L-5 cell, protein content, 145
Light, effect on growth, 136
Light scattering, 238
Lincomycin, in media, 112, 114
Linoleic acid, in media, 54, 68, 90
Lipid, in media, 68
a-Lipoic acid, in media, 54, 64
Lipopolysaccharide, as mitogen, 351
Liposome, 54
Liquid nitrogen
in freezing, 30
storage, 33
Liver
dispersion, 123
rat, perfusion, 128, 129
Liver cell
buffer solutions, 540
cloning, 163
counting, 543, 544
differentiation, 541
digestion buffer, 542, 543
establishment of cell lines, 536-544
isolation, 543
media for, 59, 537, 542,543
plating, 538,539, 543
rat
adult, 539
fetal, 538
media for, 90
Lowry protein assay, 144
interference, 146
L. pomonella, media, 457
LS cell, 89
L251A cell line, 219
Lung, dispersion, 123
Lung carcinoma, 376
Lung cell
human embryo, protein content, 145
fibroblast
fetal, 50
627
628
SUBJECT INDEX
SUBJECT INDEX
629
trace elements, 69
MCDB 501, 59, 62-70, 87
vitamins, 64, 65
MD 705/1 (Kitos), 57, 88
conditioned, 47, 153,524, 581
minimum essential (Eagle), s e e Eagle's
minimum essential medium
preparation, 161-164
costs, 204-206
Mitsuhashi and Maramorosch, s e e Mitdefined, 88-90
suhashi and Maramorosch medium
development, 91-93
mixture, 97; 99, 102-104, 106
four factors, 98
DM, 59
DM 120, 57, 88
Mohberg and Johnson's, 57
DM 145, 57, 62-70, 88
MPNL65/C, 59
DM 160, 57, 88
Nagle and Brown's, 58, 89
Dulbecco's, s e e Dulbecco's modified
NCTC-109, 57
Eagle's medium
NCTC 135 (Evans), 57, 62-70, 88
EM-I, 105
N16, 56
Fischer's, s e e Fischer's medium
199, 56, 62-70, 87, 90, 91, 129, 155
5A (McCoy), 56, 62-70, 86, 91
Parsa, 90
F10(Ham), s e e Ham's medium F10
pH, 199, 200
F12(Ham), s e e Ham's medium F12
preparation, 7-9, 94-109, 198, 199
F12 + hormones, 57
products, 225-228
F12K, 59, 87, 91, 155
quality control, 453, 454
F12M, 155
RPMI 1640, 56, 62-70, 89, 91, 155,
GHAT, 352
213
Goodwin IPL-52, s e e Goodwin IPL-52
Schneider, s e e Schneider medium
selective hybrid, 351, 352
medium
Grace, s e e Grace medium
serumless, 57, 62-70, 94-109
half-selective hybrid, 352
serum supplements, 122
Hansen S-301, s e e Hansen medium S-301
7C's, 58, 89
HAT, 351-353
Sinclair's, 58
Higuchi's, s e e Higuchi's medium
SM-20 (Halle), 59, 90
Hink, s e e Hink medium
SM-201, 59
history, 84, 85
$77, 546
sterility testing, 21
hormone-supplemented, 94-109
IMEM-ZO, 58, 62-70, 87
storage, 7-9
insect cell, 455-466
supplements, 122
L-15 (Leibovitz), 56, 62-70, 87, 91,463
suppliers, 15, 16
testing, 91
listing, 56-59
liver cells, 537
toxicity, 117
MAB 87/3 (Gorham and Waymouth), 57,
toxicity testing, 91,453,454
virus in, 225-228
62-70, 88
V-605, 56
macrophage, 499-502
maintenance, 87
White's, 56
MAP 954/1 [Waymouth (Donta)], 57, 89
Williams E, 59, 90
Williams G, 59, 62-70, 90
MB 752/1 (Waymouth), 57, 62-70, 88, 89,
90
WO~, 57
McCoy's 5A, s e e McCoy's medium 5A
Wollenberger, 90
MCDB 104, 59, 87, 91, 155
Yamane's, s e e Yamane's medium
Yunker e t a l . , s e e Yunker, Vaughn and
MCDB 105, 59, 62-70, 76, 87
MCDB 202, 59, 62-70, 76, 87, 91
Cory medium
MCDB 301, 58, 62-70, 89
Medium A2 + APG, 57
Medium alpha-MEM, 56
MCDB 401, 59, 62-70, 87, 90, 91
MCDB 411, 57, 62-70, 89
Medium AMBD 647/3, 59
630
SUBJECT INDEX
SUBJECT INDEX
as
mutagen, 312
Microcarrier culture, 184-194
cell counting, 188
cell yield, 210
culture initiation, 187
growth cycle, 187-190
harvesting, 190, 191
interferon formation, 192
large-scale, 193
preparation, 185-187
primary cells, 207, 208
seeding, 187, 188
source, 185-187
synthesis, 185-187
toxicity, 184, 185
Microculture, neurons, 582, 583
Microscope, 11, 12
suppliers, 17
Microscopy
chromosome study, 336-339
monolayer cultures, 139, 140
Microsome preparation, 224
Microtest plate, 156
Microvillus, transformed cells, 368
Minimum essential medium, see Eagle's
minimum essential medium
Mink lung cell, 414
Mithramycin, DNA determination, 147
Mitochondria, isolation, 222
Mitogen
dosage, 492
effect on lymphocyte, 486
Mitotic cell
increase, 255
selection, 192, 193
Mitotic detachment, synchronization, 252256
Mitotic index, 233, 327
Mitotic spindle inhibitor, 327
Mitosis, peak activity, 254-256
Mitsuhashi and Maramorosch medium, 454,
462
preparation, 462
Mixed culture
cytopathogenicity, 421-423
neurons and nonneuronal ceils, 581
Mixed media, see Media, mixture
Mohberg and Johnson's medium, 57
Moloney murine leukemia virus, microcarrier culture, 192
Molybdenum, in media, 52, 69
N-Methyl-N'-nitrooN-nitrosoguanidine,
631
Monitoring
antigen-antibodyreaction, 174-178
with antiserum, 176-178
cell characteristics, 164-178
by chromsomal examination, 170-174
by isozymes, 166-170
Monkey kidney cell
dissociation, 127
suspension culture, 203
Monocyte, 492, 494, 495
Monolayer culture
H T C cells,545
human adenovirus, 426
media for, 88, 89
microscopic, evaluation, 139
morphology, 139, 140
poikilotherm vertebrate cells, 474-476
properties, 133-135
protocol, 138, 139
substrate for attachment, 137, 138
techniques, 132-140
Moore's medium RPMI 1640, 56, 62-70, 89,
91
Morris hepatoma, 544
Moscona's saline, 138, 139
Mosquito cell
media, 457, 462
suspension culture, 454
Moth cell line, 450
media, 456, 462
suspension culture, 455
Mouse
cell
nontramsformed, media for, 56
tumorigenic, 376
embryo
carcinoma, 97
cooling rate, 31
fibroblast
media for, 58, 59, 90
monolayer culture, 132
media for, 87
crythrolcukemia cell, 346, 506-511
cloning, 509
growth conditions, 507-509
hemoglobin induction, 509, 510
media, 508
serum
requirements, 508
testing, 508, 509
fibroblast
effect of hormone, 101, 102
632
SUBJECT INDEX
Mouse ( c o n t ' d . )
Swiss 3T3, effect of hormone, 105, 106
karyotype, 341, 342
L cell
cloning, 163
interferon production, 295
lymphoma cell $49
cell cycle phase, 242
DNA content, 244
fluorescence spectrum, 238
lymphosarcoma MB(T-86157), suspension
culture, 203
marrow cell, growth factor, 163
melanoma cell line PG19, tumorigenicity
testing, 374
neuroblastoma cell
media for, 57
C1300, 89
nude, s e e Nude mouse
pancreatic cell, media for, 59
pituitary cell, characteristics, 531
plasmacytoma, cell fusion, 349
sarcoma virus, 413
testicular cell TM4, 96, 97
thymusless, 370
Mouse human hybrid, 347, 357
MPNL 65/C medium, 59
Mucopolysaccharide, 267, 268
Mucopolysaccharide storage disease, 444
Multicell culture, 264
Multiplication stimulating activity, 79, 96,
107, 162, 163
Murashige and Skoog inorganic salt, 480
Murine cl 1 cell, microcarrier culture, 190
Murine leukemia virus, 225, 226
assay, 412-424
enzymic, 416---421
immunological, 416-421
virological, 421-424
cells for propagation, 414
host range, 413
purification, 412-424
SL
- test, 423, 424
source, 414
XC test, 421-423
Murine lymphoblastoid cell, 218
EL4, 223
Murine lymphoma virus, transformation,
369
Murine sarcoma virus, transformation, 369
Murine thymus leukemia antigen, 219
SUBJECT INDEX
colony, 24
DNA stain, 25
immunofluorescence, 24, 25
infection, testing, 375
isolation from medium, 227
large-scale production, 227
microscopy, 25
pseudo-colony, 24
scanning electron microscopy, 28
in serum, 23
testing, 22-27
uracil uptake, 26, 27
uridine phosphorylase, assay, 25, 26
M y c o p l a s m a hyorhinis, immunofluorescence, 24, 25
Mycoplasmal contamination, see Contamination, mycoplasmal
Mycoplasma ribosomal RNA, 27
Mycostatin, in media, 198
Myeloma, 376
hybrid, 349
Myoblast, skeletal, 377
cell source, 512
culture, 511-527
differential release, 517
differentiation from fibroblasts, 517
establishment of culture, 512
inoculum size, 518-520
media, 520-524
composition, 518-520
conditioned, 524, 525
secondary suspension, 516-518
synchrony, 511,518-520
Myo-inositol, in media, 478
N
NAD, see Nicotinamide adenine dinucleotide
NADP, in media, 64
Nagle and Brown's medium, 58, 89
1-Naphthaleneacetic acid, as auxin, 478
Nasopharyngeal carcinoma, 376
National Institute on Aging Cell Repository,
442
National Science Foundation Cell Culture
Centers, 443
NCTC 109 medium, 57, 89
NCTC 135 medium, 57, 88
composition, 62-70
Neomycin, in media, 112, 114, 116
633
mesophyll, 365
Nicotiana t a b a c u m L., callus culture, 479
Nicotinamide, in media, 64
Nicotinamide adenine dinucleotide, in
media, 64
Nicotinic acid, in media, 64
634
SUBJECT INDEX
0
Oat roots x maize, 365
Oleic acid, in media, 54, 68
199 medium, 56, 87, 90, 91, 129, 155
composition, 62-70
Organ culture, defined, 263
Organelle
fractionation, 222
preparation, 221-229
subceliular, 223
Ornithine, in media, 63
Osmolarity
adjustments for lower vertebrates, 469
SUBJECT INDEX
Peritoneal macrophage, see Macrophage,
peritoneal
Petri dish, 13
Petunia
from protoplast, 367
protoplast fusion, 367
Petunia A t r o p a , 365
Petunia x carrot, 365
P e t u n i a hybrida mesophyll P. parodii
mesophyU, 365
pH
effect on culture, 72, 135
of media, 199, 200
for poikilotherm vertebrate cells, 470
Phagocyte, see also Macrophage
uptake by macrophage, 498, 499
Phase-contrast microscopy, monolayer cultures, 139
Phase fraction analysis, 241
Phenazine methosulfate, isoenzyme staining, 168, 169
Phenol red
in media, 70
pH indicator, 121, 122
Phenotype
expression, time, 313
stability, 308
Phenylalanine, in media, 53, 62
Phenylmethylsulfonyl fluoride, protease inhibitor, 273
Phosphate, in media, 68
Photography
choice of film, 338
chromosome study, 336-339
film development, 338
printing, 339
Phytohemagglutinin, as mitogen, 487, 492
Phywe flow cytometer, 236
Pigmentation, genetic marker, 565
Pineal gland
avaian, cell dissociation, 130
rat, cell dissociation, 129, 130
Pipetting technique, 38, 39
Pituitary cell
ACTH-producing, 531
corticotropin-producing, 531
/~-endorphin-producing, 531
enkephalin-producing, 531
growth hormone formation, 528
hormone-producing, 528
mouse, characteristics, 531
635
636
SUBJECT INDEX
Poly-D-lysine, 49
Polymer, in media, 70
Polymyxin B, in media, 113, 115
Polyoma virus, transformation, 370
Poly IC, s e e Polyriboinosinic: polyribocytidylic acid
Polyriboinosinic:polyribocytidylic acid, interferon producing, 294, 295
Polysaccharide, in agar
acidic, 160
sulfated, 160
Population analysis, cell cycle, 243-246
Poststalning, autoradiography, 286, 287
Potassium, in media, 68
Potato tuber moth, media, 462
PPO, s e e 2,5-Diphenyloxazole
Prague strain, Rous sarcoma virus, 380, 394
Prestaining, autoradiography, 286
Priming, interferon production, 296
Pristane, priming, 350
Progesterone, in media, 102, 107
Prolactin
pituitary tumor cell, 527
production, 183
Proline, in media, 53, 63
Pronase, tissue culture, 125, 126, 129
Propidium iodide
fluorescence spectrum, 238
fluorescent dye, 236-238, 242
preparation, 247
Protamine, 49
in media, 70
Protease, neutralization, 77
Protein
adhesion, s e e Adhesion protein
cell content, 145
cross-reacting, 177
determination, protocol, 144-146
in media, 70, 88
synthesis, mutant, selection, 317-319
Proteoglycan, 268
differentiation, 268, 269
preparation, 274
Protoplast
formation, 361
from pea, 361
plating, 483
separation, 361
stabilization, 360, 361
tobacco leaf, 362
from V i c i a , 361
wall regeneration, 361
Q
Q-banding, s e e Quinacrine banding
Quality control
cell line, 439
media, 453,454
Quasidiploid cell line, 323
Quinacrine banding, 324, 325
Quinacrine stain, 325
R
Rabbit
antimouse antiserum, 276, 277
aortic intimal cell, 96, 97
corneal cell line, 414
embryo cell, media for, 58
intimal cell, effect of hormone, 106, 107
Rabies virus, microcarrier culture, 192
Radioimmunoassay
competitive, 420, 421
viral structural proteins, 417--421
Rapeseed, from protoplasts, 367
Rapeseed mesophyll x soybean culture, 365
Rat
embryo cell, media for, 58
epithelial cell, media for, 57
follicular cell, effect of hormone, 100, 101
glioma Ce, 97
SUBJECT INDEX
effect of hormone, 103-105
media for, 57
hepatoma, 544
karyotype, 342
liver cell
media for, 90
perfusion, 128, 129
plating, 538, 539
neuroblastoma, 96
ovarian cell, 96
pancreatic cell, media for, 59
pineal gland, cell dissociation, 129, 130
pituitary cell, 529
serum, sympathetic neurons, 580
submaxillary gimmel factor, 101
submaxillary gland, extract, 104
R-banding, see Reverse banding
Redox potential, of medium, 74
Refrigeration, 9
Renal adenocarcinoma, 376
Replicate plating
cloth, 320
procedure, 320, 321
Reptile
balanced salt solution, osmolarity adjustment, 469
incubation temperature, 471,472
media, 469
Resistance, multiple loci, 315
Resistant strain, drug concentration, 314,
315
Resting cell, human lymphocyte, 486--494
Retinoic acid, in media, 103
Reverse banding, 325
R h i p i c e p h a l u s appendiculatus, media, 463
Riboflavin, in media, 54, 65
Riboflavin 5-phosphate, in media, 65
Ribonuclease, tissue culture, 125
Ribonucleic acid, isolation, 568,569
Ribose, in media, 66
Ribostamycin, in media, 116
Ricin, resistance, 309
Rinaldini's solution, 452
Ringer's balanced salt solution, 120, 121
RNA, see Ribonucleic acid
RNase, see Ribonuclease
RNA tumor virus
microcarrier culture, 191
transformation, 369
Rodent neoplasm, 584-590
Roller bottle, 13, 14, 397
637
638
SUBJECT INDEX
Selenium
deficiency, 74
in media, 52, 69, 86, 89, 90
neuroblastoma, 98
Self-absorption, in autoradiography, 289,
290
Sendai virus
cell fusion, 353-356
in nucleus transfer, 359
storage, 354
Sephadex G50M, as microcarrier, 186
Serine, in media, 63
Sertoli cell, 103
Serum, see also specific type
bovine, see Bovine serum
certification, 197
cloning, 163
commercial production, 197
deprivation, synchrony, 259-261
dialyzed, 82, s e e a l s o Serum protein
effect on growth, 77-79, 94-109
on macrophage, 501
minimal requirements, 91-93
neuronal cell, 587
pretreatment, 95
as trypsin inhibitor, 95, 128
weaning from, 91-93
Serum albumin, in media, 88
Serumless medium, 57
composition, 62-70
Serum protein, 82
minimalization, 49-51
7C's medium, 58, 89
Shearing, 223
effect on cell, 224
Silicon, in media, 69
Silkworm, media, 457
Simian virus 40, see SV40
Sinclair's medium, 58
Sindbis virus, microcarrier culture, 192
Skeletal myoblast, see Myoblast, skeletal
Skeletal muscle, dispersion, 123
Skin biopsy
explants, 446-448
method, 445, 446
Skin epithelium, differentiation, 265
Skin explant, 446--448
Slide preparation, chromosomes, 326-334
S+L
- test, 423, 424
SM-20 medium, 59, 90
SM-201 medium, 59
SUBJECT INDEX
Storage facility, 3
Streptomycin, in media, 113, 115
Stromal cell, 266
primary culture, 278
Subcell culture, defined, 263
Subculture, 95
GH cells, 532
invertebrate cell line, 451,452
poikilotherm vertebrate cells, 476, 477
YI cell line, 572, 573
Sulfate, in media, 68
Superinduction, interferon, 295
Supernatant reverse transcriptase assay, 416
Suspension culture, 14, 46--49, 81
cell lines, 202-206
chondrocyte, 563
HTC cells, 550, 551
human adenovirus, 426
invertebrate cell, 454, 455
media for, 56, 58, 89
operations, 205
plant cell, 481,482
primary cell, 206-210
scaling-up, 211-221
Swiss 3T3 cell, 96, 97
SV40
biological activity, 412
cleavage maps, 412
culture, 405
MEM, 405, 406
preparation, 404--412
transformation, 370
SV40 DNA, 405
assay, 411,412
preparation, 406--410
purified virions, 409, 410
quantitation, 411,412
radiolabeling, 410, 411
Sympathetic ganglion-like PC12 cell, 585
Sympathetic neuron
cell preparation, 575,576
cholinergic activity, 575
co-culture, 575,581
conditioned medium, 581,582
differentiation, 574
long-term culture, 574-584
media, 578-580
microculture, 582, 583
phenotypic characteristics, 583,584
use of Methocel, 580
Synchronous growth, 218
639
Synchrony, 248-262
calcium deprivation, 261,262
criteria, 249
degree of, 249-257,259
double-thymidine block, 261
evaluation, 249-252
Ficoll gradients, 257, 258
gradient centrifugation, 256-258
hydroxyurea, 259-261
isoleucine deprivation, 258, 259
mitotic detachment, 252-256
myoblast, 511,518-520
physical methods, 256, 257
serum deprivation, 259-261
Syrian hamster karyotype, 342
Syringe adapter, 9
T
Taurine, in media, 63
T ceil, 178, 493
Teleost
balanced salt solution osmolarity adjustment, 469
incubation temperature, 471
media, 469
Temperature, effect on cuRure, 55, 135, 450
Teratoma, ovarian, 376
Testicular cell
effect of hormone, 103
mouse TM4, 96, 97
Testosterone, in media, 102, 103
Tetracarcinoma, 376
Tetracycline, in media, 113, 115, 116, 565
Tetraphenylboron, as chelator, 126
Thiamin, in media, 54, 65
Thiamin monophosphate, in media, 65
Thiamin pyrophosphate, in media, 65
Thioctic acid, s e e a-Lipoic acid
Thioglycolate broth, 20
6-Thioguanine
hybrid selection, 352
resistant strains, 314
3T3 cell, 376
monolayer, 133
mouse, DNA content, 148
Threonine, in media, 53, 62
Thymidine
incorporation, 251, 252
in media, 67
640
SUBJECT INDEX
Thymidine (cont'd.)
synchrony, 261
tritiated, as mutagen, 316, 317
Thymine, in media, 67
Thymus, lymphocyte preparation, 124
Thymus cell, canine, 414
Thyrotropin-releasing hormone, in media,
99
Thyroxine, in media, 63
Tick cell line, media, 463
Tin, in media, 69
Tissue culture, 119-131
defined, 263
Tissue dispersion, 119-131
mechanical, 130, 131
shearing, 130, 131
solutions, 124
Tissue disruption, 119-131
Tissue dissociation, see Tissue dispersion
Tissue preparation, 122, 123
mincing, 124
Tobacco, from protoplast, 367
Tobacco cell, callus culture, 479
Tobacco hornworm, media, 457
Tobacco leaf, protoplast, 302
Tobacco mesophyll x chicken red cell, 365
Tobacco mesophyll x HeLa culture, 365
Tobacco mesophyll soybean culture, 365
a-Tocopherol phosphate, see Vitamin E
Togavirus, interferon producing, 293
Toluidine blue, chondrocyte staining, 563
Torenia baillonii T. fournieri petals, 365
Toxicity, media, 117, 453, 454
Trace element, in media, 52, 69, 95
Tracheal cell, embryonic bovine, 29
Tranfection-infectivityassay, 434, 435
Transferrin, in media, 95, 99-107, 109, 500
Transformation, see also Cell, transformed;
Cell transformation
neoplastic, 296-302
soft agar assay, 297,299, 300
Triatoma infestans, media, 463
Trichoplusia ni cell, 450
media, 457,460
storage, 453
suspension culture, 455
Triiodothyronine, in media, 99, 101
Trout cell, incubation temperature, 471
Trypan blue, cell viability, 151, 152
Trypsin
contamination, 125
in media, 139
neutralization, 77
Trypsin-EDTA solution, metaphase preparation, 171
Trypsin inhibitor, 95, 128
Trypsinization, 50
adenovirus, 433,434
BHK cells, 298
in cloning, 156
enzymes for, 124-128
myoblasts, 517
poikilotherm vertebrate cell, 475, 476
Trypsinizing flask, 127
Trypticase soy broth, 20
Tryptophan, in media, 53, 62
Tumor, transplantable, 276, 277
Tumorigenic cell line, see Cell line,
tumorigenic
Tumorigenicity
human adenovirus, 425
testing
nude mouse, 370-379
protocol, 373-379
threshold, 374
Tumor-specific cell surface antigen, see
Antigen, tumor-specifc-cell surface
antigen
Tween 20, in media, 70
Tween 80, in media, 70
Tylosin, in media, 113, 115, 116
Tyrode's balanced salt solution, 120, 121,
452
Tyrosinase
assay, 568
melanocytes, 564
toxicity, 567
Tyrosine, in media, 53, 62
U
Ultrafiltration, virus, 228
Ultraviolet light, as mutagen, 312
Uracil
in media, 67
uptake, mycoplasmal, 26, 27
Uridine, in media, 67
Uridine phosphorylase, assay, 25, 26
Uridine triphosphate, in media, 67
Urine, cloning, 163
Ussing chamber, 555-557
641
SUBJECT INDEX
V
Valine, in media, 53, 62
Vanadium, in media, 52, 69
Vascular endothelial cell, monolayer, 133
Vasopressin, cAMP production, 559
Velveteen cloth, for plating, 320
Ventilation, laminar air, 10
Vertebrate cell, 466-477
lower, 467
osmolarity adjustments, 469
poikilotherm, 466--477
media, 470
monolayer culture, 474-476
pH, 470
primary culture, 474-476
subculture, 476, 477
temperature requirements, 470--472
Vesicular stomatitis virus, microcarrier culture, 192
Viability, s e e Cell viability
Vicia, protoplast, 361
Vicia culture pea mesophyll, 365
Vicia mesophyll soybean culture, 365
Vinblastine, mitotic inhibitor, 256
Viokase
in media, 532, 572, 573
neuronal cell culture, 587
Viomycin, in media, 113, 115
Viral antigenic determinant, 418
Viral contamination, s e e Contamination,
viral
Viral DNA, assay, 434, 435
Virus, see a l s o specific type
acquisition, 226, 227
banding, isopycnic, 415
detection, cell fusion, 348, 349
DNA, s e e Viral DNA
harvesting, 399, 400
infectious, biohazard, 37, 42, 43
isolation, 222, 227
in media, 225-228
microcarrier culture, 191, 192
nontransforming, large-scale growth,
403
preparation, 222
production
effect of serum, 226, 227
mechanization, 400, 401
ultrafiltration, 228
purification, 401-403,414-416
testing, 28, 29
xenotropic, 413,414
Vitamin
fat-soluble, 54, 65
in media, 54, 64, 65
Vitamin A, in media, 65
Vitamin B, in media, 54, 65
Vitamin D, in media, 65
Vitamin E, in media, 65
Vitamin K, in media, 65
V-605 medium, 56
W
Walker carcinosarcoma 256 cell, 86
Water
distillation, 195
preparation, 94
cost of, 195
quality, 8
system, suppliers, 17
treatment, 8
Waymouth's medium MB 752/1, 57, 88, 90
composition, 62-70
White's medium, 56
Williams' medium E, 59, 90
Williams' medium G, 59, 90
composition, 62-70
Wisconsin 38 cell, callus culture, 479
WISH cell, protein content, 145
WI-38 cell
cloning, 155
DNA content
protein content, 145
Wollenberger medium, 9
WO5 medium, 57
Working culture, defined, 118
Wrist shaker, 203
X
XC test, 421--423
Xenotropic virus, s e e Virus, xenotropic
Y
Yamane's medium, 57
composition, 62-70
Y chromosome, 174
642
YI adrenal cell line, 570-574
effect of ACTH, 571
initiation of stock, 572
karyotype, 571
media, 571
monolayer growth, 572
stability, 570
steroidogenesis, 571
SUBJECT INDEX
storage, 573
subculture, 572, 573
Yunker, Vaughn and Cory medium, 461