cytotoxicity...hepatoma-derived cells such as hep-g2 or heparg (see section 22.2.8). coculture may...

17
CHAPTER 21 Cytotoxicity 21.1 VIABILITY, TOXICITY, AND SURVIVAL Once a cell is explanted from its normal in vivo environment, the question of viability, particularly in the course of experimental manipulations, becomes fundamental. Previous chapters have dealt with the status of the cultured material relative to the tissue of origin and how to quantify changes in growth and phenotypic expression. However, none of these data is acceptable unless the great majority of the cells are shown to be viable. Furthermore many experiments carried out in vitro are for the sole purpose of determining the potential cytotoxicity of compounds being studied, either because the compounds are being used as pharmaceuticals or cosmetics and must be shown to be nontoxic or because they are designed as anticancer agents and cytotoxicity may be crucial to their action. New drugs, cosmetics, food additives, and so on, go through extensive cytotoxicity testing before they are released for use by the public. This testing usually involves a large number of animal experiments, although in Europe these experiments will be subject to new legislation [Cox & Chrisochoidis, 2003], introduced in 2009 for topical application and in 2013 for systemic application. There is much pressure, both humane and economic, to perform at least part of cytotoxicity testing in vitro. The introduction of specialized cell lines and interactive organotypic cultures, and the continued use of long-established cultures, may make this a reasonable proposition. Toxicity is a complex event in vivo, where there may be direct cellular damage, as with a cytotoxic anticancer drug, Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications, Sixth Edition, by R. Ian Freshney Copyright © 2010 John Wiley & Sons, Inc. physiological effects, such as membrane transport in the kid- ney or neurotoxicity in the brain, inflammatory effects, both at the site of application and at other sites, and other systemic effects. Currently it is difficult to monitor systemic and physiological effects in vitro, so most assays determine effects at the cellular level. Definitions of cytotoxicity vary [Kroemer et al., 2009], depending on the nature of the study and whether cells are killed or phenotypically altered. In addition cells may die by necrosis (see Fig. 12.1), apoptosis (see Plate 17c , d), self-digestion (autophagy); may cease proliferation (cytostasis); and/or may become terminally differentiated (e.g., cornification) [Galluzi et al., 2009]. Whereas demonstrating efficacy in an anticancer agent assay may require a cytocidal effect (cell killing), demonstrating the lack of toxicity of other pharmaceuticals may require a more subtle analysis of specific targets such as an alteration in gene transcription, cell signaling, or cell–cell interaction including those effects that may give rise to an inflammatory or allergic response. Most assays oversimplify the events that they measure and are employed because they are cheap, easily quantified, and reproducible. However, it has become increasingly apparent that they are inadequate for modern drug development, which requires greater emphasis on specific molecular targets and precise metabolic regulation. Gross tests of cytotoxicity are still required, but there is a growing need to supplement them with more subtle tests of metabolic pathway regulation and signaling. Perhaps the most obvious of these tests is the induction of an inflammatory or allergic response, which need not imply cytotoxicity of the allergen and is still one of the hardest results to demonstrate in vitro. 365

Upload: others

Post on 08-Nov-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21

Cytotoxicity

21.1 VIABILITY, TOXICITY, AND SURVIVAL

Once a cell is explanted from its normal in vivo environment,the question of viability, particularly in the course ofexperimental manipulations, becomes fundamental. Previouschapters have dealt with the status of the cultured materialrelative to the tissue of origin and how to quantify changes ingrowth and phenotypic expression. However, none of thesedata is acceptable unless the great majority of the cells areshown to be viable. Furthermore many experiments carriedout in vitro are for the sole purpose of determining thepotential cytotoxicity of compounds being studied, eitherbecause the compounds are being used as pharmaceuticalsor cosmetics and must be shown to be nontoxic or becausethey are designed as anticancer agents and cytotoxicity maybe crucial to their action.

New drugs, cosmetics, food additives, and so on, gothrough extensive cytotoxicity testing before they arereleased for use by the public. This testing usually involvesa large number of animal experiments, although in Europethese experiments will be subject to new legislation [Cox& Chrisochoidis, 2003], introduced in 2009 for topicalapplication and in 2013 for systemic application. There ismuch pressure, both humane and economic, to perform atleast part of cytotoxicity testing in vitro. The introduction ofspecialized cell lines and interactive organotypic cultures, andthe continued use of long-established cultures, may makethis a reasonable proposition.

Toxicity is a complex event in vivo, where there may bedirect cellular damage, as with a cytotoxic anticancer drug,

Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications, Sixth Edition, by R. Ian FreshneyCopyright © 2010 John Wiley & Sons, Inc.

physiological effects, such as membrane transport in the kid-ney or neurotoxicity in the brain, inflammatory effects, bothat the site of application and at other sites, and other systemiceffects. Currently it is difficult to monitor systemic andphysiological effects in vitro, so most assays determine effectsat the cellular level. Definitions of cytotoxicity vary [Kroemeret al., 2009], depending on the nature of the study andwhether cells are killed or phenotypically altered. In additioncells may die by necrosis (see Fig. 12.1), apoptosis (see Plate17c, d), self-digestion (autophagy); may cease proliferation(cytostasis); and/or may become terminally differentiated (e.g.,cornification) [Galluzi et al., 2009]. Whereas demonstratingefficacy in an anticancer agent assay may require a cytocidaleffect (cell killing), demonstrating the lack of toxicity ofother pharmaceuticals may require a more subtle analysis ofspecific targets such as an alteration in gene transcription, cellsignaling, or cell–cell interaction including those effects thatmay give rise to an inflammatory or allergic response.

Most assays oversimplify the events that they measure andare employed because they are cheap, easily quantified, andreproducible. However, it has become increasingly apparentthat they are inadequate for modern drug development,which requires greater emphasis on specific molecular targetsand precise metabolic regulation. Gross tests of cytotoxicityare still required, but there is a growing need to supplementthem with more subtle tests of metabolic pathway regulationand signaling. Perhaps the most obvious of these tests is theinduction of an inflammatory or allergic response, whichneed not imply cytotoxicity of the allergen and is still one ofthe hardest results to demonstrate in vitro.

365

Page 2: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

366 CULTURE OF ANIMAL CELLS

The traditional approach to cytotoxicity has been to con-centrate on cell growth or survival. Cell growth is generallytaken to be the regenerative potential of cells, as measured byclonal growth (see Protocol 20.10), net change in populationsize (e.g., in a growth curve; see Protocols 20.7, 21.8), or achange in cell mass (total protein or DNA) or metabolic activ-ity (e.g., DNA, RNA, or protein synthesis; MTT reduction).Other aspects will be considered later (see Section 21.6).

21.2 IN VITRO LIMITATIONS

It is important that any in vitro measurement can beinterpreted in terms of the in vivo response of the same orsimilar cells, or at least that the differences that exist betweenin vitro and in vivo measurements are clearly understood.

21.2.1 PharmacokineticsThe measurement of toxicity in vitro is generally a cellularevent. For example, it would be very difficult to recreatethe complex pharmacokinetics of drug exposure in vitro, andbetween in vitro and in vivo experiments there usually aresignificant differences in exposure time to and concentrationof the drug, rate of change of the concentration, drugmetabolism (activation and detoxification), tissue penetration,clearance, and excretion. Although it may be possible tosimulate these parameters—for example, using multicellulartumor spheroids for drug penetration or timed perfusionto simulate concentration and time (C × T ) effects—moststudies concentrate on a direct cellular response, therebygaining simplicity and reproducibility.

21.2.2 MetabolismMany nontoxic substances become toxic after beingmetabolized by the liver; in addition many substances thatare toxic in vitro may be detoxified by liver enzymes. Forin vitro testing to be accepted as an alternative to animaltesting, it must be demonstrated that potential toxins reachthe cells in vitro in the same form as they would in vivo.This proof may require additional processing by purified livermicrosomal enzyme preparations [McGregor et al., 1988],coculture with activated hepatocytes [Guillouzo & Guguen-Guillouzo, 2008] (see also Appendix II: Hepatocytes), orhepatoma-derived cells such as Hep-G2 or HepaRG (seeSection 22.2.8). Coculture may use transfilter 3-D culture or2-D cellular microarrays [Khetani & Bhatia, 2008]. Geneticmodification of the target cells with the introduction of genesfor metabolizing enzymes under the control of a regulatablepromoter [Mace et al., 1994] has also been used.

21.2.3 Tissue and Systemic ResponsesThe nature of the response must also be considered carefully.A toxic response in vitro may be measured by changes in cellsurvival (see Protocol 21.3) or metabolism (see Section 21.3.4),whereas the major problem in vivo may be a tissue response(e.g., an inflammatory reaction, fibrosis, kidney failure) or a

systemic response (e.g., pyrexia, vascular dilatation). For invitro testing to be more effective, models of these responsesmust be constructed, perhaps utilizing organotypic culturesreassembled from several different cell types and maintainedin the appropriate hormonal milieu.

It should not be assumed that complex tissue andeven systemic reactions cannot be simulated in vitro.Assays for inflammatory responses, teratogenic disorders, andneurological dysfunctions may be feasible in vitro, given theright tissue-engineered models and a proper understanding ofcell–cell interaction and the interplay of endocrine hormoneswith local paracrine and autocrine factors.

21.3 NATURE OF THE ASSAY

The choice of assay will depend on the agent under study, thenature of the anticipated response, and the particular targetcell. In vitro assays can be divided into five major classes:

(1) Viability. An immediate or short-term response, such asincreased and uncontrolled membrane permeability or aperturbation of a particular metabolic pathway correlatedwith cell proliferation or survival.

(2) Survival. The long-term retention of self-renewal capa-city (5–10 generations or more).

(3) Metabolic. Assays, usually microtitration based, of inter-mediate duration that can either measure a metabolicresponse (e.g., dehydrogenase activity; DNA, RNA, orprotein synthesis) at the time of, or shortly after, exposure.Making the measurement two or three populationdoublings after exposure is more likely to reflect cellgrowth potential and may correlate with survival.

(4) Genotoxicity and Transformation. Survival in an alteredstate (e.g., one or more genetic mutations withresultant alterations in growth control or malignanttransformation).

(5) Irritancy. A response analogous to inflammation, allergy,or irritation in vivo; as yet difficult to model in vitro, butmay be possible to assay by monitoring cytokine releasein organotypic cultures.

21.3.1 ViabilityViability assays are used primarily to measure the proportionof viable cells after a potentially traumatic procedure, such asprimary disaggregation, cell separation, or cryopreservation,rather than to look at a long-term cytotoxic response.

Most viability tests rely on a breakdown in membraneintegrity measured by the uptake of a dye to which the cellis normally impermeable, such as Trypan Blue, Erythrosin,Naphthalene Black (see Plate 17a), or propidium iodide (seeFig. 20.7), or the release of a dye normally taken up andretained by viable cells (e.g., diacetyl fluorescein or NeutralRed), or the release of lactate dehydrogenase by leaky cells

Page 3: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 367

[Kendig & Tarloff 2006]. This effect presents immediatelyand does not always predict ultimate survival as dye exclusiontends to overestimate viability—for example, 90% of cellsthawed from liquid nitrogen may exclude Trypan Blue, butonly 60% prove to be capable of attachment 24 h later.

Note that routine assessment of viability at subculture canbe uninformative regarding trypsinized adherent cells as mostof the nonviable cells will be lost in the discarded mediumand prewash before trypsinization. An accurate assessment ofthe viability status at subculture requires that all the cells berecovered from the medium and prewash and combined withthe trypsinate. However, the viability of reseeded cells will beaccurately determined without this recovery.

Protocol 21.1 can be adapted for training (see Sections 28.3,Exercises 11, 28.4, 18B, 19; see also Section 28.4).

PROTOCOL 21.1. ESTIMATION OF VIABILITYBY DYE EXCLUSION

PrincipleViable cells are impermeable to Naphthalene Black,Trypan Blue [Kaltenbach et al., 1958], propidiumiodide [Darzynkiewicz & Gong, 1994], and a numberof other dyes.

OutlineMix a cell suspension with stain, and examine it bylow-power microscopy.

Materials

Sterile or aseptically prepared:

� Cells for testing (e.g., flask for trypsinization, frozenvial to thaw, or primary disaggregate)

� Growth medium appropriate to cell type� Trypsin, crude, 0.25%� D-PBSANonsterile:

� Hemocytometer� Viability stain (e.g., 0.4% Trypan Blue or 1%

Naphthalene Black in D-PBSA or HBSS)� Pasteur pipettes� Microscope� Tally counter

Procedure

1. Prepare a cell suspension at a high concentration(∼1 × 106 cells/mL) by trypsinization or bycentrifugation and resuspension.

2. Take a clean hemocytometer slide and fix thecoverslip in place (see Protocol 20.1; Fig. 20.1).

3. Mix one drop of cell suspension with one drop(Trypan Blue) or four drops (Naphthalene Black)of stain.

4. Load the counting chamber of the hemocytometer(see Protocol 20.1).

5. Leave the slide for 1 to 2 min before starting tocount (do not leave any longer, or viable cells willdeteriorate and take up the stain).

6. Place the slide on the microscope, and use a 10×objective to look at the counting grid (see Fig. 20.1;Plate 17a).

7. Count the total number of cells and the number ofstained cells.

8. Wash the hemocytometer, and return it to its box.

Analysis. Calculate the percentage of unstained cells.This figure is the percentage viability by this criterion.If the respective volumes of cell suspension and stain aremeasured accurately at step 3, then this method of viabilitydetermination can be incorporated into Protocol 20.1.

PROTOCOL 21.2. ESTIMATION OF VIABILITYBY DYE UPTAKE

PrincipleViable cells take up diacetyl fluorescein andhydrolyze it to fluorescein, to which the cellmembrane of live cells is impermeable [Rotman& Papermaster, 1966]. Live cells fluoresce green;dead cells do not. Nonviable cells may be stainedwith propidium iodide and subsequently fluorescered [Darzynkiewicz & Gong, 1994]. Viability isexpressed as the percentage of cells fluorescing green.This method may be applied to CCD analysis or flowcytometry (see Section 20.7.2; Fig. 20.7).

OutlineStain a cell suspension in a mixture of propidiumiodide and diacetyl fluorescein, and examine thecells by fluorescence microscopy or flow cytometry.

Materials

Sterile or aseptically prepared:

� Single-cell suspension� Fluorescein diacetate, 10 μg/mL, in HBSS without

phenol red� Propidium iodide, 500 μg/mL, in HBSS without

phenol red

Nonsterile:

� Fluorescence microscope� Filters:

Fluorescein: excitation 450/590 nm, emission LP515 nm

Page 4: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

368 CULTURE OF ANIMAL CELLS

Propidium iodide: excitation 488 nm, emission615 nm

Procedure

1. Prepare the cell suspension as for dye exclusion(see Protocol 21.1), but in medium without phenolred.

2. Add the fluorescent dye mixture at a proportion of1 : 10 to give a final concentration of 1 μg/mL ofdiacetyl fluorescein and 50 μg/mL of propidiumiodide.

3. Incubate the cells at 37◦C for 10 min.4. Place a drop of the cells on a microscope slide, add

a coverslip, and examine the cells by fluorescencemicroscopy with excitation at 488 or 536 nm anddetection at 562–588 or 617 nm respectively forpropidium iodide and excitation at 494 nm andemission at 521 for fluorescein.

Analysis. Cells that fluoresce green are viable, whereasthose that fluoresce red are nonviable. Viability may beexpressed as the percentage of the total number of cellsthat fluoresce green. The stained cell suspension can also beanalyzed by flow cytometry (see Section 20.7.2; Fig. 20.7).

Neutral red uptake. Living cells take up neutral red,40 μg/mL in culture medium, and sequester it in thelysosomes. However, neutral red is not retained by nonviablecells. Uptake of neutral red is quantified by fixing the cells informaldehyde and solubilizing the stain in acetic ethanol, andmeasuring absorbance on an ELISA plate reader at 570 nm[Borenfreund et al., 1990; Babich & Borenfreund, 1990].Neutral red tends to precipitate, so the medium with stain isusually incubated overnight and centrifuged before use. Thisassay does not measure the total number of cells, but it doesshow a reduction in the absorbance related to loss of viablecells and is readily automated.

21.3.2 SurvivalAlthough short-term tests are convenient and usually quickand easy to perform, they reveal only cells that are dead (i.e.,permeable) at the time of the assay. Frequently, however, cellsthat have been subjected to toxic influences (e.g., irradiation,environmental toxins, and antineoplastic drugs) show an ef-fect several hours, or even days, later. The nature of the testsrequired to measure viability in these cases is necessarily differ-ent because, by the time the measurement is made, the deadcells may have disappeared or some resistant cells may haverecovered. Therefore long-term tests are used to demonstratesurvival rather than short-term toxicity. Survival implies theretention of regenerative capacity and is usually measuredby plating efficiency (see Protocol 20.10). Plating efficiencymeasures survival by demonstrating proliferative capacity forseveral cell generations, provided that the cells plate with a

high-enough efficiency that the colonies can be consideredrepresentative of the entire cell population. Although notideal, a plating efficiency of over 10% is usually acceptable.

PROTOCOL 21.3. CLONOGENIC ASSAY FORATTACHED CELLS

OutlineTreat the cells for 24 h with the experimental agent ata range of concentrations. Trypsinize the cells, seedthem at a low cell density, and incubate them for 1 to3 weeks. Stain the cells (see Plate 6a, e), and countthe number of colonies (see Fig. 21.1).

Materials

Sterile:

� Growth medium� D-PBSA� Trypsin, crude, 0.25%� Compound to be tested at 10× the maximum

concentration to be used, dissolved in serum-freemedium (check the pH and the osmolality of thetest solution, and adjust if necessary)

� Flasks, 25 cm2

� Petri dishes, 6 or 9 cm, labeled on the side of thebase

Nonsterile:

� D-PBSA� Methanol� Crystal Violet, 1%� Hemocytometer or electronic cell counter

Procedure

1. Prepare a series of cultures in 25-cm2 flasks, threefor each of six agent concentrations, and threecontrols. Seed the cells at 5 × 104 cells/mL in4.5 mL of growth medium, and incubate themfor 48 h, by which time the cultures will haveprogressed into the log phase (see Section 20.9.2).

2. Prepare a serial dilution of the compound to give2 mL at 10× of each of the final concentrationsrequired:(a) If you are testing a compound for the first time,

use 3- to 5-fold serial dilutions over a range of3 to 5 logs.

(b) If you can predict the approximate toxic con-centration, then select a narrower, arithmeticrange over one or two decades.

3. Add 0.5 mL of 10× concentrate to each of threeflasks for each concentration, starting with controlmedium (no compound added but solvent presentin amount used for toxin) and progressing fromlowest to highest concentration.

Page 5: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 369

4. Return the flasks to the incubator.5. If the compound is slow acting or partially rever-

sible, repeat step 3 twice; that is, expose thecultures to the agent for 3 days, replacingthe medium and compound daily by changingthe medium. With fast-acting compounds, 1-hexposure may be sufficient.

6. Remove the medium from each group of threeflasks in turn (working from control then lowestconcentration of compound to highest), trypsinizethe cells, and count the cells in the controls.

7. Dilute and seed the cells into Petri dishesat the required density for clonal growth (seeProtocols 13.1, 20.10), diluting all of the culturesby the same amount as the control. Work fromthe flasks exposed to the highest concentration oftoxin down to the lowest and then the control.

8. Incubate the cultures until colonies form (usually1–3 weeks).

9. Fix the cultures in absolute methanol, and stainthem for 10 min in 1% Crystal Violet (seeProtocol 15.3).

10. Wash the dishes in tap water, drain, and stand inan inverted position to dry.

11. Count the colonies with >50 cells (>5 doublings).

Analysis of survival curve(1) Calculate the plating efficiency at each drug concentra-

tion.(2) Calculate the relative plating efficiency, which is the

plating efficiency at each concentration as a fraction ofthe control—the surviving fraction.

For high grade or acute cytotoxin, treat flasks for 1– 72 h depending on nature of cytotoxin and cell cycle duration

For low grade cytotoxin or presumed chronic exposure treat cloning cultures for 1–3 weeks, depending on the nature of the cytotoxin.

Trypsinize the monolayer, resuspend cells, and count.

Dilute serially to between 10 and 200 cells/ml (based on cell count of controls)

Incubate for from 1–3 weeks (depending on growth rate)

Increasing concentration of cytotoxin

Fig. 21.1. Clonogenic Assay for Adherent Cells. Cells are trypsinized, counted, and diluted as formonolayer dilution cloning (see Protocol 13.1). The test substance can be added before trypsinizationor after seeding for cloning (see Section 21.3.2). The colonies are fixed and stained when they reach areasonable size for counting by eye but before they overlap.

Page 6: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

370 CULTURE OF ANIMAL CELLS

0 1 2 3 4 5 6 7 8 9 100.001

0.01

0.1

1

Sur

vivi

ng fr

actio

n

Concentration of cytotoxin

IC50 IC90

Fig. 21.2. Survival Curve. Semilog plot of the surviving fraction ofcells (ratio of colonies forming from test cells to colonies formingfrom control cells) against the concentration of cytotoxin. Typicallythe curve has a ‘‘knee,’’ and the IC90 lies in the linear range of thecurve. The IC50, falling on the knee, is a less stable value.

(3) Plot the surviving fraction on a log scale against theconcentration on a linear or log scale, depending on theconcentration range used (see Fig. 21.2).

(4) Determine the IC50 or IC90, which is the concentrationof compound promoting 50% or 90% inhibition of colonyformation, respectively. As this is a semilog plot, the IC90

is more appropriate, as it is more likely to fall on thelinear part of the curve, whereas the IC50 tends to fall onthe knee of the curve, giving a less stable value.

(5) Analyze the curve for differences in sensitivity:(a) Slope of the curve and length of the knee. A shal-

lower slope and/or longer knee means reducedsensitivity; a steeper slope and/or shorter knee meansincreased sensitivity. Both the length of knee andthe slope influence the IC50 and the IC90, althougha more significant difference can be observed in theIC90 (see Fig. 21.3).

(b) Resistant fraction. The fraction of resistant cells isindicated by a flattening of the lower end of thecurve.

(c) Total resistance is indicated by the lack of any gradienton the curve.

(d) Area under the curve: Complex survival curves may becompared by calculating the area under the curve, butthis is done for expediency and is not mathematicallyvalid.

Variable parameters in survival assayConcentration of agent. A wide range of concentrations

in log increments (e.g., 1 μM–1 mM, and control) should beused for the first attempt and a narrower range (log or linear),

0.001

0.01

0.1

1

Sur

vivi

ng fr

actio

n

0 1 2 3 4 5 6 7 8 9 10

Concentration of cytotoxin

Completely resistant

More sensitive

Sensitive

Less sensitive

1% Resistant fraction

Very sensitive

Fig. 21.3. Interpretation of Survival Curves. Semilog plot of cellsurvival against the concentration of cytotoxin. The slope increaseswith increasing sensitivity and decreases with reduced sensitivityuntil it becomes totally flat for complete resistance. Partial resistanceas a resistant fraction is shown by the curve flattening out at thelower end.

based on the results from the first range, for subsequentattempts.

Invariant agent concentrations. Some conditions thatare tested cannot easily be varied—such as the quality ofmedium, water, or an insoluble plastic. In these cases theserum concentrations can be varied. As serum may have amasking effect on low-level toxicity, an effect may only beseen in limiting serum (see Effect of Medium Constituents,below, and Fig. 21.4b).

Duration of exposure to agent. Some agents act rapidly,whereas others act more slowly. Exposure to ionizingradiation, for example, need last only a matter of minutesto achieve the required dose, whereas testing some cycle-dependent antimetabolic drugs may take several days toachieve a measurable effect. Duration of exposure (T ) anddrug concentration (C) are related, although C × T is notalways a constant. Prolonging exposure, usually by replacingthe drug daily, can increase sensitivity beyond that predictedby C × T because of cell cycle effects and cumulative damage.

Time of exposure to agent. When the agent is solubleand expected to be toxic, the procedure in Protocol 21.3should be followed, but when the quality of the agent isunknown, stimulation is expected, or only a minor effect isexpected (e.g., 20% inhibition rather than several-fold), theagent may be incorporated during clonal growth rather thanat preincubation. Confirmation of anticipated toxicity—suchas for a cytotoxic drug—requires a conservative assay witha minimal drug exposure, as compared to that in vivo,applied during culture before trypsinization for cloning.

Page 7: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 371

0.001

0.01

0.1

1

Sur

vivi

ng fr

actio

n

5-Fluorouracil, M

0

With feeder layer

Without feeder

layer

(a) (b)

1 10 100 10000.1 0.01 0.1 1 10 100 10001

10

100

Formaldehyde (mM)

Rel

ativ

e pl

atin

g ef

ficie

ncy

High serum medium

HSM w/o serum

Low serum medium

Buccal epithelial growth medium (BEGM) w/o pituitaryextract BEGM w/o pituitary extract and cysteine

Fig. 21.4. Effect of Culture Conditions on Survival. (a) Cell density. Human glioma cells were platedout in the presence (dashed line) and absence (solid line) of a feeder layer after treatment with variousconcentrations of 5-fluorouracil. A 10% resistant fraction is apparent at 1 × 10−4 M drug only in thepresence of a feeder layer. In the absence of the feeder layer, the small number of colonies makingup the resistant fraction were unable to survive alone. (b) Medium constituents. High serum medium(HSM; solid line and squares) compared with HSM without serum (dashed line and circles) showsabout a 5-fold increase in the IC50 with serum; removing cysteine decreases the IC5010-fold (datafrom [Nilsson et al., 1998]).

Confirmation of the lack of toxicity—such as for tap water ora nontoxic pharmaceutical—requires a more stringent assay,with prolonged exposure added at seeding for clonal growthand maintained during clonal growth. The toxin may needto be replaced weekly; daily replacement would alone impaircloning efficiency.

Cell density during exposure. The density of the cellsduring exposure to an agent can alter the response of the cellsand the agent; for example, HeLa cells are less sensitive tothe alkylating agent mustine at high cell densities [Freshneyet al., 1975].

Cell density during cloning. The number of coloniesmay fall at high concentrations of a toxic agent, but it ispossible to compensate for this effect by seeding more cellsso that approximately the same number of colonies form ateach concentration. This procedure removes the risk of a lowclonal density influencing survival and improves statisticalreliability, but it is prone to the error that cells from higherdrug concentrations are plated at a higher cell concentration,a factor that may also influence survival. It is preferableto plate cells on a preformed feeder layer, whose density(5 × 103 cells/cm2) greatly exceeds that of the cloning cells.This step ensures that the cell density is uniform regardlessof clonal survival, which contributes little to the total celldensity. Note that cloning on a feeder layer can sometimes

reveal a resistant fraction of cells that is not apparent withoutthe feeder layer (Fig. 21.4a).

Effect of medium constituents. The composition ofthe culture medium will also affect the way that cellsrespond to a toxin, partly because different media will havedifferent effects on proliferation but also because individualconstituents may affect the stability, binding, and metabolismof the toxin. Serum and cysteine, for example, reduce thetoxicity of formaldehyde on buccal epithelium 5- and 10-foldrespectively (Fig. 21.4b). Serum proteins may bind toxins,quite apart from their effect on cell growth and cysteineand other sulphydryls bind and detoxify reactive chemicalsintracellularly and in the medium [Nilsson et al., 1998]. Ascysteine auto-oxidizes in the medium, the age of the mediumwill add another variable [Grafstrom, personal communication].

Colony size. Some agents are cytostatic (i.e., they inhibitcell proliferation) but not cytotoxic, and during continuousexposure they may reduce the size of colonies withoutreducing the number. In this case the size of the coloniesshould be determined by densitometry [McKeehan et al.,1977], automatic colony counting, or visually counting thenumber of cells per colony. For colony counting, the thresholdnumber of cells per colony (e.g., 50 as in Protocol 21.9) ispurely arbitrary, and it is assumed that most of the coloniesare greatly in excess of this number. Colonies should be

Page 8: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

372 CULTURE OF ANIMAL CELLS

grown until they are quite large (>1 × 103 cells), when thegrowth of larger colonies tends to slow down; smaller, but stillviable, colonies tend to catch up with these larger colonies.For colony sizing, stain the cultures earlier, before the growthrate of larger colonies has slowed down, and score all of thecolonies.

Solvents. Some agents to be tested have low solubilitiesin aqueous media, and it may be necessary to use an organicsolvent to dissolve them. Ethanol, propylene glycol, anddimethyl sulfoxide have been used for this purpose, butmay themselves be toxic to cells. Hence the minimumconcentration of solvent should be used to obtain a solution.The agent may be made up at a high concentration in, forexample, 100% ethanol, then diluted gradually with BSSand finally diluted into medium. The final concentration ofsolvent should be <0.5% and a solvent control must be included(i.e., a control with the same final concentration of solventbut without the agent being tested).

Take care when using organic solvents with plastics orrubber. It is better to use glass with undiluted solvents and touse plastic only when the solvent concentration is <10%.

Although calculating the plating efficiency is one ofthe best methods for testing cell survival rates, it shouldbe remembered that plating efficiency only applies to theclonogenic component of the cell population, which maynot be representative of the whole cell population. Thequestion does not arise if controls plate with 100% efficiency;in practice, however, control plating efficiencies of 20% orless are more likely and the response is being measured in asubset of the total cell population.

21.3.3 Assays Based on Cell ProliferationCell counts after a few days in culture can also be usedto determine the effect of various compounds on cellproliferation, but at least in the early stages of testing, acomplete growth curve is required (see Protocols 20.7–21.9)because the interpretation of cell counts at a single point intime can be ambiguous (see Fig. 20.10, day 7). Growth curveanalyses, using cell counting, are feasible only with relativelysmall numbers of samples, as they become cumbersome in alarge screen, although automating growth curves in multiwellplates using image analysis (Incucyte; Chip-Man; see Section20.9.3; Figs. 20.11, 20.12) can make this feasible.

In cases where there are many samples, a single pointin time—such as the number of cells three to five daysafter exposure—can be used. The time should be selectedas within the log phase, and preferably mid-log phase, ofcontrol cells. Any significant effect should be backed up witha complete growth curve over the whole growth cycle or byan alternative assay, such as a survival curve by clonogenicassay (see Protocol 21.3) or MTT assay (see Protocol 21.4).

21.3.4 Metabolic Cytotoxicity AssaysPlating efficiency tests are labor intensive and time-consumingto set up and analyze, particularly when a large number of

samples is involved (although this can be automated; seeSection 26.4), and the duration of each experiment may beanywhere from two to four weeks. Furthermore some celllines have poor plating efficiencies, particularly freshly isolatednormal cells, so a number of alternatives have been devisedfor assaying cells at higher densities (e.g., in microtitrationplates; see Section 21.3.5). None of these tests measuressurvival directly, however. Instead, the net increase in thenumber of cells (i.e., the growth yield; see Section 21.3.3), theincrease in the total amount of protein or DNA, or continuedmetabolic activity, such as the reduction of a tetrazolium salt(MTT or XTT) to formazan or the synthesis of proteinor DNA, is determined. Survival in these cases is definedas the retention of metabolic or proliferative ability by thecell population as a whole some time after removal of thetoxic influence. However, such assays cannot discriminatebetween a reduction in metabolic or proliferative activity percell and a reduced number of cells, and therefore any novel orexceptional observation should be confirmed by clonogenicsurvival assay.

21.3.5 Microtitration AssaysThe introduction of multiwell plates revolutionized theapproach to replicate sampling in tissue culture. Theseplates are economical to use, lend themselves to automatedhandling, and can be of good optical quality. The mostpopular are 96-well microtitration plates or microplates (seePlate 17b), each well having 28 to 32 mm2 of growth area,0.1 or 0.2 mL medium, and up to 1 × 105 cells. Microtitrationoffers a method by which large numbers of samples may behandled simultaneously, but with relatively few cells persample. With this method, the whole population is exposedto the agent, and viability is determined subsequently, usuallyby measuring a metabolic parameter such as the ATP orNADH/NADPH concentration. Assay kits are available (seeAppendix II: Cytotoxicity Assays).

The end point of a microtitration assay is usually anestimate of the number of viable cells, if the assay is doneafter the removal of the toxin. Although this result can beachieved directly by cell counts or by indirect methods,such as isotope incorporation, cell viability as measured byMTT reduction [Mosmann, 1983] is widely used as theendpoint [Cole, 1986; Alley et al., 1988]. MTT is a yellowwater-soluble tetrazolium dye that is reduced by live, butnot dead, cells to a purple formazan product that is insolublein aqueous solutions. However, a number of factors caninfluence the reduction of MTT [Vistica et al., 1991]. Theassay described in Protocol 21.4, provided by Jane Plumb ofthe Cancer Research UK Centre for Oncology and AppliedPharmacology, University of Glasgow, Scotland, UK, hasbeen shown to give the same results as a standard clonogenicassay [Plumb et al., 1989] (see also Section 21.3.6). It illustratesthe use of microtitration in the assay of anticancer drugs,but would be applicable, with minor modifications, to anycytotoxicity assay.

Page 9: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 373

PROTOCOL 21.4. MTT-BASED CYTOTOXICITYASSAY

PrincipleCells in the exponential phase of growth are exposedto a cytotoxic drug. The duration of exposure isusually determined as the time required for maximaldamage to occur, but is also influenced by thestability of the drug. After removal of the drug,the cells are allowed to proliferate for two tothree population-doubling times (PDTs) in order todistinguish between cells that remain viable and arecapable of proliferation and those that remain viablebut cannot proliferate. The number of surviving cellsis then determined indirectly by MTT dye reduction.The amount of MTT-formazan produced can bedetermined spectrophotometrically once the MTT-formazan has been dissolved in a suitable solvent.

OutlineIncubate monolayer cultures in microtitration platesin a range of drug concentrations (Fig. 21.5). Removethe drug, and feed the plates daily for two to threePDTs; then feed the plates again, and add MTT toeach well. Incubate the plates in the dark for 4 h,and then remove the medium and MTT. Dissolve thewater-insoluble MTT-formazan crystals in DMSO,add a buffer to adjust the final pH, and record theabsorbance in a plate reader.

Materials

Sterile:

� Growth medium� Trypsin (0.25% + EDTA, 1 mM, in PBSA)� MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenylte-

trazolium bromide (Sigma), 50 mg/mL, filtersterilized

� Sorensen’s glycine buffer (0.1 M glycine, 0.1 MNaCl adjusted to pH 10.5 with 1 M NaOH)

� Microtitration plates (Iwaki)� Pipettor tips, preferably in an autoclavable tip box� Petri dishes (non–TC-treated), 5 cm and 9 cm or

reservoir (Corning)� Universal containers or tubes, 30 mL and 100 mL

Nonsterile:

� Plastic box (clear polystyrene, to hold plates)� Multichannel pipettor� Dimethyl sulfoxide (DMSO)� DMSO dispenser (optional); such as Labsystems

Microplate Dispenser (Cat No 5840 127, fromThermo Fisher; see also Fig. 4.7)

� ELISA plate reader (Molecular Devices, withSOFTmax PRO; see also Fig. 4.7; Appendix II:Plate Readers)

� Plate carrier for centrifuge (for cells growing insuspension; see Appendix II: Microtitration PlateCentrifugation)

Procedure

Plating out cells

1. Trypsinize a subconfluent monolayer culture, andcollect the cells in growth medium containingserum.

2. Centrifuge the suspension (5 min at 200 g) to pelletthe cells. Resuspend the cells in growth medium,and count them.

3. Dilute the cells to 2.5 to 50 × 103 cells/mL,depending on the growth rate of the cell line,and allowing 20 mL of cell suspension permicrotitration plate.

4. Transfer the cell suspension to a 9-cm Petri dish,and, with a multichannel pipette, add 200 μL ofthe suspension into each well of the central 10columns of a flat-bottomed 96-well plate (80 wellsper plate), starting with column 2 and ending withcolumn 11, placing 0.5 to 10 × 103 cells into eachwell.

5. Add 200 μL of growth medium to the eight wellsin columns 1 and 12. Column 1 will be used toblank the plate reader; column 12 helps maintainthe humidity for column 11 and minimizes the‘‘edge effect.’’

6. Put the plates in a plastic lunch box, and incubatein a humidified atmosphere at 37◦C for 1 to 3 days,such that the cells are in the exponential phase ofgrowth at the time that drug is added.

7. For nonadherent cells, prepare a suspension infresh growth medium. Dilute the cells to 5 to100 × 103 cells/mL, and plate out only 100 μLof the suspension into round-bottomed 96-wellplates. Add drug immediately to these plates.

Drug addition

8. Prepare a serial fivefold dilution of the cytotoxicdrug in growth medium to give eight concentra-tions. This set of concentrations should be chosensuch that the highest concentration kills most of thecells and the lowest kills none of the cells. Oncethe toxicity of a drug is known, a smaller rangeof concentrations can be used. Normally threeplates are used for each drug to give triplicatedeterminations within one experiment.

Page 10: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

374 CULTURE OF ANIMAL CELLS

9. For adherent cells:(a) Remove the medium from the wells in columns

2 to 11. This can be achieved with ahypodermic needle attached to a suctionline.

(b) Feed the cells in the eight wells in columns 2and 11 with 200 μL of fresh growth medium;these cells are the controls.

(c) Transfer the drug solutions to 5-cm Petri dishes,and add 200 μL to each group of four wellswith a four-tip pipettor.

(d) Add the cytotoxic drug to the cells in columns3 to 10. Only four wells are needed for eachdrug concentration, such that rows A throughD can be used for one drug and rows E throughH for a second drug.

10. For nonadherent cells, follow steps 9b–d butprepare the drug dilution at twice the desiredfinal concentration; add 100 μL of diluted drug orcontrol medium to the 100 μL of cells already inthe wells.

11. Return the plates to the plastic box, and incubatethem for a defined exposure period.

Growth period

12. At the end of the drug exposure period, removethe medium from all of the wells containing cells,and feed the cells with 200 μL of fresh medium.Centrifuge plates containing nonadherent cells(5 min at 200 g) to pellet the cells. Then removethe medium, using a fine-gauge needle to preventdisturbance of the cell pellet.

13. Feed the plates daily for 2 to 3 PDTs.

Estimation of surviving cell numbers

14. Feed the plate with 200 μL of fresh medium at theend of the growth period, and add 50 μL of MTTto all of the wells in columns 1 to 11.

15. Wrap the plates in aluminum foil, and incubatethem for 4 h in a humidified atmosphere at 37◦C.Note that 4 h is a minimum incubation time, andplates can be left for up to 8 h.

16. Remove the medium and MTT from the wells(centrifuge for nonadherent cells), and dissolvethe remaining MTT-formazan crystals by adding200 μL of DMSO to all of the wells in columns 1to 11.

17. Add glycine buffer (25 μL per well) to all of thewells containing DMSO.

18. Record absorbance at 570 nm immediately,because the product is unstable. Use the wellsin column 1, which contain medium and MTT butno cells, to blank the plate reader.

Set up microtitration plate and incubate for about two population doublings

When cells are in exponential growth, add drug or toxin

Remove drug and allow cells to recover in growth medium

After two or three more population doublings, remove medium and repace with MTT or XTT. Read on plate reader after 3–4 h. Use absorbance to plot inhibition curve and calculate IC50

Increasing drug or toxin concentration

Controls (solvent only)

IC50, A

IC50, B

CELL A

CELL B

CELL A

CELL B

CELL A

CELL B

CELL A

CELL B

Blanks (no cells)

Blanks (no cells)

Fig. 21.5. Microtitration Assay. Stages in the assay of two differentcell lines exposed to a range of concentrations of the same drugand then allowed to recover before the estimation of survival bythe MTT reaction (see Protocol 21.4). The far left column has nocells and can be used as a blank to set the plate reader. This arrayis applicable when using plate sealers, when all wells are equivalent;however, with lids, there is a risk of an edge effect, probably due toevaporation, and it is better to leave the far left and far right columnsblank (i.e., with medium only, as in Protocol 21.4), and some usersleave the top and bottom rows blank as well. (See also Plate 17b).

Analysis of MTT assay(1) Plot a graph of the absorbance (y-axis) against the

concentration of drug (x-axis).(2) Calculate the IC50 as the drug concentration that is

required to reduce the absorbance to half that of thecontrol. The mean absorbance reading from the wellsin columns 2 and 11 is used as a control (columns 1and 12 when plate sealers are used as in Fig. 21.5). Theabsorbance values in control columns should be the same.Occasionally they are not, however, and this is taken toindicate uneven plating of cells across the plate.

(3) The absolute value of the absorbance should be plottedso that control values may be compared, but the datacan then be converted to a percentage-inhibition curve(Fig. 21.6) to normalize a series of curves.

Page 11: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 375

0 2 4 6 8 10

0

20

40

60

80

100

Inhi

bitio

n, %

of c

ontr

ol

Concentration of cytotoxin

IC50

Fig. 21.6. Percentage Inhibition Curve. Test well values arecalculated as a percentage of the controls and plotted against theconcentration of cytotoxin. Typically a sigmoid curve is obtained,and ideally the IC50 will lie in the center of the inflexion of thecurve.

Variations in MTT AssayOther applications. A similar assay has also been used to

determine cellular radiosensitivity [Carmichael et al., 1987b].MTT can be used to determine the number of cells aftera variety of treatments other than cytotoxic drug exposure,such as growth factor stimulation. However, in each case it isessential to ensure that the treatment itself does not affect theability of the cell to reduce the dye and absorbance remainslinear with cell number.

Duration of exposure. As with clonogenic assays (seeProtocol 21.3), some agents may act more quickly, andthe exposure period and recovery may be shortened. Thecells must remain in exponential growth throughout (seeSections 12.4.3, 20.9.2), and the cell concentration at theend should still be within the linear range of the MTTspectrophotometric assay. When using a cell line for thefirst time, parallel plates should be set up for cell countsto generate a growth curve (see Protocol 21.4) and forMTT-formazan absorbance to ensure that absorbance isproportional to the number of cells. If the growth curveshows that the cells are moving into the stationary phaseor the absorbance is nonlinear when plotted against cellconcentration, shorten the assay and proceed directly to step14 of Protocol 21.4.

Duration of exposure is related to the number of cellcycles that the cells have gone through during exposure andrecovery. Cell cycle time will influence the choice betweena short-form and long-form assay (Figs. 21.7, 21.8). Withrapidly dividing cells, not only will the cell density increasemore rapidly during exposure but, in addition, the responseto cycle-dependent drugs will be quicker. When first tryingan assay, it may be desirable to sample on each day of drug

exposure and recovery. If a stable IC50 is reached earlier, thenthe assay may be shortened.

End point. Sulforhodamine, a fluorescent dye that stainsprotein, can also be used to estimate the amount of protein(i.e., cells) per well on a plate reader with fluorescencedetection [Boyd, 1989]. It stains all cells and does notdiscriminate between live and dead cells. Labeling with[3H]thymidine (DNA synthesis), [3H] leucine [Freshneyet al., 1975] or [35S]methionine [Freshney & Morgan, 1978](protein synthesis), or other isotopes can be substituted forMTT reduction. Quantitation is achieved by microtitrationplate scintillation counting on a specially adapted scintillationcounter (Perkin Elmer) or by preparing an autofluorogramand reading it on a densitometer [Freshney & Morgan,1978].

In practice, it may not matter which criterion is usedfor determining viability or survival at the end of an assay;it is rather the design of the assay, such as duration ofdrug exposure and recovery, phase of the growth cycle (celldensity, growth rate, etc.), that is more important. In a shortassay with no or minimal recovery period, the endpoint mustmeasure only viable cells (e.g., MTT), but in a longer assaythe end point measures the difference between wells that haveincreased and those that have not, or have even decreased.In a monolayer assay, at least, nonviable cells will have beenlost, and the increase or decrease relative to control wellsis what is measured; whether by MTT, sulforhodamine, orisotope incorporation into DNA or protein becomes lessimportant.

Handling. A variety of automated instruments are avai-lable to reduce the handling time required per sample,including autodispensers, diluters, cell harvesters, andprogrammable plate readers (see Fig. 4.7; Appendix II:Microtitration Equipment).

0 1 2 3 4 5 6 7 8 9 10

Short form: cells with PDT <24 h

Long form: cells with PDT >24 h

Seed plates

Add cytotoxin

Remove cytotoxin

Assay viability

Days

Fig. 21.7. Assay Duration. Pattern for short-form and long-formassays. The upper diagram represents an assay that is suitable for cellwith a PDT < 24 h, and the bottom diagram represents an assaythat is suitable for cells with a PDT > 24 h, although intermediatetime scales are also possible.

Page 12: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

376 CULTURE OF ANIMAL CELLS

0 1 2 3 4 5

IC50

103

102

101

100

10−1

10−2

10−3

Cell cycles in controls from time of addition of cytotoxin

Exposure Recovery

Initial cytotoxicity

Maximum sensitivity

Fig. 21.8. Time Course of the Fall in IC50. Idealized curve for an agent with a progressive increasein cytotoxicity with time, but eventually reaching a maximum effect after three cell cycles. Not allcytotoxic drugs will conform to this pattern [Freshney et al., 1975].

21.3.6 Comparison of Microtitration withClonogenic SurvivalThe volume of medium required per sample formicrotitration is less than one-fiftieth of that required forcloning, although the number of cells is approximately thesame for both techniques. Microtitration assays are alsoshorter and more amenable to automated handling, datagathering, and analysis. Microtitration, however, is unableto distinguish between differential responses between cellswithin a population and the degree of response in eachcell—for example, a 50% inhibition of a metabolic parametercould mean that 50% of the cells respond or that each cellis inhibited by 50%—but this becomes less important in anassay with a prolonged recovery period, where the relativeincrease by cell proliferation becomes the major criterion ofsurvival.

A comparison of the IC50 derived by microtitration andplating efficiency assays showed a good correlation betweenthe two methods (Fig. 21.9) for the assay of antineoplasticdrugs [Morgan et al., 1983; Plumb et al., 1989]. Thecorrelation for IC90 was not as tight.

A significant feature of microtitration assays, particularlywith a photometric or radiometric end point, is the generationof large amounts of data, often in a format that is readilyanalyzed by computer (see Sections 20.8.1, 20.8.2). It isimportant, however, to scan the raw data as well as thedata-reduced end point because computer analysis may makedifferent assumptions or corrections to deal with aberrantdata points, which are not apparent unless the raw data areavailable for scrutiny.

21.3.7 Drug InteractionThe investigation of cytotoxicity often involves the studyof the interaction of different drugs; drug interaction is

−12 −10 −8 −6 −4

−12

−10

−8

−6

−4

Log 1

0 IC

50 [M

], cl

onog

enic

ass

ay

Log10 IC50 [M], microtitration

Fig. 21.9. Correlation between Microtitration and ClonogenicSurvival. Measurement of the IC50 values of a group of five celllines from human glioma and six drugs (vincristine, bleomycin, VM-26 epidophyllotoxin, 5-fluorouracil, methyl CCNU, mithramycin).Most of the outlying points were derived from one cell line thatlater proved to be a mixture of cell types. The broken line isthe regression, with the data points from the heterogeneous cellline omitted. Microtitration IC50 was derived by [35S]methionineincorporation [Freshney & Morgan, 1978; figure after Freshneyet al., 1982a].

readily determined by microtitration systems, in whichseveral different ratios of interacting drugs can be examinedsimultaneously. Analysis of drug interaction can be performedby using an isobologram to interpret the data [Chou, 2006].A rectilinear plot implies an additive response, whereas acurvilinear plot implies synergy if the curve dips below thepredicted line and antagonism if it goes above.

Page 13: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 377

21.4 APPLICATIONS OF CYTOTOXICITY ASSAYS

21.4.1 Anticancer Drug ScreeningDrug screening for the identification of new anticancer drugscan be a tedious and often inefficient method of discoveringnew active compounds. The trend is now more towardmonitoring effects on specific molecular targets using highthrough-put screens. However, there have been attempts toimprove screening by adopting rapid, easily automated assays,like those based on the determination of the number ofviable cells by staining the cells with MTT [Mosmann, 1983;Carmichael et al., 1987a; Plumb et al., 1989]. To furthercut down on manipulations, the MTT incubation step maybe omitted and the end point determined by measuring theamount of total protein with sulforhodamine B [Boyd, 1989].Although this method is quicker and easier than the MTTassay, it should be remembered that nonviable, and certainlynonreplicating, cells will still stain, so the assay should beconfirmed when activity is detected, using a more reliableindicator such as clonogenicity or MTT reduction.

21.4.2 Predictive Drug Testing for TumorsThe possibility has often been considered that measurementof the chemosensitivity of cells derived from a patient’s tumormight be used in designing a chemotherapeutic regime forthe patient [Freshney, 1978]. This technique has never beenexhaustively tested, although the results of small-scale trialswere encouraging [Hamburger & Salmon, 1977; Bateman etal., 1979; Hill, 1983; Thomas et al., 1985; Von Hoff et al.,1986]. What is required is the development of reliable andreproducible culture techniques for neoplastic cells from themost common tumors (e.g., breast, lung, colon), such thatcultures of pure tumor cells capable of cell proliferation overseveral cell cycles may be prepared routinely. As many ofthe cells within a tumor have a limited life span, the maintargets for chemotherapy are the clonogenic populationswith infinite repopulation capacity [Al-Hajj et al., 2004;Jones et al., 2004]. Advances in stem cell recognition (seeSection 23.1) may make isolation of tumor stem cells feasible.It was hoped that the soft agar clonogenic assay [Hamburger& Salmon, 1977] might isolate transformed stem cells forassay but, although the technique seemed initially promising,isolated clones did not have long-term regenerative capacity.Routine isolation of tumor cell populations with long-termrepopulation efficiency has yet to be achieved, but when itis, it may be possible to improve targeting and specificity ofanticancer drugs, making predictive testing more meaningful.Assays might then be performed in a high proportion ofcases, hopefully within two weeks of receipt of the biopsyand potentially effective drugs selected by genetic profiling.

The major problem, however, is one of logistics. Thenumber of patients with tumors for which the correct targetcells (1) will grow in vitro sufficiently to be tested, (2) canbe expected to respond, and (3) will produce a response thatcan be followed up, is extremely small. Hence it has proved

difficult to use any in vitro test as a predictor of responseor even to verify the reliability of the assay. The correlationof insensitivity in vitro with nonresponders is high, butfew clinicians would withhold chemotherapy because of anin vitro test, particularly when the agent in question wouldprobably not be used alone. Ultimately it may be moreprofitable to isolate tumor stem cells and screen them byexpression analysis for specific molecular targets and back thisup with an in vitro cytotoxicity assay (see also Section 26.4.2).

21.4.3 Testing PharmaceuticalsA number of pharmaceutical companies maintain a programof in vitro toxicity testing on the assumption that it mightprove more economical and ethically acceptable than animaltesting. Legislation enforcing the use of animal tests is difficultto introduce as the complexity of the wide range of effectsseen in vivo is still very difficult to model in vitro. However,there is considerable political pressure to introduce suchlegislation, and this is driving large-scale comparative surveysto determine whether any of the many existing tests maybe acceptable [Knight & Breheny, 2002; Vanparys, 2002;ECVAM, 2008; Hartung & Daston 2009].

21.5 GENOTOXICITY

Genotoxicity implies damage to DNA (mutagenesis) leadingto permanent alteration in gene expression (transformation)which may lead to malignancy (carcinogenesis). DNA stressis often accompanied by increased expression of p53 andrelated genes, which can be used to assay genotoxicity(Gentronix). In vitro assays for transformation includedemonstration of anchorage independence (see Protocols13.4, 13.5), reduced density limitation of cell proliferation(see Protocol 17.3), and evidence of mutagenesis. (See alsoTable 17.1 and Genotoxicity Testing in Appendix II.)Mutagenesis can be assayed by sister chromatid exchange;this procedure is described in Protocol 21.5, which wascontributed by Maureen Illand and Robert Brown when atthe Cancer Research UK Centre for Oncology and AppliedPharmacology, University of Glasgow, Scotland.

21.5.1 Mutagenesis Assay by Sister ChromatidExchangeSister chromatid exchanges (SCEs) are reciprocal exchangesof DNA segments between sister chromatids at identical lociduring the S-phase of the cell cycle. As SCEs are moresensitive indicators of mutagenic activity than chromosomebreaks, they have become a major tool in mutagenesis research[Latt, 1981].

With the development of the thymidine analogbromodeoxyuridine (BUdR) and its subsequent use in DNAlabeling experiments, the resolution of SCEs was greatlyimproved in comparison with previous methods, whichinvolved the incorporation of radioactive nucleotides into

Page 14: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

378 CULTURE OF ANIMAL CELLS

replicating DNA [Taylor, 1958]. Later the fluorescence plusGiemsa (FPG) technique of [Perry and Wolf [1974] forthe scoring of SCEs was enhanced, and for the first time,permanent staining of SCEs was demonstrated. Previously,during the scoring process, rapid bleaching of the fluorescentstain occurred [Latt, 1981].

The FPG method involves two distinct steps: (1) Cells arelabeled with BUdR for two complete cycles and then treatedwith colcemid to block the cells in metaphase. After BUdRexposure, the DNA of one chromatid of each chromosomecontains bromouracil in one strand, while the DNA of itssister chromatid contains bromouracil in both strands. (2)Chromosomes are then prepared from these cells and stainedwith the fluorescent dye Hoechst 33258, and then the BUdRis photodegraded with ultraviolet light; this is followed byGiemsa staining. These final steps highlight the differentialincorporation of bromouracil into the sister chromatids.DNA that contains bromouracil quenches the fluorescenceof Hoechst–DNA complexes. Therefore the chromatidcontaining bromouracil substituted in both strands fluorescesweakly and stains weakly with Giemsa, while the chromatidcontaining bromouracil in only one strand fluoresces moreintensely, degrades the BUdR, and subsequently stains darklywith Giemsa (see Plate 17e, f ). If any SCEs occur, this stainingpattern produces what are called harlequin chromosomes.

PROTOCOL 21.5. SISTER CHROMATIDEXCHANGE

OutlineTrypsinize metaphase-arrested cells that have beenlabeled with BUdR for two cell cycles, incubatethe cells in hypotonic buffer, and then fix the cells.Prepare slides of the cells, after treating the cells withHoechst 33258, and photodegrade the chromosomespreads. Stain the chromosomes with Giemsa, andvisualize on a light microscope under oil immersion.

Materials

Sterile:

� D-PBSA� PE: 10 mM EDTA in D-PBSA� Trypsin: 0.12% in PE� BUdR (Sigma): 1 mM in sterile UPW� Karyomax: Colcemid, 10 μg/mL (Invitrogen)� Growth mediumNonsterile:

� SSC, 2 × :1 : 10 dilution of 20 × SSC (seeAppendix I)

� Hypotonic buffer: 0.075 M KCl� Sorensen’s buffer: phosphate buffer, 0.066 M, pH

6.8 (tablets from Merck)

� Methanol:acetic acid: 3:1, ice cold and freshlyprepared

� Giemsa solution, 0.76%: Place 1 g of Giemsapowder (Merck) in 66 mL of glycerol and heat in awater bath at 56◦C to 60◦C for 11/2 to 2 h. Coolthe solution, and add 66 mL of absolute alcohol.

� Giemsa, diluted to 3.5% in Sorensen’s buffer,pH 6.8

� Hoechst 33258 (Sigma), 20 μg/mL, in UPW� Latex photo-mountant or adhesive� Xylene� DPX mountant (Merck)� Coverslips, 22 × 15 mm� Coplin jar� Slide rack (Thermo Fisher)� Short wave UV lamp in irradiation box

Δ Safety Note. Hoechst 33258 is carcinogenic; weigh itout and dissolve it in a fume hood.

Pretreatment

1. Seed the cells at the appropriate density (e.g.,1 × 106 cells per 75-cm2 flask), and incubate for2 days at 37◦ C.

2. Add BUdR to the growth medium at a finalconcentration of 10 μM.

3. Incubate the cells in the dark at 37◦C for a further48 h (∼2 cell cycles).

4. Add colcemid to the cells 1 to 6 h beforeharvesting, depending on the cycling time of thecells. For human cell lines, the final concentrationof colcemid should be 0.01 μg/mL.

Harvesting cells

5. Wash the cells with D-PBSA, and trypsinize themwith 1 mL of 0.12% trypsin in PE.

6. Resuspend the cells in 10 mL of growth medium.Transfer the cell suspension to 50-mL centrifugetubes.

7. Centrifuge the suspension at 1200 g for 5 min.8. Remove the supernate, leaving approximately

0.2 mL above the pellet. Flick the side of thetube to resuspend the pellet.

9. Slowly add 10 mL of hypotonic buffer(prewarmed to 37◦C), and incubate the cellsuspension for 10 to 15 min at room temperature.

10. Spin the cells in a benchtop centrifuge at 1200 gfor 5 min.

11. Remove the supernate, leaving 0.2 mL above thepellet. Flick the side of the tube to resuspend thepellet.

12. Add 10 mL of ice-cold fixative, initially drop bydrop, mixing well after each addition. Leave thetube on ice for 10 min.

Page 15: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 379

13. Repeat steps 10 through 12 once more, lettingthe cells remain in fixative overnight at 4◦C, toimprove slide preparations.

14. Spin the fixed cells at 1200 g for 5 min, andresuspend the cells in 3 to 5 mL of methanol/acetic acid.

15. Store the cells at −20◦C.

Slide preparation

16. Slides should be clean and grease-free before use,so wipe them with absolute alcohol.

17. Using a short glass Pasteur pipette, take upapproximately 500 μL of the fixed cells.

18. Hold the slide at a downward angle, and holdingthe Pasteur pipette at least 15 cm (6 in.) abovethe slide, drop 3 drops of the cell suspension ontothe slide (see Protocol 15.7).

19. Air dry the slide in the dark.20. Check the slide under phase contrast to ensure

that the metaphase spreads are evenly distributedacross the slide and that the chromosomes arewell separated.

Harlequin staining

21. Immerse the slides in a Coplin jar of Hoechst33258 at a concentration of 20 μg/mL for 10 min.(Wear gloves, as Hoechst is toxic.)

22. Transfer the slides to a slide rack, and drop 500 μLof 2× SSC onto each slide.

23. Cover the slides with a 22 × 50-mm coverslip,and seal the edges with a temporary seal, such aslatex photo-mountant, to prevent evaporation.

24. Place the slides in the slide rack, coverslipsfacing downwards, and place the slide rack ona shortwave UV box. Maintain a distance ofapproximately 4 cm between the slides and theUV source. The longer the slides are exposed toUV, the paler the pale chromatid will become;expose the slides for about 25 to 60 min.

25. Remove the coverslips from the slides, and washthe slides three times in UPW, 5 min per wash.Cover the slide holder with aluminum foil.

26. Air dry the slides in the dark.27. Stain the slides in a Coplin jar containing 3.5%

Giemsa solution in Sorensen’s buffer, pH 6.8, for3 to 5 min.

28. Carefully rinse the slides in tap water, and drainthem with a paper tissue.

29. Air dry the slides on the bench for 1 h. Dip eachslide into xylene, drop 4 drops of DPX mountant(Merck) onto the slide, and mount a 22 × 50-mmcoverslip, expressing any air bubbles with tissue.(Carry out this final step in a fume hood, as xylenefumes are toxic. Also, wear gloves.)

30. Air dry the slides in a fume hood overnight.

Analysis

31. Under the 40× objective of a light microscope,scan the slides for metaphase spreads.

32. Find an area on the slides where most of themetaphase spreads are located, and examine thisarea under oil immersion.

33. When no sister chromatid exchanges (SCEs)have occurred, each chromosome has onecontinuously staining pale chromatid and onecontinuously staining dark chromatid. One SCEhas occurred when there is one area of darkstaining and then light staining on one chromatid,and on the sister chromatid one area of lightstaining and then dark staining (see Plate 17e,f ). Each point of the discontinuity in staining isscored as one SCE.

34. Count the number of SCEs per cell and also thenumber of chromosomes per cell.

35. Larger chromosomes usually have a greaternumber of SCEs than smaller ones, and theincidence of SCEs may vary from cell to cell.Therefore, scoring SCEs per chromosome is amore accurate measure of SCE rate. SCE score iscalculated by the following formula:

Mean number of SCEs cellMean number of chromosomes/cell

Aim to score approximately 50 spreads per cellline being studied.

Variations. Pulse labeling of cells with BUdR andsubsequent staining, as described previously, can detectdifferences in early and late replicating regions ofchromosomes during the cell cycle. When cells are labeledwith BUdR at the latter part of the cell cycle, DNA thatreplicates early will stain darkly with Giemsa, due to very littleBUdR incorporation, and for regions of the chromosome thatare pulsed at the earlier stages of the cell cycle, only thoseregions that replicate their DNA early will stain faintly withGiemsa [Latt, 1973].

Additionally cells that have undergone only one cell cycleof continuous BUdR labeling show differential staining ofchromatids only at certain bands (lateral asymmetry) becauseof the differences in thymine content of the DNA [BritoBabapulle, 1981]. After photodegradation of BUdR, thefluorescent dye acridine orange can also be used to stain SCEs.With this dye, green fluorescence is observed at regions thathave double-stranded DNA and thus will have little BUdRincorporation, and red fluorescence is observed at regions thathave single-stranded DNA, which will have incorporated theBUdR. Consequently the red fluorescence is equivalent to

Page 16: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

380 CULTURE OF ANIMAL CELLS

lighter staining with Giemsa, and the green fluorescence isequivalent to the darker staining with Giemsa [Karenberg &Freelander, 1974].

21.5.2 CarcinogenicityThe potential for in vitro testing for carcinogenesis isconsiderable [Berky and Sherrod, 1977; Grafstrom, 1990a,b;Zhu et al., 1991; Tweats et al., 2007], but this is one areain which in vivo testing is far from adequate; the modelsare poor, and the tests often take weeks, or even months,to perform. The development of a satisfactory in vitro test ishampered (1) by the lack of a universally acceptable criterionfor malignant transformation in vitro and (2) by the inherentstability of human cells used as targets.

The most generally accepted tests so far assume that mostcarcinogens are mutagenic (see Section 17.3). This assumptionis the basis of the Ames test [Ames, 1980], wherein bacteriaare used as targets and activation can be carried out withliver microsomal enzyme preparations. This test has a highpredictive value, but nevertheless, dissimilarities in uptake,susceptibility, and type of cellular response have led tothe introduction of alternative tests using mammalian andhuman cells as targets, such as sister chromatid exchange (seeSection 21.5.1).

Some of these tests are also mutagenesis assays, usingsuspensions of L5178Y lymphoma cells as targets [Cole etal., 1990] and the induction of mutations or reversion,or cytological evidence of sister chromatid exchange (seeProtocol 21.5), as evidence of mutagenesis. Others [Styles,1977] have used transformation as an end point, assayingclonogenicity in suspension (see Section 17.5.1) as a criterionfor transformation. Critics of these systems say that bothuse cells that are already partially transformed as targets;even the BHK21-C13 cell used by some workers is acontinuous cell line and may not be regarded as completelynormal. Furthermore the bulk of the common cancersarise in epithelial tissues and not in connective tissuecells.

The demonstration of increased oncogene expressionor amplification, or the presence of increased or alteredoncogene products, may provide more reliable criteria, insome cases functionally related to the carcinogen. This isnow possible with microarray analysis. Likewise the deletionor mutation of suppressor genes is open to molecular analysis,where deletions or mutations in the p53, Rb, p16, and L-CAM (E-cadherin) genes would cover a high proportionof malignant transformation events. It is now feasible toconsider expression analysis and an alternative to mutagenesisin carcinogenicity testing [Nuwaysir et al., 1999; Desai et al.,2002; Vondracek et al., 2002].

21.6 INFLAMMATION

There is an increasing need for tissue culture testing to revealthe inflammatory responses that are likely to be induced by

pharmaceuticals and cosmetics with topical application or byxenobiotics that may be inhaled or ingested and may beresponsible for many forms of allergy. This is an area thatis only at the early stages of development but bears greatpromise for the future. It is a sensitive topic in more waysthan one. Animal rights groups are naturally incensed at theneedless use of large numbers of animals to test new cosmeticsthat have little benefit except commercial advantage to themanufacturer, particularly when the testing of substances (e.g.,shampoos) involves the Draize test, in which the compoundis added to a rabbit’s eye. More important, clinically, isthe apparent increase in allergenic responses produced bypharmaceuticals and xenobiotics. These responses are littleunderstood and poorly controlled, largely because of theabsence of a simple reproducible in vitro test.

Since the advent of filter well technology, several modelsfor skin (e.g., see Plate 20) and cornea have appeared [Braa andTriglia, 1991; Triglia et al., 1991; Fusenig, 1994b; Roguet etal., 1994; Kondo et al., 1997; Brinch & Elvig, 2001; Cantonet al., 2010], utilizing the facility for coculture of differentcell types that the filter well system provides. In these systemsthe interaction of an allergen or irritant with a primary target(e.g., epidermis) is presumed to initiate a paracrine response,which triggers the release of a cytokine from a second, stromalcomponent (e.g., dermis) (Fig. 21.10). This cytokine can thenbe measured by ELISA technology to monitor the degree ofthe response. Although still in the early stages of development,kits for the measurement of irritant responses are available, forexample, Epiderm (MatTek) [Koschier at al., 1997]; Episkin(Saduc) [Cohen et al., 1997]; SkinEthic [Brinch & Elvig,2001]; review [Schafer-Korting et al., 2008] (see Plate 21).

Filter well insert

Filter Test solution Target monolayer

Medium Collagen gel

Assay medium for products(cytokines, prostaglandins, NO, etc.)

Assay stromal cells for response (receptor activation, signal transduction, gene expression)

Assay target monolayer cells for response (as for stromal cells)

Fig. 21.10. Organotypic Assay. Assay system for exposing onecell layer (e.g., epidermal keratinocytes) cocultured with anotherassociated cell type (e.g., skin fibroblasts in collagen gel; see Plate 20)to an irritant and measuring the response by cytokine release (seealso Plate 21).

Page 17: Cytotoxicity...hepatoma-derived cells such as Hep-G2 or HepaRG (see Section 22.2.8). Coculture may use transfilter 3-D culture or 2-D cellular microarrays [Khetani & Bhatia, 2008]

CHAPTER 21 CYTOTOXICITY 381

Protocols for epidermal, buccal, and corneal culture suitablefor modeling irritant responses are given in Chapter 22 (seeProtocols 22.1, 22.2, 22.11).

It would seem that this type of system may be a majorarea of development, with the real prospect that allergenscreening from patients’ own skin in organotypic culturemay become possible, analysis of similar cultures from GI

tract may reveal allergens responsible for irritable bowelsyndrome, and analysis of organotypic cultures from trachealand bronchial cells may identify patient-related causes ofasthma. In each case, and in many others, there is thepossibility of specific mechanistic studies into the processesof abnormal cell interaction that typify many allergic anddegenerative diseases.