034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

7
Please cite this article in press as: C. Valiathan, et al., A rapid survival assay to measure drug-induced cytotoxicity and cell cycle effects, DNA Repair (2011), doi:10.1016/j.dnarep.2011.11.002 ARTICLE IN PRESS G Model DNAREP-1633; No. of Pages 7 DNA Repair xxx (2011) xxx–xxx Contents lists available at SciVerse ScienceDirect DNA Repair jo u rn al hom epa ge: www.elsevier.com/locate/dnarepair A rapid survival assay to measure drug-induced cytotoxicity and cell cycle effects Chandni Valiathan a,c,d , Jose L. McFaline b,c,d , Leona D. Samson a,b,c,d,e,a Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA b Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA c Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA d Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA e Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA a r t i c l e i n f o Article history: Received 30 August 2011 Received in revised form 28 October 2011 Accepted 4 November 2011 Available online xxx Keywords: Clonogenic survival assay Cell cycle Flow cytometry a b s t r a c t We describe a rapid method to accurately measure the cytotoxicity of mammalian cells upon expo- sure to various drugs. Using this assay, we obtain survival data in a fraction of the time required to perform the traditional clonogenic survival assay, considered the gold standard. The dynamic range of the assay allows sensitivity measurements on a multi-log scale allowing better resolution of compara- tive sensitivities. Moreover, the results obtained contain additional information on cell cycle effects of the drug treatment. Cell survival is obtained from a quantitative comparison of proliferation between drug-treated and untreated cells. During the assay, cells are treated with a drug and, following a recov- ery period, allowed to proliferate in the presence of bromodeoxyuridine (BrdU). Cells that synthesize DNA in the presence of BrdU exhibit quenched Hoechst fluorescence, easily detected by flow cytometry; quenching is used to determine relative proliferation in treated vs. untreated cells. Finally, this assay can be used in high-throughput format to simultaneously screen multiple cell lines and drugs for accurate measurements of cell survival and cell cycle effects after drug treatment. © 2011 Elsevier B.V. All rights reserved. 1. Introduction Survival of cells upon exposure to toxic agents is an important phenotypic measure used to understand the biological importance of certain proteins and pathways in either preventing or enabling cell survival after toxic stress. For example, key proteins involved in DNA repair or the DNA damage response have been identified by measuring the effect of silencing or over-expressing these proteins on cell survival after DNA damage. The gold standard for assessing the survival of cells after drug treatment in such experiments continues to be the clonogenic survival assay that is extremely sensitive and has a dynamic range of several orders of magnitude. Unfortunately, it suffers from being very low throughput as well as time and labor intensive. Typically, the clonogenic survival assay takes 10–14 days and requires a large number of cell culture plates, thus limiting its practical application to a few cell lines and to a Abbreviations: PI, propidium iodide; CEN, chicken erythrocyte nuclei; BCNU, 1,3-bis(2-chloroethyl)-1-nitrosourea. Corresponding author at: Department of Biological Engineering, Koch Institute for Integrative Cancer Research and Center for Environmental Health Sciences, Mas- sachusetts Institute of Technology, Bldg 56-235, Cambridge, MA 02139, USA. Tel.: +1 617 258 7813; fax: +1 617 452 2066. E-mail addresses: [email protected] (C. Valiathan), jose [email protected] (J.L. McFaline), [email protected] (L.D. Samson). limited number of doses or agents. Moreover, for cell lines grown in suspension, or for those that fail to form colonies, the clonogenic survival assay is done by either monitoring growth from single cells or following their ability to form colonies in soft agar. These approaches are even more time intensive taking up to 2–3 weeks to complete a single experiment. Currently, the only available high-throughput techniques for measuring sensitivity involve the correlation of viability to membrane permeability (trypan blue or propidium iodide exclusion) or measurement of metabolic activity (e.g. the MTT assay). Unfortunately, metabolic activity primarily reflects mitochondrial function; in addition changes in metabolic activity do not always correlate well with cell viability after treatment, and do not differentiate between cytotoxic and static effects after treatment. Similarly, membrane permeability only takes into account cells that undergo cell death after treatment and fails to identify sensitivity due to activation of a static program such as arrest or senescence [1]. Perhaps more importantly, these methods have an inherently limited dynamic range for detection of sensitivity, generally less than a single order of magnitude vs. three or four orders of magnitude for the clonogenic survival assay. In this paper, we describe a rapid method for measuring the drug sensitivity of cells with a dynamic range comparable to that of the clonogenic survival assay. This assay has a much higher through- put compared to traditional clonogenic survival assays. In addition to obtaining survival information, it can also be used to deduce cell cycle effects of drug treatment. This method is based on the fact that 1568-7864/$ see front matter © 2011 Elsevier B.V. All rights reserved. doi:10.1016/j.dnarep.2011.11.002

Upload: marnevasconcellos

Post on 12-Nov-2014

999 views

Category:

Technology


3 download

DESCRIPTION

 

TRANSCRIPT

Page 1: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

G

D

A

Ca

b

c

d

e

a

ARRAA

KCCF

1

pocimotcsUttt

1

fsT

j

1d

ARTICLE IN PRESS Model

NAREP-1633; No. of Pages 7

DNA Repair xxx (2011) xxx– xxx

Contents lists available at SciVerse ScienceDirect

DNA Repair

jo u rn al hom epa ge: www.elsev ier .com/ locate /dnarepai r

rapid survival assay to measure drug-induced cytotoxicity and cell cycle effects

handni Valiathana,c,d, Jose L. McFalineb,c,d, Leona D. Samsona,b,c,d,e,∗

Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USADepartment of Biology, Massachusetts Institute of Technology, Cambridge, MA, USAKoch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USACenter for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USADepartment of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

r t i c l e i n f o

rticle history:eceived 30 August 2011eceived in revised form 28 October 2011ccepted 4 November 2011vailable online xxx

eywords:

a b s t r a c t

We describe a rapid method to accurately measure the cytotoxicity of mammalian cells upon expo-sure to various drugs. Using this assay, we obtain survival data in a fraction of the time required toperform the traditional clonogenic survival assay, considered the gold standard. The dynamic range ofthe assay allows sensitivity measurements on a multi-log scale allowing better resolution of compara-tive sensitivities. Moreover, the results obtained contain additional information on cell cycle effects ofthe drug treatment. Cell survival is obtained from a quantitative comparison of proliferation between

lonogenic survival assayell cyclelow cytometry

drug-treated and untreated cells. During the assay, cells are treated with a drug and, following a recov-ery period, allowed to proliferate in the presence of bromodeoxyuridine (BrdU). Cells that synthesizeDNA in the presence of BrdU exhibit quenched Hoechst fluorescence, easily detected by flow cytometry;quenching is used to determine relative proliferation in treated vs. untreated cells. Finally, this assay canbe used in high-throughput format to simultaneously screen multiple cell lines and drugs for accuratemeasurements of cell survival and cell cycle effects after drug treatment.

. Introduction

Survival of cells upon exposure to toxic agents is an importanthenotypic measure used to understand the biological importancef certain proteins and pathways in either preventing or enablingell survival after toxic stress. For example, key proteins involvedn DNA repair or the DNA damage response have been identified by

easuring the effect of silencing or over-expressing these proteinsn cell survival after DNA damage. The gold standard for assessinghe survival of cells after drug treatment in such experimentsontinues to be the clonogenic survival assay that is extremelyensitive and has a dynamic range of several orders of magnitude.nfortunately, it suffers from being very low throughput as well as

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

ime and labor intensive. Typically, the clonogenic survival assayakes 10–14 days and requires a large number of cell culture plates,hus limiting its practical application to a few cell lines and to a

Abbreviations: PI, propidium iodide; CEN, chicken erythrocyte nuclei; BCNU,,3-bis(2-chloroethyl)-1-nitrosourea.∗ Corresponding author at: Department of Biological Engineering, Koch Institute

or Integrative Cancer Research and Center for Environmental Health Sciences, Mas-achusetts Institute of Technology, Bldg 56-235, Cambridge, MA 02139, USA.el.: +1 617 258 7813; fax: +1 617 452 2066.

E-mail addresses: [email protected] (C. Valiathan),ose [email protected] (J.L. McFaline), [email protected] (L.D. Samson).

568-7864/$ – see front matter © 2011 Elsevier B.V. All rights reserved.oi:10.1016/j.dnarep.2011.11.002

© 2011 Elsevier B.V. All rights reserved.

limited number of doses or agents. Moreover, for cell lines grownin suspension, or for those that fail to form colonies, the clonogenicsurvival assay is done by either monitoring growth from singlecells or following their ability to form colonies in soft agar. Theseapproaches are even more time intensive taking up to 2–3 weeksto complete a single experiment. Currently, the only availablehigh-throughput techniques for measuring sensitivity involve thecorrelation of viability to membrane permeability (trypan blue orpropidium iodide exclusion) or measurement of metabolic activity(e.g. the MTT assay). Unfortunately, metabolic activity primarilyreflects mitochondrial function; in addition changes in metabolicactivity do not always correlate well with cell viability aftertreatment, and do not differentiate between cytotoxic and staticeffects after treatment. Similarly, membrane permeability onlytakes into account cells that undergo cell death after treatmentand fails to identify sensitivity due to activation of a static programsuch as arrest or senescence [1]. Perhaps more importantly, thesemethods have an inherently limited dynamic range for detectionof sensitivity, generally less than a single order of magnitude vs.three or four orders of magnitude for the clonogenic survival assay.

In this paper, we describe a rapid method for measuring the drugsensitivity of cells with a dynamic range comparable to that of the

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

clonogenic survival assay. This assay has a much higher through-put compared to traditional clonogenic survival assays. In additionto obtaining survival information, it can also be used to deduce cellcycle effects of drug treatment. This method is based on the fact that

Page 2: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

ARTICLE IN PRESSG Model

DNAREP-1633; No. of Pages 7

2 C. Valiathan et al. / DNA Repair xxx (2011) xxx– xxx

s in th

tradftPBaiwaasfcvo

ttpttwpaiDopcowitlgeita

2

2

t

PBS + 1% BSA containing propidium iodide (50 �g/ml) and immedi-ately analyzed by flow cytometry to obtain cell cycle profiles. None

Fig. 1. Timeline of key step

he fluorescence of Hoechst, a dye that preferentially binds AT-richegions in the DNA, is quenched when bromodeoxyuridine (BrdU),

thymine analog, is incorporated into DNA [2,3]. Cells that haveivided zero, one or two times in the presence of BrdU can be dif-erentiated based on the level of quenched Hoechst fluorescence,hus giving a measure of cell proliferation [4]. In a previous report,oot et al. [5] took advantage of the Hoechst quenching property ofrdU to measure survival of cells after exposure to a DNA damaginggent. We have extensively modified the technique to be performedn a multi-well format (96-well plate for suspension cells and 24-

ell plate for adherent cells), drastically decreasing the setup timend reducing the number of cells required for a survival curve tos little as 106–3 × 106 cells. Furthermore, we have broadened thecope of the assay so that it can simultaneously be used with dif-erent cell types and different cytotoxic agents. The assay can beompleted within half the time taken to perform a clonogenic sur-ival assay while maintaining high sensitivity and a dynamic rangef three to four logs of magnitude.

Fig. 1 shows a concise representation of the steps involved inhe assay. After cells are treated they are allowed to recover forhe duration of two doubling times and subsequently allowed toroliferate (if they can) for the duration of another two doublingimes in the presence of BrdU. To make our approach amenableo screening multiple agents with diverse mechanisms of action,e allow cells two doubling times after treatment for toxicity toresent. This allows for simultaneous detection of the toxicity ofgents that act immediately vs. those that require formation ofntermediates to slowly build up in cells. As an example, variousNA damaging agents are dependent on replication for toxicity toccur, and several days may need to pass prior to an observablehenotypic response. The duration of time prior to BrdU additionan be optimized depending on the agents to be tested. At the endf the assay, cells are gently lysed to obtain nuclei that are stainedith propidium iodide (PI) and Hoechst dye. Nuclei fluorescence

s measured by flow cytometry to quantify the percentage of cellshat have proliferated in the presence of BrdU. The relative pro-iferation rate of treated samples compared to untreated controlsives a measure of the sensitivity of cells to treatment. The ease,conomy and efficiency of this assay will enable rapid progressn systematic approaches to understanding the biological impor-ance of many proteins and pathways whose modulation leads ton observed phenotype after exposure to cytotoxic agents.

. Materials and methods

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

.1. Cell culture

The human lymphoblastoid cell lines TK6 [6] and TK6 deriva-ives (MT1 [7] and TK6 + MGMT [8]) were grown in suspension

e experimental procedure.

in RPMI medium supplemented with 10% equine serum, 1% l-glutamine and 1% penicillin–streptomycin. TK6 and MT1 cell lineswere derived from the same parent and both lack the DNA repairprotein MGMT whereas the TK6 + MGMT cell line contains recon-stituted MGMT. A genetically diverse set of human lymphoblastoidsuspension cell lines were obtained from the Coriell Institutefor Medical Research and cultured in RPMI medium (Invitrogen)supplemented with 15% FBS, 1% penicillin–streptomycin and1% l-glutamine. The cell lines were numbered 1–24 for ease ofhandling. The Coriell catalog numbers for the cell lines 1–24are respectively, GM15029, GM13036, GM15215, GM15223,GM15245, GM15224, GM15236, GM15510, GM15213, GM15221,GM15227, GM15385, GM15590, GM15038, GM15056, GM15072,GM15144, GM15216, GM15226, GM15242, GM15268, GM15324,GM15386 and GM15061. The adherent U87MG glioblastomacell line was obtained from ATCC (Rockville, MD) and grown inDMEM medium supplemented with 10% fetal bovine serum, 1%l-glutamine and 1% penicillin–streptomycin.

2.2. Determining the optimal BrdU concentration

Cells are grown in the presence of different concentrations ofBrdU (0–100 �M) for one doubling time. Cells are then lysed andstained as described below with Hoechst and propidium iodidefor flow cytometry analysis. The optimal BrdU dose is deter-mined as that which quenched Hoechst fluorescence of G1 cellsby half after one doubling time. This dose allows the effectiveresolution of cells that have undergone one division after BrdUaddition from those that have undergone none or two divisionsafter addition. This optimal dose was determined as 45 �M for thelymphoblastoid suspension cell lines and 20 �M for the U87MG celllines.

2.3. Cell cycle profile analysis by flow cytometry

BrdU is sometimes known to cause a G2/M arrest in culturedhuman cells. TK6, TK6 derivatives and U87MG cell lines were grownin the presence of the optimal BrdU concentration for at least twodoubling times, during which samples were collected at multipletime points, washed with cold PBS and fixed overnight in cold 100%ethanol. Fixed cells were washed with PBS + 1% BSA, resuspended in

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

of the cell lines showed a G2/M arrest when grown in the pres-ence of BrdU. If however such an arrest is observed, the effect canbe overcome by adding deoxycytidine at an equal concentration asthe added BrdU [3].

Page 3: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

ING Model

D

A Repa

2

fdciaTttta

2

(daemit9a5pww9meos

2

flte,ipoc2ttmadtdma

2

dadUtfa

replicate their DNA in the presence of BrdU. Therefore, as the cells

ARTICLENAREP-1633; No. of Pages 7

C. Valiathan et al. / DN

.4. Drug treatment

Cell lines were treated in duplicate at multiple doses of BCNUrom a 100 mM stock solution in 100% ethanol. A maximum of sixoses were assayed and therefore 12 wells accommodated dupli-ate exposures for each survival curve. Treatment was performedn serum free media for 1 h, after which the drug was washed awaynd the cells were returned to fresh serum-containing media.his treatment scheme can be accommodated for cytotoxic agentshat require longer exposure times as well as short-lived agentshat do not require to be washed away after treatment. The drugreatment procedure is described below for both suspension anddherent cell lines.

.4.1. Suspension cellsBefore drug treatment, cells were grown to mid-log phase

6 × 105 cells/ml for TK6 and its derivatives). 270 �l of cells at aensity of 4.5 × 105 cells/ml was plated in each well of one row of

round-bottom 96-well plate. If multiple cell lines were assayed,ach cell line was plated in one row of a 96-well plate in serum freeedia. The drug was diluted to 10× the final dose concentrations

n serum free media in another 96-well plate. 30 �l of the solu-ion with 10× the desired dose was transferred to each well in the6-well plate containing cells, after which the cells were incubatedt 37 ◦C for 1 h. After 1 h, cells were spun down at 1200 rpm for

min, drug-containing media was removed using a multi-channelipette and the cells were washed with 200 �l warm 1× PBS perell. The cells were then centrifuged and resuspended in 300 �l ofarm fresh media containing serum, transferred to a flat-bottom

6-well plate and incubated at 37 ◦C for the duration of two nor-al doubling times. If the drug is not to be washed away, the

xperiment is set up in a flat-bottom 96-well plate at a densityf 1.7 × 105 cells/ml in serum containing media, and the washingtep is omitted.

.4.2. Adherent cellsBefore drug treatment, U87MG cells were grown to 80% con-

uence, washed with warm 1× PBS, trypsinized and dilutedo 4 × 104 cells/ml. Due to the limited number of adher-nt cells that can attach to the wells in a 96-well plate

U87MG were treated in 24-well plate format. The rest of the assays continued in a 96-well format, as described below, making sam-le processing and data collection faster and more efficient. 1 mlf the diluted cells was plated in each well of a 24-well plate andells were allowed to attach overnight at 37 ◦C, 5% CO2. One such4-well plate was set up for each cell line that was assayed. Athe time of treatment, the cells were removed from the incuba-or and the media replaced with 900 �l of warm, fresh serum-free

edia. The drug was diluted to 10× the final dose concentrationsnd 100 �l of the 10× drug was added per well such that there wereuplicate wells per dose and six doses including the untreated con-rol. The cells were returned to the incubator for 1 h after which therug-containing media was replaced with warm serum-containingedia. The cells were returned to the incubator for a period equiv-

lent to two doubling times for normally growing cells.

.5. Bromodeoxyuridine addition

After allowing cells to recover for two doubling times afterrug treatment, the cells are grown in the presence of BrdU fornother two doubling times. The optimal BrdU concentration wasetermined to be 45 �M for TK6 and its derivatives and 20 �M for

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

87MG cells as described above. If BrdU concentration is greaterhan 20 �M, then BrdU must be replenished every 12 h. There-ore, after the first two doubling times, BrdU was added to TK6nd TK6 derivatives at a concentration of 45 �M (from a 10 mM

PRESSir xxx (2011) xxx– xxx 3

stock solution) and replenished by simply adding BrdU to eachwell every 12 h for the duration of two normal doubling times.Similarly BrdU was added to U87MG cells at a concentration of20 �M. Since cells become photosensitive upon BrdU addition, caremust be taken to keep cells in the dark at all times post BrdUaddition.

2.6. Nuclei isolation and staining for flow cytometry

At the end of four doubling times after drug treatment, cells weretransferred to a v-bottom 96-well plate. For suspension cells, thecells were transferred directly to a v-bottom 96-well plate using amulti-channel pipette. For adherent cells, the cell plate was spundown at 1500 rpm for 5 min to pull down any unattached cells.The media was removed and replaced with 100 �l of 0.25% trypsinEDTA. Trypsin was quenched with 200 �l of serum-containingmedia and the cells from each well in the 24-well plate weretransferred to individual wells of a v-bottom 96-well plate. Oncetransferred both cell types were spun down at 1500 rpm for 5 min,media was removed with a multi-channel pipette and cells werewashed with cold 1× PBS. The cells were then resuspended in300 �l of 1× lysis/staining buffer (0.1 M Tris–HCl pH 7.5, 0.1%Igepal CA-60, 1 mM CaCl2, 5 mM MgCl2, 0.2% BSA (w/v), 1.2 �g/mlHoechst 33258 and 1 × 104 chicken erythrocyte nuclei (CEN)/ml)and incubated on ice for 15 min after which 6 �l of 200 �g/mlpropidium iodide was added to each well using a multi-channelpipette. Two wells containing only the lysis/staining buffer and pro-pidium iodide are also prepared as blank controls. Samples weremixed well using a multi-channel pipette and analyzed on a BDLSR II flow cytometer equipped with a 96-well plate robot arm.

2.7. Data collection

Events were visualized on the side scatter vs. forward scatterplot to gate out debris (Fig. 2A), and gated on the PI-height vs. PI-area plot to exclude doublets that fall below the diagonal (Fig. 2B).30,000 events that passed these two criteria were collected andviewed on a PI-area vs. Hoechst-area plot (Fig. 2C). During datacollection, the voltages for PI and Hoechst were adjusted to posi-tion CEN at the (30 K, 30 K) point to make subsequent data analysiseasier.

3. Results

3.1. Detecting cells that have undergone zero, one or two celldivisions after drug treatment

BCNU is a DNA damaging agent commonly used in the clinicto treat glioblastoma despite the fact that it can have severe sideeffects on hematopoietic cells [9,10]. We used the multi-well assayto test the BCNU sensitivity of the human lymphoblastoid celllines TK6 and TK6 derivatives (MT1 and TK6 + MGMT) that growin suspension and the human U87MG glioblastoma cell line thatgrows attached. All data were analyzed using FlowJo (TreeStarInc.). For each cell line and drug dose, the debris and doubletswere gated out as described in Fig. 2. The remaining events wereobserved on a PI-area vs. Hoechst-area plot. Fig. 3 delineates theregions corresponding to cells that are in the first, second or thirdcell cycle after treatment, recovery and incubation with BrdU. Asseen in Fig. 3A, Hoechst fluorescence of cells decreases as they

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

replicate and divide, they move from the region labeled 1st cellcycle leftwards to the region labeled 2nd cell cycle, and so on. Foreach sample, gates were drawn as in Fig. 3C and the number ofevents in each gate was determined.

Page 4: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

ARTICLE IN PRESSG Model

DNAREP-1633; No. of Pages 7

4 C. Valiathan et al. / DNA Repair xxx (2011) xxx– xxx

Fig. 2. (A) Side scatter vs. forward scatter plot shows the position of nuclei, CEN and debris. All events in gate R1 are to be included in the data collection and analysis steps;( t be exP chanc

3t

ous

dsdcvCusotsv

Fcw

B) PI-height vs. PI-area plot shows the doublet events below the diagonal that musI-area vs. Hoechst-area plot of events included in gates R1 and R2. PI and Hoechstoordinates for all samples.

.2. Calculating the fraction of proliferated cells after drugreatment

For each cell type and dose assayed, the number of events in eachf the regions corresponding to the second and third cell cycle wassed to calculate the number of proliferating cells in the sample ashown below in formula (1).

12

(#events in cell cycle 2

#CEN

)+ 1

4

(#events in cell cycle 3

#CEN

)(1)

The formula calculates the total number of cells that haveivided after drug treatment by halving the number of cells in theecond cell cycle (these cells have undergone one cell division) andividing the number of cells in the third cell cycle by four (theseells have undergone two cell divisions). Since all samples of equalolume were spiked with the same number of CEN, the number ofEN counted per sample is proportional to the volume of samplesed for flow cytometry data collection. Therefore the cell den-ity of proliferating cells is calculated by dividing the total number

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

f proliferating cells per sample by the number of CEN counted inhat sample. Comparing the density of proliferated cells in a treatedample with that in an untreated sample gives the % control growthalue that is used to plot a survival curve.

ig. 3. (A) PI-area vs. Hoechst-area plot showing the regions that would contain nuclei inell cycle (CC3) – black; (B) plot showing the G1, S and G2 populations for each cell cyclehich the CEN and cell cycle gates have been drawn.

cluded. Events in gate R2 are included in the data collection and analysis steps; (C)nel voltages must be adjusted to position CEN at (30 K, 30 K) or any other practical

3.3. The multi-well assay has a large dynamic range, yielding logscale killing for suspension and adherent cell lines treated with acytotoxic agent

BCNU’s efficacy as a chemotherapeutic agent arises from its abil-ity to generate extremely cytotoxic DNA inter-strand crosslinks[11]. DNA crosslinks are formed in a multistep process, the first ofwhich is the formation of O6-chloroethylguanine lesions [12]. TheO6-methylguanine methyl transferase (MGMT) protein is knownto remove chloroethyl adducts from the O6 position of guanine[13,14]. Thus, MGMT provides protection against BCNU-inducedcytotoxicity, and cells lacking MGMT are particularly sensitive tothis agent [15–17]. With this in mind, we used our multi-well assayto measure BCNU sensitivity of cell lines either lacking MGMT (TK6,MT1 and U87MG) or expressing MGMT (TK6 + MGMT) to determinethe range of sensitivity accurately measured by the assay.

BCNU sensitivity of suspension (TK6 and MT1) and adherent(U87MG) cell lines that each lack MGMT was measured usingour multi-well assay. The TK6 and MT1 cell lines have previously

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

been shown to be extremely BCNU sensitive using the clonogenicsurvival assay as shown in Fig. 4A [8]. Results from the multi-wellassay, shown in Fig. 4B, are remarkably similar to those from thecolony-forming assay. Additionally, we measured BCNU sensitivity

the first cell cycle (CC1) – light grey, second cell cycle (CC2) – dark grey and third. G11, S1, G21 are G1, S and G2 in cell cycle 1 and so on. (C) Example of a sample for

Page 5: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

ARTICLE IN PRESSG Model

DNAREP-1633; No. of Pages 7

C. Valiathan et al. / DNA Repair xxx (2011) xxx– xxx 5

F traditional clonogenic survival assay reproduced from [8]; (B) killing curves for BCNUt ) killing curves for BCNU treatment of the U87MG cell line using the multi-well assay.

oMcWsMB

3m

ofsdiwamiipmslgCcTonst[icnc

3t

tid

Fig. 5. Survival curves for a panel of 24 genetically diverse lymphoblastoid cell linesexposed to multiple doses of BCNU (80, 60, 40, 20, 10 and 0 �M). Cell lines are

ig. 4. (A) Killing curves for BCNU treatment of TK6 and MT1 cell lines using thereatment of the TK6, MT1 and TK6 + MGMT cell lines using the multi-well assay; (C

f the TK6 + MGMT cell line (the TK6 cell line reconstituted withGMT) using the multi-well assay and found that TK6 + MGMT

ells show extreme resistance to BCNU, as expected (Fig. 4B).e also tested the assay on adherent cells by measuring BCNU

ensitivity of the adherent U87MG glioblastoma cell line that lacksGMT [18]. Again, as expected the U87MG cells showed extreme

CNU sensitivity (Fig. 4C), also with log-scale killing.

.4. Measuring and comparing the cytotoxicity of BCNU in aultiple cell lines

Recent efforts in our group have focused on identifying the rangef alkylation sensitivity in 24 lymphoblastoid cell lines derivedrom unrelated, healthy individuals of diverse ancestry [19]. Thistudy demonstrated that cells derived from healthy individualsisplay an unexpectedly wide range of sensitivity to the alkylat-

ng agent N-methyl-N′-nitro-N-nitrosoguanidine (MNNG), a drugith a mechanism of action analogous to the cancer chemotherapy

gent temozolomide. Temozolomide is frequently used in the treat-ent of certain brain cancers and lymphomas [20,21]. This range

n sensitivity has profound implications in the context of inter-ndividual differences in response to chemotherapy, with someatients demonstrating innate resistance or sensitivity to treat-ent. Here, we used our multi-well assay to assess the range in

ensitivity of the same 24 genetically diverse lymphoblastoid cellines to the chemotherapeutic agent BCNU. The cell lines wererown in suspension and assayed in a 96-well format as described.ells were exposed to multiple doses of BCNU in biological tripli-ates or duplicates to obtain the survival curves shown in Fig. 5.he set of 24 genetically varied cell lines displayed a wide rangef sensitivities, with survival measured over three orders of mag-itude. From this study, we identified cell lines that had extremeensitivity or resistance to BCNU, which have been used for fur-her investigation of factors affecting BCNU sensitivity or resistance22]. The multi well assay described here allows for rapid screen-ng of panels of cell lines for their sensitivity to a wide variety ofytotoxic agents. This information will aid in identifying mecha-isms of resistance and sensitivity to commonly used cytotoxichemotherapeutic agents.

.5. The multi-well assay detects cell-cycle effects of drugreatment

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

Flow cytometry plots obtained using our assay for U87MG cellsreated with BCNU show that with increasing doses of BCNU, theres not only a decrease in the total number of cells but that thisecrease is also accompanied by a steady increase in the fraction

numbered from 1 to 24 and their corresponding catalog numbers are provided inSection 2. The mean of biological triplicates or duplicates is shown and error barsdenote the standard deviation from the mean.

of cells in late S and G2/M phase of the first cell cycle (Fig. 6). Fromthese observations we can conclude that at higher BCNU doses sur-viving cells are unable to divide during the BrdU pulse and remainin the region corresponding to the first cell cycle. Moreover, sur-viving cells are arrested at late S or G2/M for the entire durationafter BrdU addition. A comparison of these results with previouslypublished results is provided in Section 4.

4. Discussion

The multi-well assay we describe here produces survivalmeasurements on a multi-log scale comparable to the clonogenicsurvival assay. As described in Section 3, BCNU sensitivities ofthe suspension cell lines TK6 and MT1 are remarkably similar

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

to that measured by the clonogenic survival assay. Moreover,the TK6 + MGMT cell line is identified as being resistant to BCNUtreatment as expected [15,17]. These results highlight three impor-tant aspects of our assay: (i) the assay can measure sensitivity

Page 6: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

ARTICLE IN PRESSG Model

DNAREP-1633; No. of Pages 7

6 C. Valiathan et al. / DNA Repair xxx (2011) xxx– xxx

F ng BCNm has apr

artmhbc

almlvsdeaerMsusdwamiwccscmifitr

cgsfimt

ig. 6. U87MG cells show increasing accumulation of cells in late S/G2 with increasiarks the area corresponding to the late S/G2 region of the first cell cycle which

egion containing the G1 cells in the first cell cycle seen in panel A.

nd resistance equally well, thus yielding accurate results thateflect those obtained from the clonogenic survival assay; (ii)he multi-well assay is capable of measuring cell sensitivity on a

ulti-log scale, thus showing the large dynamic range the assayas in contrast to other available quick assays; (iii) the assay cane used to measure cell survival of both suspension and adherentell lines to cytotoxic agents, thus increasing its applicability.

The multi-well format of the assay has a reduced setup timellowing one to simultaneously assay either a large number of cellines, doses or agents. This greatly improves cell survival measure-

ent efficiency to yield results in a fraction of the time and in aess labor-intensive manner as compared to the clonogenic sur-ival assay. As demonstrated, the assay enabled us to measure theensitivity of a panel of 24 genetically diverse cell lines to multipleoses of BCNU in biological triplicates or duplicates, and thus gen-rate survival profiles for the panel of cell lines. The results showed

wide range of sensitivities across genetically diverse cell lines,nabling us to identify cell lines with extreme BCNU sensitivity oresistance for further study [22]. In our previous study monitoringNNG-sensitivity across the panel of 24 cell lines, growth mea-

urements at one time point following exposure to one dose weresed to assess sensitivity, and we observed only a 9-fold change inensitivity from most sensitive to most resistant [23]. The approachescribed here, gave us the relative sensitivity of the cell lines toithin three orders of magnitude, analogous to clonogenic survival

ssays, within a fraction of the time. Furthermore, comparing theost sensitive and most resistant cells lines from our previous stud-

es with MNNG and the current study with BCNU, allows us to assesshether the sensitivity of cells to cytotoxic agents is specific to the

lass of cytotoxic agents used, or simply represents the response toytotoxic damage in general. As an example, while cell line 6 washown to be the most sensitive to MNNG in our previous report, thisell line clusters with the least sensitive cell lines after BCNU treat-ent. In contrast, cell line 4 appears to be among the most sensitive

n both cases. The difference observed in cell line 6 may reflect dif-erences in the ability to repair DNA interstrand crosslinks that arenduced by BCNU but not by MNNG. Currently, we are investigatinghe mechanisms of BCNU resistance and sensitivity in the two mostesistant and two most sensitive cell lines [22].

The ability to gather proliferation data from a small number ofells and with higher throughput, as compared to traditional clono-enic survival assays, has the potential to be used to measure the

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

ensitivity of cell sub-populations that have previously proven dif-cult to analyze. Glioblastoma is both the most common and mostalignant form of brain cancer. Despite aggressive therapy, this

umor frequently displays a resistant phenotype. A sub-population

U doses of 0 �M, 20 �M and 60 �M (A, B and C respectively). The box in the figuresproximately double the value of Hoechst and PI fluorescence as compared to the

of cells, dubbed glioblastoma stem cells, have been hypothesized tocontribute to this resistant phenotype [24,25]. Glioblastoma stemcells are identified and enriched from primary tumor samples dueto their ability to form suspended cell aggregates, termed neuro-spheres, in certain culture conditions [25]. The protocol describedhere would enable screening for compounds to target this resis-tant sub-population regardless of whether or not they can formcolonies.

In addition to inducing cell death, many cytotoxic agents affectregular cell cycle progression, with cells undergoing arrest eitherin the G1, S or G2/M phases of the cell cycle. The nature of the arrestchanges with the drug or cell line used, and provides insight intomechanisms of drug action and possible ways to modify cell sensi-tivity to a particular drug. The data obtained using our assay showedthat the BCNU-induced decrease in surviving U87MG glioblastomacells is accompanied by a concomitant arrest in late S/G2 phaseof the cell cycle (Fig. 6). Previous studies have shown that U87MGcells show an accumulation of cells in late S or G2/M after BCNUtreatment [26] (assayed by PI staining and cell cycle profile anal-ysis by flow-cytometry). Traditional clonogenic survival assays, aswell as more recent high-throughput survival assays, yield no infor-mation on possible cell cycle arrest from the drug treatment. Anysuch cell cycle effects have been determined by separate exper-iments usually using flow cytometry. In comparison, results fromthe multi-well assay we describe here are rich in cell-cycle informa-tion, and provide added insight into the long-term cell cycle effectsof the drug treatment.

In conclusion, we have presented a rapid and efficient methodthat takes advantage of current flow cytometry technology andproperties of proliferating cells to measure the sensitivity of bothsuspension and adherent cells to cytotoxic agents. The flexibilityof the method, its large dynamic range and its broad applicabil-ity make it a powerful tool with great potential in many differentapplications including both small and large scale screening ofsensitivity of numerous cell lines to known or suspected toxicagents.

Conflict of interest statement

The authors declare that there are no conflicts of interest.

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

Acknowledgements

The authors would like to thank Dr. John Silber of the Univer-sity of Washington for useful comments in the preparation of this

Page 7: 034 a rapid survival assay to measure drug induced cytotoxicity and cell cycle effects

ING Model

D

A Repa

mHCw

R

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

ARTICLENAREP-1633; No. of Pages 7

C. Valiathan et al. / DN

anuscript. This work was supported by the National Institute ofealth [U54-CA112967, R01-CA055042, P30-ES002109 and P30-A014051]. LDS is an American Cancer Society Professor and CVas supported by a Merck-MIT graduate fellowship.

eferences

[1] G. Nikkhah, J.C. Tonn, O. Hoffmann, H.P. Kraemer, J.L. Darling, W. Schachenmayr,R. Schonmayr, The MTT assay for chemosensitivity testing of human tumors ofthe central nervous system. Part II: evaluation of patient- and drug-specificvariables, J. Neurooncol. 13 (1992) 13–24.

[2] P.S. Rabinovitch, M. Kubbies, Y.C. Chen, D. Schindler, H. Hoehn, BrdU-Hoechstflow cytometry: a unique tool for quantitative cell cycle analysis, Exp. Cell Res.174 (1988) 309–318.

[3] M. Poot, H. Hoehn, M. Kubbies, A. Grossmann, Y. Chen, P.S. Rabinovitch,Cell-cycle analysis using continuous bromodeoxyuridine labeling and Hoechst33358-ethidium bromide bivariate flow cytometry, Methods Cell Biol. 41(1994) 327–340.

[4] M.G. Ormerod, M. Kubbies, Cell cycle analysis of asynchronous cell populationsby flow cytometry using bromodeoxyuridine label and Hoechst-propidiumiodide stain, Cytometry 13 (1992) 678–685.

[5] M. Poot, J.R. Silber, P.S. Rabinovitch, A novel flow cytometric technique for drugcytotoxicity gives results comparable to colony-forming assays, Cytometry 48(2002) 1–5.

[6] T.R. Skopek, H.L. Liber, B.W. Penman, W.G. Thilly, Isolation of a human lym-phoblastoid line heterozygous at the thymidine kinase locus: possibility for arapid human cell mutation assay, Biochem. Biophys. Res. Commun. 84 (1978)411–416.

[7] V.S. Goldmacher, R.A. Cuzick Jr., W.G. Thilly, Isolation and partial characteriza-tion of human cell mutants differing in sensitivity to killing and mutation bymethylnitrosourea and N-methyl-N′-nitro-N-nitrosoguanidine, J. Biol. Chem.261 (1986) 12462–12471.

[8] M.J. Hickman, L.D. Samson, Role of DNA mismatch repair and p53 in signalinginduction of apoptosis by alkylating agents, Proc. Natl. Acad. Sci. U. S. A. 96(1999) 10764–10769.

[9] X. Durando, J.J. Lemaire, J. Tortochaux, I. Van-Praagh, F. Kwiatkowski, C. Vincent,C. Bailly, P. Verrelle, B. Irthum, J. Chazal, J.O. Bay, High-dose BCNU followedby autologous hematopoietic stem cell transplantation in supratentorial high-grade malignant gliomas: a retrospective analysis of 114 patients, Bone MarrowTransplant. 31 (2003) 559–564.

10] F.H. Hochberg, L.M. Parker, T. Takvorian, G.P. Canellos, N.T. Zervas, High-doseBCNU with autologous bone marrow rescue for recurrent glioblastoma multi-

Please cite this article in press as: C. Valiathan, et al., A rapid survival assRepair (2011), doi:10.1016/j.dnarep.2011.11.002

forme, J. Neurosurg. 54 (1981) 455–460.11] P.E. Gonzaga, P.M. Potter, T.Q. Niu, D. Yu, D.B. Ludlum, J.A. Rafferty, G.P.

Margison, T.P. Brent, Identification of the cross-link between human O6-methylguanine-DNA methyltransferase and chloroethylnitrosourea-treatedDNA, Cancer Res. 52 (1992) 6052–6058.

[

PRESSir xxx (2011) xxx– xxx 7

12] W.P. Tong, M.C. Kirk, D.B. Ludlum, Formation of the cross-link 1-[N3-deoxycytidyl),2-[N1-deoxyguanosinyl]ethane in DNA treated with N,N′-bis(2-chloroethyl)-N-nitrosourea, Cancer Res. 42 (1982) 3102–3105.

13] B. Kaina, G. Fritz, S. Mitra, T. Coquerelle, Transfection and expression of humanO6-methylguanine-DNA methyltransferase (MGMT) cDNA in Chinese hamstercells: the role of MGMT in protection against the genotoxic effects of alkylatingagents, Carcinogenesis 12 (1991) 1857–1867.

14] S.L. Gerson, MGMT: its role in cancer aetiology and cancer therapeutics, Nat.Rev. Cancer 4 (2004) 296–307.

15] R. Maze, J.P. Carney, M.R. Kelley, B.J. Glassner, D.A. Williams, L. Samson,Increasing DNA repair methyltransferase levels via bone marrow stem celltransduction rescues mice from the toxic effects of 13-bis(2-chloroethyl)-1-nitrosourea, a chemotherapeutic alkylating agent, Proc. Natl. Acad. Sci. U. S. A.93 (1996) 206–210.

16] W. Meikrantz, M.A. Bergom, A. Memisoglu, L. Samson, O6-alkylguanine DNAlesions trigger apoptosis, Carcinogenesis 19 (1998) 369–372.

17] M. Wu, M.R. Kelley, W.K. Hansen, W.J. Martin 2nd, Reduction of BCNU toxicity tolung cells by high-level expression of O(6)-methylguanine-DNA methyltrans-ferase, Am. J. Physiol. Lung Cell. Mol. Physiol. 280 (2001) L755–L761.

18] W.P. Roos, L.F. Batista, S.C. Naumann, W. Wick, M. Weller, C.F. Menck, B. Kaina,Apoptosis in malignant glioma cells triggered by the temozolomide-inducedDNA lesion O6-methylguanine, Oncogene 26 (2007) 186–197.

19] R.C. Fry, J.P. Svensson, C. Valiathan, E. Wang, B.J. Hogan, S. Bhattacharya,J.M. Bugni, C.A. Whittaker, L.D. Samson, Genomic predictors of interindivid-ual differences in response to DNA damaging agents, Genes Dev. 22 (2008)2621–2626.

20] E.T. Wong, R. Tishler, L. Barron, J.K. Wu, Immunochemotherapy with rituximaband temozolomide for central nervous system lymphomas, Cancer 101 (2004)139–145.

21] R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K.Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K.Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O.Mirimanoff, Radiotherapy plus concomitant and adjuvant temozolomide forglioblastoma, N. Engl. J. Med. 352 (2005) 987–996.

22] C. Valiathan, P. Limsirichai and L.D. Samson, NF-Y plays a role in transcriptionchanges induced by a DNA cross-linking agent, and is shown to bind promotersof key DNA damage response genes, in preparation 2011.

23] R.C. Fry, P. Navasumrit, C. Valiathan, J.P. Svensson, B.J. Hogan, M. Luo, S.Bhattacharya, K. Kandjanapa, S. Soontararuks, S. Nookabkaew, C. Mahidol, M.Ruchirawat, L.D. Samson, Activation of inflammation/NF-kappaB signaling ininfants born to arsenic-exposed mothers, PLoS Genet. 3 (2007) e207.

24] N. Sanai, A. Alvarez-Buylla, M.S. Berger, Neural stem cells and the origin ofgliomas, N. Engl. J. Med. 353 (2005) 811–822.

25] A.M. Bleau, D. Hambardzumyan, T. Ozawa, E.I. Fomchenko, J.T. Huse, C.W.Brennan, E.C. Holland, PTEN/PI3K/Akt pathway regulates the side population

ay to measure drug-induced cytotoxicity and cell cycle effects, DNA

phenotype and ABCG2 activity in glioma tumor stem-like cells, Cell Stem Cell4 (2009) 226–235.

26] G.W. Xu, J.S. Mymryk, J.G. Cairncross, Pharmaceutical-mediated inactivation ofp53 sensitizes U87MG glioma cells to BCNU and temozolomide, Int. J. Cancer116 (2005) 187–192.