Life Science 2008 Ms

download Life Science 2008 Ms

of 14

Transcript of Life Science 2008 Ms

  • 8/6/2019 Life Science 2008 Ms

    1/14

    Centchroman induces G0/G1 arrest and Caspase-dependent Apoptosis

    involving Mitochondrial Membrane Depolarization in MCF-7 and

    MDA MB-231 Human Breast Cancer Cells

    Manisha Nigam, Vishal Ranjan, Swasti Srivastava, Ramesh Sharma, Anil K. Balapure

    Tissue and Cell Culture Unit (TCCU), Central Drug Research Institute, Lucknow, 226001, India

    Received 6 July 2007; accepted 27 November 2007

    Abstract

    Studies with Centchroman (CC) as a candidate anti-breast cancer agent are into phase III multicentric clinical trial in stage III/IV breast cancer. We

    have previously demonstrated its anti-neoplastic activity in Estrogen Receptor positive (ER+ve) MCF-7 Human Breast Cancer Cells (HBCCs). We

    now present the basis for anti-neoplastic activity of CC, mediated through apoptosis in both ER+ve/ve MCF-7 and MDA MB-231 HBCCs

    respectively, compared to Tamoxifen (TAM) as a positive control. All the experiments were performed with 48 h estrogen-deprived cells exposed to

    CC/TAM for the subsequent 48 h. Cytotoxic potential of CC was assessed through SRB assay. Cell-cycle analysis, Time-dependent cytotoxicity,

    Reactive Oxygen Species (ROS) and Mitochondrial Membrane Permeability were investigated by Flow Cytometry. Early-stage apoptosis was

    detected by AnnexinPI staining. Caspases were assayed colorimetrically whereas nuclear derangements were assessed morphologically through PI

    staining and finally by DNA fragmentation analysis. Cell viability studies confirmed the IC50 of CC in MCF-7 and MDA MB-231 cells to be 10 and

    20 M (Pb0.001) respectively, suggesting enhanced susceptibility of the former cell type to CC. FACS data reveals CC mediated G 0/G1 arrest

    (Pb0.01) along with the presence of prominent sub-G0/G1 peak (Pb0.001) in both the cell types suggesting ongoing apoptosis. Phosphatidylserine

    externalization, mitochondrial events, caspase evaluation and nuclear morphology changes reveal initiation/progression of caspase-dependentapoptosis even at a dose of 1 M which eventually leads to DNA fragmentation in both the cell types. Results demonstrate that CC induces caspase-

    dependent apoptosis in MCF-7 and MDA MB-231 cells irrespective of ER status similar to TAM in terms of anti-neoplastic activity.

    2007 Elsevier Inc. All rights reserved.

    Keywords: MCF-7; MDA MB-231; Centchroman; Tamoxifen; Apoptosis

    Introduction

    Apoptosis contributes to the anti-tumor activity of many

    chemotherapeutic drugs (Kastan and Bartek, 2004; Zhivotovskyand Kroemer, 2004; Danial and Korsmeyer, 2004; Chipuk and

    Green, 2005). Its absence activates a non-apoptotic pathway

    Necroptosis (Degterev et al., 2005). Tamoxifen (TAM) widely

    used to treat metastatic, hormone responsive breast cancer

    (Clarke et al., 2001; Macgregor and Jordan, 1998; Shiau et al.,

    1998; Muss 1992) displays its cytoproliferative/-static/-toxic

    (Taylor et al., 1984; Perry et al., 1995a) effects in ER (Estrogen

    Receptor) +ve/

    ve Human Breast Cancer Cells (HBCCs) byER-dependent/-independent pathways (Goldenberg and Froese,

    1982; Perry et al., 1995b; Obrero et al., 2002). Recently, a report

    on TAM induced rapid cell death in MCF-7 cells suggests

    caspase-independent mitochondria mediated cell death (Kallio

    et al., 2005). However, prolonged TAM administration results

    into eventual resistance to the drug and augments the risk of

    endometrial carcinoma (Bergman et al., 2000) etc. Therefore,

    the controversy about TAM and carcinogenesis has encouraged

    search for potentially safer drug for long-term treatment.

    Centchroman (CC) [67/20; INN: Ormeloxifene], a non-

    steroidal anti-estrogen (AE) with mild estrogenic and strong

    Available online at www.sciencedirect.com

    Life Sciences 82 (2008) 577590www.elsevier.com/locate/lifescie

    Corresponding author. Tel.: +91 941 506 3603; fax: +91 522 262 3405, +91

    522 262 3938.

    E-mail addresses: [email protected] (M. Nigam),

    [email protected] (V. Ranjan), [email protected]

    (S. Srivastava), [email protected] (R. Sharma),

    [email protected] (A.K. Balapure), C.D.R.I Communication No. 7029.

    0024-3205/$ - see front matter 2007 Elsevier Inc. All rights reserved.doi:10.1016/j.lfs.2007.11.028

    mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]://dx.doi.org/10.1016/j.lfs.2007.11.028http://dx.doi.org/10.1016/j.lfs.2007.11.028mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]
  • 8/6/2019 Life Science 2008 Ms

    2/14

    anti-estrogenic activity (Anand and Ray, 1977; World Health

    Organization, 1993; Central Drug Research Institute, 1994,

    1996; Kamboj et al., 1977) is one of the most extensively

    documented molecules for contraceptive purposes. However, it

    has also been reported to be accountable for 40.5% regression of

    lesions in phase III multicentric clinical trials in stage III/IV

    advanced breast cancer patients (Mishra et al., 1989; CentralDrug Research Institute, 1995). Our previous studies have

    demonstrated the anti-neoplasticity of CC in ER+ve MCF-7

    cells similar to TAM (Srivastava et al., 2004). Encouraged by

    these studies with reported excellent therapeutic index (Singh,

    2001), we have further explored the basis of its anti-

    neoplasticity, in not only MCF-7 cells but in ERve MDA

    MB-231 cells too with TAM as a positive control. Furthermore,

    normal cell types i.e. non-tumoral cell lines (HEK 293, hGF,

    MDCK) and another cancerous cell line Hep G2 were also

    evaluated for cytotoxic effects of drugs. Cytotoxicity studies

    reveal CC to be cytotoxic similar to TAM as determined by

    parallel IC50 values for both the cell types. FACS analysissuggests that CC induces G0/G1 phase arrest and apoptosis

    displayed by accumulation of cells in sub-G0/G1 fraction.

    AnnexinPropidium Iodide (PI) double staining confirms CC

    initiating apoptosis events even at 1 M substantiated by

    mitochondrial membrane depolarization studies. Interestingly,

    Reactive Oxygen Species (ROS) level imply the drug to be an

    anti-oxidant except at 1 M. Activation of caspases with

    apoptosis associated nuclear derangements and internucleoso-

    mal DNA ladder confirmed that CC induces apoptosis in MCF-

    7 and MDA MB-231 cells irrespective of ER.

    Materials and methods

    Materials

    Tamoxifen (TAM), Dulbecco's Modified Eagle's Medium

    (DMEM), N-[2-Hydroxyethyl] piperazine-N-2-ethanesulfonic

    acid (HEPES), Penicillin, Streptomycin, Gentamicin sulfate,

    Sulforhodamine-B (SRB), Phosphate Buffered Saline (PBS)

    pH 7.4, Propidium Iodide (PI), Ethidium Bromide (EtBr),

    Annexin V-FITC Apoptosis Detection Kit, Ribonuclease-A,

    Rhodamine 123 (Rh 123), 3-[(3-cholamidopropyl) dimethy-

    lammonio]-1-propanesulfonate (CHAPS), Caspase-8 substrate

    (Z-IETD-pNA), Caspase-9 substrate (Ac-LEHD-pNA), Cas-

    pase-3 substrate (Z-DEVD-pNA), Trichloroacetic acid (TCA)were purchased from Sigma Chemical Co., St. Louis, MO, USA.

    2,7-Dichlorofluorescin Diacetate (DCFDA) was from Merck

    Calbiochem and Fetal Calf Serum (FCS) was procured from

    GIBCO BRL Laboratories, New York, USA. 100 bp DNA ladder

    was sourced from Bangalore Genei, India. All other chemicals

    were of analytical grade.

    Cell culture

    HEK 293 (Human Embryonic Kidney, Epithelial), MDCK

    (MadinDarby Canine Kidney, Epithelial), Hep G2 (Human

    Hepatocellular Carcinoma), MCF-7 and MDA MB-231

    (Human Breast Cancer, Epithelial) cells were procured from

    the National Center for Cell Sciences (NCCS), Pune, India.

    These cells are routinely being cultured as reported previously

    by us (Saxena et al., 1995; Shagufta et al., 2006; Gupta et al.,

    2006; Srivastava et al., 2006). Briefly, the cells were cultured in

    DMEM, pH 7.4 containing Penicillin (100 U/ml), Streptomycin

    (100 g/ml), Gentamicin (60 g/ml) supplemented with 10%

    FCS and 10 mM HEPES in a humidified 5% CO2 incubator at37 C.

    The Primary Human Gingival Fibroblast (hGF) Cell Line was

    developed in-house from biopsy samples obtained aseptically

    after the informed consent from volunteers with clinically

    healthy gingiva.

    Briefly, the harvested gingival explants were transported

    from the local Medical and Dental School in chilled DMEM

    containing 20% fetal calf serum in minimal time on ice. They

    were then chopped into small pieces of approximately 1 mm3

    sizes in a petridish containing the same medium in a laminar

    flow to ensure sterility. Subsequently, they were transferred into

    a T-25 tissue culture flask with adequate medium for completeimmersion. The flasks were incubated in a humidified CO2incubator at 37 C with 5% CO2 and left undisturbed. By Day 5,

    a mixed population of epithelial and fibroblasts emanates in an

    around the explants as examined by Nikon Phase Contrast

    Microscopy. By Day 15, the epithelial cells perished while the

    fibroblast cells with typical oblong, flattened, spindle shaped

    morphology were retained. The medium in the flask was

    replaced with fresh medium as and when required. Complete

    monolayer was evidenced by Day 21. DMEM containing 10%

    FCS was employed to subculture the monolayer into fresh flasks

    following trypsination.

    For experimental purpose, the cells from a confluent flask

    were trypsinated and cultured for a total of 4 days. Initially forthe first 2 days, the cells were pre-cultured in phenol red-free

    DMEM containing 10% Dextran Coated Charcoal stripped FCS

    (DCC/FCS) (Soto and Sonnenschein, 1985). For the subsequent

    48 h, the cells were exposed to the ligands (CC/TAM). The cell

    number used and the concentration of ligands that the cells were

    exposed to have been described individually as below.

    Cell growth and cytotoxicity assay

    SRB (Shagufta et al., 2006) assay was conducted to ascertain

    the cytostatic/toxic/proliferative effect(s) of CC in normal cell

    types versus HBCCs and Hep G2. Briefly, 104

    cells/well wereplated in a 96-welled plate and exposed to 125 M CC/TAM.

    SRB assay was performed after completion of incubation with

    the drugs.

    Cellular morphological study

    HematoxylinEosin (HE) staining of cells for morphological

    studies was performed as previously reported by us ( Sharma

    et al., 2002). 0.2106 cells of each type were plated on sterile

    microscope cover glass in a 6-welled plate and exposed to 1

    20 M CC/TAM. The HE staining was performed, cells

    observed under Nikon Diaphot Phase Contrast Microscope and

    photographed.

    578 M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    3/14

    Study of cell-cycle kinetics and time-dependent apoptosis analysis

    To analyze the cell-cycle kinetics and apoptosis, 0.2106

    cells were plated in a 6-welled plate and exposed to 125 M

    CC/TAM. For time-dependent analysis of apoptosis, cells were

    treated with IC50 doses i.e. 10 M CC/TAM for MCF-7 and

    20 M CC/TAM for MDA MB-231 for varying time periods.Following trypsinization and washing with chilled Phosphate

    Buffered Saline (PBS) pH 7.4, the cells were permeabilized

    with chilled 70% ethanol for 30 min at 4 C. After rewashing

    with chilled PBS, the cells were resuspended in 500 l of PBS

    containing PI (40 g/ml) and RNase (100 g/ml). Flow

    Cytometry was performed on BecktonDickinson Fluorescence

    Activated CellSorter (FACS) employing the Cell Quest Software.

    Cells with hypodiploid DNA (content less than that of G0/G1-

    phase cells) were considered to be apoptotic (sub-G0/G1).

    Annexin V-FITC and PI staining analysis

    For evaluating apoptosis, 0.4106 cells of each type were

    plated onto 35 mm culture dishes containing cover slips and

    treated with 120 M CC/TAM. Following Annexin V-FITC

    and PI staining according to manufacturer's protocol, the cells

    on cover slips were analyzed on Nikon Eclipse E200

    Fluorescence Microscope and photographed.

    Modulation of Mitochondrial Membrane Potential (m) and

    intracellular Reactive Oxygen Species (ROS) generation

    Mitochondrial Membrane Potential (m) was measured by

    the uptake of Rhodamine 123 (Rh 123) as a function ofm.

    Rh 123 dye is readily sequestered by the functional mitochon-dria and subsequently washed out of the cells once the m is

    lost resulting in decreased fluorescence. 0.2106 cells of each

    type were plated in a 6-welled plate, exposed to 125 M of

    CC/TAM for 48 h, washed and finally harvested in chilled PBS

    containing 5 g/ml Rh 123. The samples were incubated at

    37 C for 15 min in dark, washed twice with chilled PBS and

    fluorescence intensities were determined on Flow Cytometer.

    Reactive Oxygen Species (ROS) generated upon CC exposure

    in the cells was determined through 2,7-Dichlorofluorescin

    Diacetate (DCFDA) staining. 0.2106 cells of both the types

    were plated in a 6-welled plate and exposed to 125 M CC/

    TAM for 24 h. Subsequently, the cells were washed twice withchilled PBS and incubated with 10 M DCFDA at 37 C for

    30 min in dark followed by washing twice with chilled PBS.

    Cells were trypsinated and analyzed through Flow Cytometer

    with excitation and emission at 490 and 530 nm respectively.

    Colorimetric caspase assay

    To analyze the role of Caspases, the cells were cultured and

    treated with 120 M CC/TAM in T-75 flasks. Activities of

    caspases were colorimetrically assayed per manufacturer's

    protocol (Sigma Chemical Co, St. Louis, MO, USA). Briefly,

    the cells were lysed in 250 l of lysis buffer (25 mM HEPES pH

    7.5, 0.1% CHAPS, 5% Sucrose, 5 mM DTT, 2 mM EDTA) at

    4 C for 30 min. 200 g of cell lysate protein was mixed in assay

    buffer in a final volume of 100 l, followed by the addition of

    10 l of 2 mM of the individual substrates specific for each

    caspase. For Caspase-8 Z-IETD-pNA, for Caspase-9 Ac-LEHD-

    pNA, and lastly for Caspase-3 Z-DEVD-pNA was respectively

    employed. Following incubation of the substrate with cell lysate

    at 37 C for 30 min, the liberated p-nitroaniline (pNA) was readat 405 nm on SpectraMAX 190 Microplate Reader.

    Nuclear morphological study

    For monitoring the drug mediated changes in nuclear

    morphology, the cells were cultured and treated similarly as

    for AnnexinPI staining analysis reported previously. The

    protocol for staining the cells was same as for FACS analysis,

    except that trypsinization was not performed in this case.

    Finally, the cover slips were analyzed under Nikon Eclipse

    E200 Fluorescence Microscope and photographed. The percen-

    tage of apoptotic nuclei was calculated by counting theapoptotic nuclei per field (particular focused area) from ten

    randomly chosen areas of the same picture.

    DNA fragmentation analysis

    Fragmented versus intact DNA was extracted according to

    Sellins and Cohen (1987) with minor modifications. Briefly, the

    cells were cultured and treated similarly as for caspase assay.

    Following lysis in the lysis buffer (10 mM TrisHCl pH 7.4,

    0.2% Triton X-100, 1 mM EDTA), the lysates were spun at

    20,000 g for 10 min at 4 C to separate the fragmented versus

    intact chromatin. Both fractions were precipitated overnight with

    1 ml 25% TCA at 4 C. The pelleted DNA was hydrolyzed with160 l of 5% TCA at 90 C for 15 min and quantified

    colorimetrically (Burton, 1956). Percent fragmentation refers

    to the ratio of DNA in the supernatant to the total DNA recovered

    in the supernatant and pellet respectively.

    For qualitative DNA fragmentation studies, the cells were

    cultured and treated similar to as in the caspase assay. The

    DNA was isolated according to Hogquist et al. (1991) with

    minor modifications. Briefly, the scraped cells were lysed in

    the lysis buffer (25 mM TrisHCl pH 7.8, 25 mM EDTA,

    0.5% Triton X-100) at 4 C for 2 h. After centrifugation at

    10,000 g, RNase-A (50 g/ml) was added to the supernatant

    for 1 h at 37 C followed by Proteinase-K (100 g/ml) for 2 hat 4 C. DNA was extracted using phenolchloroform and

    precipitated with 3 M sodium acetate and chilled absolute

    ethanol. The pelleted DNA was dissolved in TrisEDTA

    (pH 8.0) and electrophoresed on 1.8% agarose gel pre-stained

    with ethidium bromide (0.5 g/ml).

    Statistical analysis

    All the studies were performed using TAM as positive

    control and the results are expressed as meanSE from one of

    the three similar experiments each performed in triplicate.

    Student's t-test was used to determine the level of significance

    and a P-valueb0.05 was regarded as significant.

    579M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    4/14

    Results

    CC induces cytotoxicity in HBCCs

    For analyzing the effect of CC on cell survival, SRB assay

    (Fig. 1) was performed in Normal (HEK, hGF and MDCK)

    versus Cancerous cell types (Hep G2, MCF-7 and MDA MB-231 cells) employing TAM as a positive control. The results

    illustrate CC mediated dose-dependent decline in the viability

    of MCF-7 and MDA MB-231 cells versus untreated controls.

    However, the normal and Hep G2 cells were resistant to both

    CC/TAM exposure up to 25 M dose. On the contrary, a

    precipitous decline in drug treated MCF-7 cells was noticed

    from 115 M beyond which the curve plateaued off for CC/

    TAM. Correspondingly similar profile was observed with MDA

    MB-231 cells except that the leveling off was beyond 20 M.

    This suggests relatively enhanced susceptibility of MCF-7 cells

    unlike MDA MB-231 with efficacy of CC comparable to TAM.

    Besides, both the assays demonstrate that 1 M CC displays

    insignificant anti-proliferative activity in contrast to TAM

    showing estrogenic profile. In accordance with the MTT assay(data not shown), the IC50 value of CC from SRB assay for MCF-

    7 and MDA MB-231 cells was 9.08M (Pb0.001) and 20.16M

    (Pb0.001) respectivelysimilar to TAM. Cytotoxicity results were

    in agreement with those from HE staining studies (Fig. 2).

    Microscopical examination displays dose-dependent decline in

    the population of both the cell types upon exposure to CC/TAM as

    compared to control thereby revealed similar results.

    Fig. 1. Cytotoxicity evaluation of Centchroman (CC) in Cancerous [(A) MCF-7, (B) MDA MB-231 and (C) Hep G2] versus Normal [(D) MDCK, (E) HEK, (F) hGF]

    Cell Lines. 0.01 106 cells were pre-cultured for 48 h in phenol red-free DMEM (DCC treated FCS) and subsequently exposed with various doses of CC for 48 h using

    Tamoxifen (TAM) as a positive control. IC50 values were calculated using SRB Assay. Percentage survival was determined as per the formula (Absorbance of drug-

    treated cells/ Absorbance of Control cells) 100 and compared with Control, untreated cells regarded as 100%. Data shown are the mean S.E. of one of the threesimilar experiments each performed in triplicate. *Pb0.05; Pb0.01;#Pb0.001.

    580 M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    5/14

    CC induces G0/G1 arrest

    Cell-cycle analysis (Table 1 and Fig. 3A and B) reveals CC

    having comparable yet disparate influences on MCF-7 and MDA

    MB-231cells similar to TAM.Moreover,detection of hypodiploid

    (apoptotic) sub-G0/G1 population of cells confirms its role as an

    inducer of apoptosis. 1 M CC initiates G0/G1 arrest whereas S-

    and G2/M-phase inhibition in MCF-7 cells becomes more

    pronounced at 10 and 20 M (Pb0.01). However, the G0/G1

    arrested fraction decreases significantly beyond 10 M. Corre-

    spondingly in MDA MB-231 cells, CC initiates significant G0/G1 phase arrest at 10 M (Pb0.01) and G2/M-phase inhibition

    (Pb0.05) even at 1 M whose severity increases with the dose.

    Noticeably like MCF-7 cells, higher dose of CC (2025 M)

    shows a significant decline in G0/G1 arrest (Pb0.001) while

    pushing these fractions into sub-G0/G1 phase. However, the S-

    phase remains largely unmodulated with both the AEs except at

    high dose of 25 M showing abrupt decline (Pb0.01). There is a

    Fig. 2. Morphological analysis of Centchroman (CC) treated (A) MCF-7 and (B) MDA MB-231 cells after Hematoxylin and Eosin (HE) staining. Cells were pre-

    cultured for 48 h in phenol red-free DMEM(DCC treated FCS)and then exposed to different doses of CC/TAM for 48 h. Subsequently, HE staining was performed and

    observed under Phase Contrast Microscope (magnification 100). All pictures are typical of three independent experiments each performed under identical conditions.

    Table 1

    Cell-Cycle analysis of Centchroman (CC) treated MCF-7 and MDA MB-231 cells

    Treatment MCF-7 MDA MB-231

    Apoptosis Cell-Cycle Apoptosis Cell-Cycle

    Sub-G0/G1 G0/G1 S G2/M Sub-G0/G1 G0/G1 S G2/M

    Control 4.52 0.67 55.84 3.67 12.05 0.70 27.59 1.21 5.41 0.39 60.08 0.44 8.14 0.36 26.57 0.24

    TAM 1 M 3.74 0.25 64.38 4.74 13.220.56 18.640.82# 6.44 0.41 62.56 0.45 8.30 0.70 22.70 0.42*

    TAM 10 M 13.63 1.09# 67.215.02 8.420.66 10.760.34# 10.440.78 67.66 5.42* 7.65 0.91 14.65 0 .14#

    TAM 20 M 74.70 6.02# 12.881.62# 6.010.70# 6.410.73# 12.401.14 64.21 3.33 8.61 0.56 14.66 1.03#

    TAM 25 M 86.65 5.31# 2.440.14# 3.800.22# 7.010.41# 38.432.23# 46.413.15* 3.000.21 11.511.41#

    CC 1 M 3.41 0.19 63.19 4.75 10.78 1.41 22.62 1.71* 8.10 0.54 61.01 0.64 7.22 0.64 23.69 1.82*

    CC 10 M 12.02 1.01# 68.033.27 7.020.55 12.930.11# 11.050.88 70.580.43# 7.640.61 10.360.19#

    CC 20 M 80.07 6.77# 12.340.70# 3.550.24# 4.010.12# 21.460.89# 66.53 4.21* 6.04 0.55 5.57 0.98#

    CC 25 M 94.10 8.02#

    1.390.12#

    2.420.25#

    2.080.10#

    60.192.36#

    29.412.32#

    2.680.24 7.710.54#

    Briefly, 0.5 106 cells were pre-cultured for 48 h in phenol red-free DMEM (DCC treated FCS) and then exposed to different doses of CC/TAM for 48 h. Following

    incubation the cells were harvested, permeabilized, stained with Propidium Iodide (40 g/ml) and analyzed by Flow Cytometer (employing Cell Quest Software). All

    phases are represented in percentage. Data shown are the meanS.E. of one of the three similar experiments each performed in triplicate.*Pb0.05; Pb0.01;#Pb0.001.

    581M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    6/14

    Fig. 3. Cell-cycle analysis of Centchroman (CC) treated MCF-7 and MDA MB-231 cells. Briefly, 0.5106 (A) MCF-7 and (B) MDA MB-231 cells were pre-cultured

    for 48 h in phenol red-free DMEM (DCC treated FCS) and then exposed to different doses of CC/TAM for 48 h. For time-dependent apoptosis analysis, cells were

    treated with IC50 doses i.e. (C) 10 M CC/TAM for MCF-7 and (D) 20 M CC/TAM for MDA MB-231 for varying time periods. Following incubation the cells were

    harvested, permeabilized, stained with Propidium Iodide (40 g/ml) and analyzed by Flow Cytometer. All pictures are typical of three independent experiments each

    performed under identical conditions.

    582 M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    7/14

    significant yet steep decline in G0/G1, S and G2/M phase by both

    the ligands at 20M in MCF-7 cells and 25M in MDA MB-231

    cells. Time kinetics of CC induced apoptosis was explored by

    Flow Cytometry using IC50 values of CC/TAM in MCF-7 and

    MDA MB-231 cells (Fig. 3C and D). Control cells of both the type

    displayed only basal apoptosis. Significant rise in apoptotic

    fraction (Pb0.01) was observed after 12 h of drug treatment whichincreased with the time. In both the cell types, after 48 h,

    approximately 50% of the cell population was found to be

    apoptosed (Pb0.001).

    CC induces apoptosis in HBCCs

    This study analyses CC induced apoptosis in MCF-7 and

    MDA MB-231 cells. Control, untreated cells of both the types

    display rare faint green Annexin V-FITC fluorescence depicting

    non-descript, early apoptosis (Fig. 4). Fair amount of Annexin

    positive fraction at 1 M CC/TAM indicates onset of apoptosis

    in MCF-7 and MDA MB-231 cells. The magnitude of Annexin

    positive MCF-7 cells versus CC was higher than with TAM at

    1 M indicating enhanced susceptibility. CC/TAM at 10 M

    enhances PI positive fraction (red fluorescence) heralding cell

    membrane deformation. Once again CC seems remarkably

    better than TAM since more AnnexinPI positive fraction was

    observed. CC at 20 M imparts intense red staining to thenuclear region with no sign of residual green stain in MCF-7

    cells. Similar fluorescence profiling was noticed with MDA

    MB-231 cells. However, appearance of PI positive cells began at

    1M CC whose magnitude was greater at 10 and 20M than for

    TAM indicating subtle differences in the response.

    CC induces mitochondrial events

    To explore the mitochondrial events in CC induced apoptosis,

    we measured changes in m and ROS level. Control MCF-7

    and MDA MB-231cells elicited maximal Rh 123 fluorescence

    Fig. 4. Detection of Apoptosis in (A) MCF-7 and (B) MDA MB-231 cells by Annexin-V-FITC and Propidium Iodide (PI) staining. Briefly 0.4 106 cells were pre-

    cultured for 48 h in phenol red-free DMEM (DCC treated FCS) and then exposed to different doses of CC/TAM for 48 h. Subsequently, the cells were stained withAnnexin V-FITC (green fluorescence) and PI (red fluorescence) and analyzed by fluorescence microscopy (magnification 400).

    583M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    8/14

    reflecting intact, functional mitochondria (Fig. 5A). CC/TAM

    treatment results in rapid dose-dependent dissipation of mas detected by consequent decrease in mean fluorescence.

    Significant decrease in m starts at 1 M CC/TAM (Pb0.05)

    in both cell types except MDA MB-231 with TAM. However,

    abrupt decline in m begins at 10 M for both the ligands

    and cell types declining further at 20 M becoming negligibleat 25 M (Pb0.001 for all). Intracellular ROS induction

    consequent upon drug treatment (Fig. 5B and C) revealed

    similar dose-dependent decline in both the cells where CC

    accentuates ROS greater than TAM. Interestingly, MCF-7 cells

    generate significant amount of ROS (Pb0.01) at 1 M CC

    versus MDA MB-231 cells. However, ROS production declined

    beyond this dose in both the cell types but significantly in MCF-

    7 cells (Pb0.01).

    CC induces caspase-dependent apoptosis

    Caspase-8, -3 and -9 were assayed colorimetrically to

    determine their role in CC/TAM induced apoptosis. Unlike

    MCF-7 cells, MDA MB-231 cells displayed Caspase-3

    activation at 1 M CC/TAM (Pb0.05) with dose-dependent

    enhancement (Fig. 6). Contrarily, in both the cell types,

    Caspase-8 and -9 activities increased in a dose-dependent

    manner with CC/TAM. In MDA MB-231 cells like MCF-7

    cells, CC was comparatively more efficient (Pb0.001 at 10 M

    in MCF-7 and 20 M in MDA MB-231) than TAM (Pb0.01 at

    10 M in MCF-7 and 20 M in MDA MB-231) in inducingCaspase-8. However, for Caspase-9, both the drugs and cell

    types showed similar significance profile.

    Analysis of nuclear morphology

    This study was performed to ascertain the detailed effect(s) of

    CC on nuclear morphology as compared to TAM. Control MCF-

    7 and MDA MB-231 cells depict elliptical nuclei with uniform PI

    staining. CC/TAM initiates chromatin condensation at 1 M,

    peaking at 10 M and 20 M in either cell (Fig. 7A and B). Dark

    red staining in the nucleus with higher intensity around the rim,

    shrinkage (Pyknosis) along with crescenting and blebbing

    (deformed nuclear morphology) was observed. Qualitativelyand quantitatively, CC scores over TAM causing nuclear

    degeneration in either cells suggesting greater efficacy for the

    former.

    The foregoing is commensurate with the quantitative data for

    percentage of apoptotic nuclei (Fig. 7C). Smooth, rounded versus

    deformed nuclei were counted as normal versus apoptotic.

    Control MCF-7 and MDA MB-231 cells contain 13% 1.52

    and 12.9% 1.56 apoptotic nuclei respectively showing basal cell

    death. 1 M CC in MCF-7 and MDA MB-231 cells resulted in

    21%2.62 and 27%2.51 of the nuclei to apoptose respectively.

    Conversely, TAM at same dose in either cells failed to registerany

    Fig. 5. Analysis of Centchroman (CC) induced alterations in Mitochondrial

    Membrane Potential (m) and Reactive Oxygen Species (ROS). Briefly,

    0.2106 MCF-7 and MDA MB-231 cells were pre-cultured for 48 h in phenol

    red-free DMEM (DCC treated FCS treated) and then exposed to different doses

    of CC/TAM. (A) For the assessment of loss in m, trypsinized cells were

    incubated with the fluorophore Rhodamine 123 (5 g/ml) for 30 min at 37 C in

    dark. The unreacted dye was removed by washing the cells twice with chilled

    PBS and analyzed by Flow Cytometry. For measuring ROS production, the

    (B) MCF-7 and (C) MDA MB-231 cells were exposed to the ligands for 24 h,

    washed twice with chilled PBS. Following this, the cells were incubated with

    10M DCFDAfluorophorefor 30min at 37C in dark and subsequentlywashed

    twice with chilled PBS and trypsinated. Finally the stained cells were analyzed

    through Flow Cytometry. Data shown are the meanS.E. of one of the threesimilar experiments eachperformed in triplicate. *Pb0.05;Pb0.01;#Pb0.001.

    584 M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    9/14

    significant effect. In MCF-7 cells, 10 M CC caused a drasticincrease in apoptotic nuclei to 58% 3.01. In MDA MB-231 cells,

    10 M CC significantly augments these values to 29%1.03

    whereas 20M pushes these values to approximately 65% 5.02.

    In brief, it can be concluded that CC causes nuclear damage

    in MCF-7 and MDA MB-231 cells to varying extents.

    CC induces DNA fragmentation in HBCCs

    Quantitative and qualitative DNA fragmentation analysis was

    carried out to substantiate the nuclear morphological studies.

    Quantitatively the controls show negligible DNA fragmentation

    (Table 2) due to ongoing apoptosis, 17.0%1.20 in MCF-7 and19.4%0.90 in MDA MB-231 cells. 1 M CC causes insig-

    nificant fragmentation in both the cell types. However, MCF-7

    unlike MDA MB-231 cells display greater sensitivity to CC

    induced DNA fragmentation at 10 M [61.8%1.10 and 29.7%

    2.10 in MCF-7 and MDA MB-231 cells respectively]. Subse-

    quently, 20 M CC causes a slight increase to 68.0%4.20 in

    MCF-7 cells and drastic rise up to 66.1%3.20 in MDA MB-231

    cells. Similar fragmentation profile was obtained with TAM.

    Insignificant fragmentations observed at 1 M CC in both the cell

    types made us restrict our DNA laddering analysis (qualitative) to

    only 10 and 20 M doses that reveal characteristic internucleo-

    somal ladder (Fig. 8). Throughout the course of investigations, a

    common theme that has emerged is that CC has anti-neoplastic

    effect comparable to TAM and therefore affords an alternate

    approach for the management of breast cancer.

    Discussion

    This study explores the basis of anti-neoplasticity induced by

    CC in Human Breast Cancer Cells. We havealreadydemonstratedthat CC displays anti-neoplasticity in MCF-7 HBCCs (Srivastava

    et al., 2004). We now report apoptosis to be the major player in

    accomplishing CC induced cytotoxicity in MCF-7 (ER+ve) and

    MDA MB-231 (ERve) cells respectively evaluating the role of

    ER, if any, employing TAM as a positive control. Normal cell

    lines (HEK 293, hGF, MDCK) were employed for evaluating the

    cytotoxic potential of CC in non-tumoral cell types (Fig. 1).

    Moreover like MDA MB-231, the other cancerous cell line Hep

    G2, lacking functional ERs (Boix et al., 1993) was used as a

    model to study and compare the cytotoxic profile of CC. This

    strategy was employed to ascertain the cytotoxicity induced by

    CC in the absence of ER. CC and TAM are Type-I AEs withstrong anti-estrogenic and weakestrogenic potential (Clarke et al.,

    2001; World Health Organization, 1993; Kamboj et al., 1977).

    Since an apoptotic cell rarely displays all the features character-

    istic of cell death, hence several parameters have been examined.

    Cytotoxicity (Fig. 1) and HEstaining (Fig. 2) studies reveal that in

    all the cancerous cell types, CC causes a dose-dependent decline

    in the percentage of surviving cells compared to corresponding

    controls similar to TAM. Moreover in the Normal cell types,

    both the drugs elicited cytotoxicity at comparatively higher doses

    i.e. N25 M for HEK 293, Hep G2 and MDCK (data not shown)

    whereas 25 M in hGF suggesting inactivity of both CC/TAM

    towards Normal cells. This disparity of action of CC/TAM towards

    Normal versus Cancerous cells may be attributed to differences intissue-specific levels and expression patterns of Cytochrome P450

    (CYP) isoforms mediating cell-specific cytotoxicity (Sridar et al.,

    2002). CYPs' is a multigene family consisting of constitutive and

    inducible enzymes that may influence the response of tumors to

    anti-cancer drugs. This is because several anticancer agents can be

    either activated or detoxified by this enzyme system. Studies

    in vitro have implicated many CYP isoforms (e.g., CYP3A,

    CYP2D6, CYP2C9, CYP2C19, CYP2B6, and CYP1A2) in the

    biotransformation of TAM (Crewe et al., 1997). The principle

    metabolites of TAM are N-desmethyltamoxifen (formed by

    CYP3A4), 4-hydroxytamoxifen and endoxifen (formed by

    CYP2D6) (Crewe et al., 1997). Implication of CYP2B6 has alsobeen suggested to be involved in the metabolic activation of TAM

    (Desta et al., 2004). 7-desmethylcentchroman is considered as a

    possible active metabolite of CC in vivo. (Paliwal and Gupta.,

    1996). In order to exert its effects via anti-estrogenic metabolites

    that are more potent than the parent compound, TAM must be

    activated by the Cytochrome P450 system (Coezy et al., 1982) and

    therefore a similar mechanism might also be applicable for CC.

    Moreover, it has been reported that CYP1B1, CYP2B6 and

    CYP2D6 are overexpressed in tumors (Sridar et al., 2002). We

    suggest that the differential expression of CYPs may be one of the

    possible reason(s) accounting for disparity in the susceptibility of

    normal versus cancerous cells to CC/TAM. However in Hep G2

    cells, these enzymes have been reported to get deprived of during

    Fig. 6. Assessment of caspase activities in Centchroman (CC) induced apoptosis

    in (A) MCF-7 and (B) MDA MB-231cells. Cells were pre-cultured for 48 h in

    phenol red-free DMEM (DCC treated FCS) in T-75 flasks and subsequently

    exposed to different doses of CC/TAM for 48 h. Cell lysates were prepared and

    treated with Caspase-8 substrate (Z-IETD-pNA), Caspase-9 substrate (Ac-

    LEHD-pNA) and Caspase-3 substrate (Z-DEVD-pNA) individually as

    described in Materials and Methods. Activities were measured as a function

    of pNA released at 405 nm on SpectraMAX 190 Microplate Reader. Data shown

    are the meanS.E. of one of the three similar experiments each performed in

    triplicate. *Pb0.05; Pb0.01; #Pb0.001.

    585M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    10/14

    in vitro cultivation (Maria et al., 2000) thereby exhibiting

    insensitivity of drugs in this cell type. IC50 values for CC in

    MCF-7 and MDA MB-231 cells were in accordance with TAM as

    reported (Perry et al., 1995b). Disparity in the doses of CC/TAM

    eliciting anti-proliferation in MCF-7 (ER+ve) and MDA MB-231

    (ERve) cells corroborates the significance of ER (Perry et al.,

    1995b). While ER- has been established as the foremost mediator

    of proliferation, the role of ER- remains obscure. Studies have

    reported high levels of ER- in normal breast tissue and its

    frequent reduction during carcinogenesis (Park et al., 2003; Roger

    Fig. 7. Analysis of Centchroman (CC) induced alterations in nuclear morphology of (A) MCF-7 and (B) MDA MB-231 cells by Propidium Iodide (PI) staining.

    0.4106 cells were pre-cultured for 48 h in phenol red-free DMEM (DCC treated FCS) and subsequently exposed to different doses of CC/TAM for 48 h. Following

    incubation, the cells were stained with PI (40 g/ml) and analyzed by fluorescence microscope (magnification 400). Arrows indicate condensed or blebbed

    (apoptotic) nuclei. For quantification (C) pictures showing deformed nuclear morphology (condensed, blebbed or crescented morphology) were counted as Apoptotic

    whereas rounded nuclei with uniform PI staining were counted as Normal. Data shown are the meanS.E. of one of the three similar experiments each performed in

    triplicate. *Pb0.05; Pb0.01; #Pb0.001. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

    586 M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    11/14

    et al., 2001; Shaaban et al., 2003; Skliris et al., 2003). Recently, it

    has been reported that ER- enhances the ability of AEs through

    their mutual interaction i.e. ER-promotes TAM induced G1 arrest

    by enhancing G2 arrest and expression of pro-apoptotic genes

    (Gallagher et al., 2007). In MCF-7 and MDA MB-231 there are

    two types of ER- (I) Wild type [wt] and (II) a product of

    alternative splicing [corresponding to exon 5], lacking about

    139 bp of the hormone-binding domain (Vladusic et al., 1998). In

    MDAMB-231 cells unlikeMCF-7,thespliced variant is abundant,

    whereas wt is barely present. This non-functional ER- could

    thus lead to altered estrogen/anti-estrogen binding resulting in the

    lack of anti-estrogenic response. Moreover, it has been alreadyreported that ER- also increases the potency of anti-estrogens by

    shifting their dose-response curves, implying therefore that lower

    concentrations of AEs may achieve the similar response if ER-

    would have been present (Gallagher et al., 2007). This might

    explain the higher IC50 in MDA MB-231 cells because of non-

    functional ER-. Notably, the anti-estrogenicity of CC and TAM

    are comparable. Flow Studies (Table 1 and Fig. 3) confirm CC

    induced apoptosis with disparate effects on cell-cycle of MCF-7

    and MDA MB-231 cells at any given molarity. Previous studies

    demonstrate that TAM induces G0/G1 blockade in MCF-7 and

    MDA MB-231 cells (Perry et al., 1995a). Our studies reveal that

    at 1 and 10 M CC/TAM, both the cell types mostly display G0 /

    G1 arrest with concurrent dose-dependent increase in sub-G0/G1

    peak. This is possibly due to the cells under acute stress gettingample time to recover and reverting to normal status rather than

    perishing. But at higher doses (i.e., beyond 20 M), the cells are

    pushed into sub-G0/G1 i.e. apoptotic phase failing to undo the

    drug induced damage.

    In MCF-7 cells CC/TAM, besides inducing G0/G1 blockade,

    also affords S- and G2/M phase inhibition with similar

    consistency and level of significance. In MDA MB-231 cells,

    CC/TAM inhibits only the G2/M phase leaving S-phase

    unperturbed vis--vis control. Contrarily, G0/G1 arrest is

    significant from 10 M for both ligands leaving S-phase

    unperturbed only up to 20 M beyond which it significantly

    declines. In summary, analysis of cell-cycle kinetics and disparityof CC/TAM responsiveness in the two cell types may suggest

    different cell-cycle check points operable at any given time.

    Moreover, time-dependent apoptosis analysis confirms that IC50dose, in both the cell types, initiates significant apoptosis post-

    12 h of exposure. Despite varying IC50 doses, the time-dependent

    profile of CC/TAM induced apoptosis displays similar profile.

    Following 48 h incubation, approximately half of the cell

    population gets apoptosed. We have already accounted for this

    disparity of drug susceptibility of two cell types in question on the

    basis of ER status.

    Apoptotic cells undergo characteristic changes in plasma

    membrane architecture e.g. externalization of Phosphatidylser-

    ine (PS) which provides a recognition signal for phagocytosis ofapoptotic cells and stimulus for production of anti-inflammatory

    mediators (Huynh et al., 2002; Bratton and Henson, 2005).

    Apoptosis was evaluated by Annexin V-FITC and PI double

    staining (Fig. 4). Acquisition of green fluorescence (Annexin V-

    FITC) begins at a dose as low as 1 M with CC/TAM in either

    cell whose magnitude is greater for CC, indicating early

    apoptosis. Concomitant to acquisition of green fluorescence,

    Table 2

    Quantitative analysis of Centchroman (CC) induced DNA fragmentation in

    MCF-7 and MDA MB-231 cells

    Treatment Percentage DNA fragmentation

    MCF-7 MDA MB-231

    Control 17.0 1.20 19.4 0.90

    TAM 1 M 18.0 1.28 21.1 1.10TAM 10 M 53.8 4.00 29.52.00*

    TAM 20 M 65.6 5.04# 56.64.20#

    CC 1 M 22.4 2.00 23.5 1.50

    CC 10 M 61.8 1.10 29.72.10*

    CC 20 M 68.0 4.20# 66.13.20#

    The cells were pre-cultured for 48 h in phenol red-free DMEM (DCC treated

    FCS) and subsequently exposed to 120 M CC/TAM in T-75 flasks for 48 h.

    Following lysis in the lysis buffer, the lysates were spun at 20,000 gfor 10 min

    at 4 C to separate the fragmented versus intact chromatin. Both fractions were

    quantified colorimetrically as described in Materials and Methods. Percent

    fragmentation refers to the ratio of DNA in the supernatant to the total DNA

    recovered in the supernatant and pellet respectively. Data shown are the mean

    S.E. of one of the three similar experiments each performed in triplicate.

    *Pb

    0.05;

    Pb

    0.01;#

    Pb

    0.001.

    Fig. 8. Qualitativeanalysis of Centchroman (CC) induced DNA fragmentationin (A) MCF-7 and(B) MDAMB-231 cells. Briefly, the cells were pre-cultured for48 h in phenol

    red-free DMEM (DCC treated FCS) in T-75 flasks and subsequently exposed to different doses of CC/TAM for 48 h. After culmination of incubation, DNA was isolated andsubjected to 1.8% agarose gel electrophoresis as described in Materials and Methods. The gels were stained with Ethidium bromide and visualized under UV light.

    587M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    12/14

    traces of yellowish and intense red PI stained nuclear material is

    observed at 10 and 20 M of CC/TAM, showing severe cell

    membrane deformation at this stage as depicted by decrease in

    green fluorescence.

    Disruption of Mitochondrial Membrane Permeability (m) is

    an early event of apoptosis leading to cell death despite removal of

    stimulus before the cell actually succumbs (Kroemer, 2002). It has been reported that TAM dissipates m in MCF-7 cells

    (Strohmeier et al., 2002). We analyzed that CC attenuates min both the cell types beginning at 1 M and beyond (Fig. 5A).

    Cancer cells, particularlyhighly invasive or metastatic, may require

    a critical level of oxidative stress to maintain a balance between

    proliferation and apoptosis. Constitutively enhanced production of

    H2O2 (peroxides) in some cancer cells promotes proliferation and

    can induce cell-cycle arrest and/or apoptosis above a certain

    threshold (Chen et al., 2005). The pro-oxidant activity of 1 M CC

    in both the cell types (Fig. 5B and C) may be attributed to the

    persistent sub-lethal oxidative stress promoting proliferation

    in vitro (Brown and Bicknell, 2001). However, a severe declinein the ROS level beyond this dose probably suggests anti-oxidant

    behavior of the ligands. These analyses thus indicate implication of

    critical mitochondrial events in CC induced apoptosis.

    Apoptosis is typically accompanied by the activation of

    Caspases. TAM has been shown to induce Caspase-dependent

    cell death in MCF-7 and MDA MB-231 cells (Mandlekar et al.,

    2000). To evaluate the role of Caspases in CC mediated apoptosis,

    we assayed Caspase-8, -9 and -3 (Fig. 6A and B). Results illustrate

    that CC induces both Caspase-8, -9 in MCF-7 and MDA MB-231

    cells along with Caspase-3 in latter cell type.Activation of Caspase-

    8 suggests involvement of extrinsic pathway in CC induced

    apoptosis. Caspase-3 activity was undetectable in MCF-7 cells

    possibly attributed to the functional deletion of CASP-3 gene(Janicke et al., 1998). Thus, CC induces Caspase-dependent

    apoptosis irrespective of ER status.

    Nuclear morphological studies (Fig. 7A and B) aptly

    correlated with AnnexinPI analysis. 1 M CC/TAM in each

    cell type initiates apoptosis associated derangements in nuclear

    morphology as condensed chromatin gets (dense red patches)

    concentrated around the rim of the nucleus. Further increment in

    drugdosage in either cells increase the severity illustrated by rapid

    shrinkage, blebbing and crescenting and finally at maximal

    dosage residual nuclei become totally deformed shrunken,

    shriveled entities. Noticeably, MCF-7 cells seemingly succumb

    faster to the deleterious effects of CC than TAM as displayed bygreater overall reduction in size and number of nuclei with in-

    creasingdose. Similar response was observedwith the MDA MB-

    231 cells. The mechanism underlying these changes can be

    attributed to degradation of nuclear, cytoskeletal proteins e.g.

    Lamins by Caspases resulting in chromatin condensation

    (Loeffler et al., 2001; Susin et al., 1999). Besides, Apoptosis

    Inducing Factor (AIF) induces Caspase-independent nuclear con-

    densation (Danial and Korsmeyer, 2004).

    Quantitative data for the percentage of apoptotic nuclei

    (Fig. 7C) also supports the preceding. Unlike TAM, CC at

    1 M in both the cell types causes a significant increase in the

    percentage of apoptotic nuclei with respect to control. Exposure to

    10 M CC in MCF-7 cells causes 4.5 fold increase in percent

    apoptotic nuclei whereas the corresponding rise in MDA MB-231

    cells was 2.2 fold. This illustrates higher vulnerability of MCF-7

    cells to CC which is almost double to that in MDA MB-231cells

    possibly reflecting the ER status. Interestingly, CC response peaks

    at 10 M sustaining through 20 M for MCF-7 in terms

    of magnitude because at latter dose thepercentage apoptotic nuclei

    did not increase significantly. Correspondingly, in MDA MB-231cells at 20 M CC the increase was found to be 5.0 and 4.4 fold

    showing thereby the maximal response. TAM also responded

    similarly in both the cell types but the overall data confirms that

    CC augments the percentage of apoptotic nuclei more than TAM.

    Alterations in nuclear morphology may not necessarily

    involve DNA fragmentation (Srivastava et al., 2006) leading

    us to investigate drug mediated DNA fragmentation. This is

    more so since our studies provide evidence for the activation of

    Caspases which are principal actors of DNA fragmentation.

    Apart from this, Caspase-independent apoptotic DNA degrada-

    tion has been attributed to nuclear translocation of mitochondrial

    proteins, Endonuclease-G and Apoptosis Inducing Factor (AIF)(Danial and Korsmeyer, 2004). TAM induced DNA fragmenta-

    tion has been reported in MCF-7 and MDA MB-231 cells (Perry

    et al., 1995b). Quantitatively, 1 M CC in both cell types causes

    an insignificant increase in the percentage of fragmented DNA

    compared to respective controls (Table 2). However, 10 M CC

    in MCF-7 cells induces 3.6 fold increase in fraction of fragmented

    DNA whereas the corresponding increase in MDA MB-231 cells

    was 1.6 fold. This illustrates higher susceptibility of MCF-7 cells

    to CC which is approximately double as compared to MDA MB-

    231 cells reinforcing the role of ER. Further, 20M CC inMCF-7

    cells induces 3.9 fold increase whereas the corresponding

    response in MDA MB-231 cells was 3.5 fold. TAM was also

    found to exhibit similar profile in both cell types but CC seems to be more competent. These results were in agreement with

    qualitative analysis (Fig. 8) which reveals that 10 and 20 M CC

    induces DNA laddering in both cell types. Overall analysis

    suggests that CC induced apoptosis results in DNA fragmentation

    of MCF-7 and MDA MB-231 cells.

    In MCF-7 cells, DNA fragmentation might be contributed by

    Caspase-3-independent mechanisms in agreement with Caspase

    activity assay (Fig. 6). On the contrary, consistent with Caspase

    activity assay, Caspase-3-dependent/-independent or both the

    mechanisms might be active in MDA MB-231 cells. Apparent

    in phase II clinical studies with CC treatment, patients with low

    or undetectable ER responded partially thereby hypothesizingother mechanisms to be operative in the activity of AE against

    breast cancer (Mishra et al., 1989).

    Conclusion

    Our studies clearly demonstrate that CC seems to be a potent

    anti-neoplastic agent possessing remarkable similarity to TAM.

    CC induces apoptosis in MCF-7 and MDA MB-231 HBCCs

    irrespective of their ER status. However, further analyses of the

    intrinsic/extrinsic pathways in conjunction with non-genomic

    events and their regulation with CC are needed. Since

    prolonged AE therapy is usually associated with several side

    effects (Clarke et al., 2001, Macgregor and Jordan, 1998), CC

    588 M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    13/14

    seems to be a newer anti-neoplastic agent with negligible side

    effects and some associated beneficial effects. The L-enantiomer

    (Levormeloxifene) of CC has been shown to be anti-atherogenic

    and inhibits bone resorption without stimulation of endometrial

    epithelium in ovariectomized rats (Singh, 2001). Prolonged

    administration of CC to rats and rhesus monkeys does not

    induce toxicity and is therefore considered safe for chronicadministration (Mukerjee et al., 1997). Moreover, treatment of

    CC for more than 4 years on women volunteers showed

    normal hematological and biochemical parameters (Singh,

    2001). DL-Centchroman and its D- and L-enantiomers lack

    mutagenicity, genotoxicity and reduce toxic effects of known

    mutagens (Giri et al., 2001). To our knowledge, this is the first

    such report detailing the induction of apoptosis by CC in MCF-7

    and MDA MB-231 HBCCs. In conjunction with previous

    clinical trials in advanced cases of estrogen dependent breast

    cancer (Rajan, 1996a,b), our study suggests a future prospect for

    a better anti-breast cancer agent. Moreover, resolving its (CC)

    other potential isomers by understanding structure

    activityrelationships could increase the prospects of their potential

    usage (Macgregor and Jordan, 1998).

    Acknowledgements

    Director, C.D.R.I. is thanked for permitting to carry out the

    work. Mr. A. L. Vishwakarma is thanked for the experiments

    with Flow Cytometry. Manisha Nigam (31/4(763)/2004-EMR-I)

    and Vishal Ranjan (31/4(770)/2005-EMR-I) gratefully acknowl-

    edge the fellowship received from C.S.I.R. New Delhi. The

    work has been supported by funds from CMM-0018 and MOH,

    New Delhi.

    References

    Anand, N., Ray, S., 1977. Centchromana post-coital contraceptive agent.

    Indian Journal of Experimental Biology 15, 11421143.

    Bergman, L., Beelen, M.L.R., Gallee, M.P.W., Hollema, H., Benraadt, J., van

    Leeuwen, F.E., Comprehensive Cancer Centres' ALERT Group, 2000. Risk

    and prognosis of endometrial cancer after tamoxifen for breast cancer.

    Lancet 356, 881887.

    Boix, L., Bruix, J., Castells, A., Fuster, J., Bru, C., Visa, J., Rivera, F., Rodes, J.,

    1993. Sex hormone receptors in hepatocellular carcinoma. Is there a

    rationale for hormonal treatment? Journal of Hepatology 23, 187191.

    Bratton, D.L., Henson, P.M., 2005. Autoimmunity and apoptosis: refusing to go

    quietly. Nature Medicine 11, 2627.

    Brown, N.S., Bicknell, R., 2001. Hypoxia and oxidative stress in breast canceroxidative stress: its effects on the growth, metastatic potential and response

    to therapy of breast cancer. Breast Cancer Research 3, 323327.

    Burton, K.A., 1956. A study of the conditions and mechanism of the

    diphenylamine reaction for the colorimetric estimation of deoxyribonucleic

    acid. Biochemical Journal 62, 315323.

    Central Drug Research Institute, 1994. Centchroman: contraceptive, antiestro-

    gen. Drugs Future 19, 684685.

    Central Drug Research Institute, 1995. Area antifertility: centchroman phase III

    trials in advanced cancer of breast. Annual Report of the Central Drug

    Research Institute 199495. Maheshwari & Sons, Lucknow, India, p. 1.

    Central Drug Research Institute, 1996. Ormeloxifene: centchroman (EN:

    090296). Drugs Future 21, 759760.

    Chen, W.Y., Wu, C.C., Lan, Y.H., Chang, F.R., Teng, C.M., Wu, Y.C., 2005.

    Goniothalamin induces cell cycle-specific apoptosis by modulating the redox

    status in MDA MB-231cells. European Journal of Pharmacology 522, 2029.

    Chipuk, J.E., Green, D.R., 2005. Do inducers of apoptosis triggers caspase

    independent cell death? Nature Reviews. Molecular Cell Biology 6,

    268275.

    Clarke, R., Leonessa, F., Welch, J.N., Skaar, T.C., 2001. Cellular and molecular

    pharmacology of antiestrogen action and resistance. Pharmacological

    Review 53, 2571.

    Coezy, E., Borgna, J.L., Rochefort, H., 1982. Tamoxifen and metabolites in

    MCF7 cells: correlation between binding to estrogen receptor and inhibitionof cell growth. Cancer Research 42, 317323.

    Crewe, H.K., Ellis, S.W., Lennard, M.S., Tucker, G.T., 1997. Variable

    contribution of cytochromes P450 2D6, 2C9 and 3A4 to the 4-hydroxylation

    of tamoxifen by human liver microsomes. Biochemical Pharmacology 53,

    171178.

    Danial, N.N., Korsmeyer, S.J., 2004. Cell death: critical control points. Cell 116,

    205219.

    Degterev, A., Huang, Z., Boyce, M., Li, Y., Jagtap, P., Mizushima, N., Cuny, G.D.,

    Mitchison, T.J., Moskowitz, M.A., Yuan, J., 2005. Chemical inhibitor of

    nonapoptotic cell death with therapeutic potential for ischemic brain injury.

    Nature Chemical Biology 1, 112119.

    Desta, Z., Ward, B.A., Soukhova, N.V., Flockhart, D.A., 2004. Comprehensive

    evaluation of tamoxifen sequential biotransformation by the human

    cytochrome P450 system in vitro: prominent roles for CYP3A and

    CYP2D6. Journal of Pharmacology and Experimental Therapeutics 310,10621075.

    Gallagher, L.H., Valentine, C.D., Bader, S. El., Kushner, P.J., 2007. Estrogen

    receptor beta increases the efficacy of antiestrogens by effects on apoptosis

    and cell cycling in breast cancer cells. Breast Cancer Research and

    Treatment. (Printed online).

    Giri, A.K., Mukhopadhyay, A., Sun, J., Hsie, A.W., Ray, S., 2001. Antimutagenic

    effects of centchromana contraceptive and a candidatedrug forbreast cancer

    in multiple mutational assays. Mutagenesis 14, 613619.

    Goldenberg, G.J., Froese, E.K., 1982. Drug and hormone sensitivity of ER+ve

    and ERve human breast cancer cells. In Vitro Cancer Research 42,

    51475151.

    Gupta, A., Dwivedy, A., Govind, K., Sharma, R., Balapure, A.K., Singh, M.M.,

    Ray, S., 2006. Rapid synthesis of 4-benzylidene and 4-[bis-(4-methoxyphenyl)-

    methylene-2-substituted phenyl-benzopyrans as potential selective estrogen

    receptor modulators (SERMs) using McMurry coupling reaction. BioorganicMedicinal Chemistry Letters 6, 60066012.

    Hogquist, K.A., Nett, M.A., Unanue, E.R., Chaplin, D.D., 1991. Interleukin 1 is

    processed and released during apoptosis. Proceedings of the National

    Academy of Sciences of the United States of America 88, 84858489.

    Huynh, M.N., Fadok, V.A., Henson, P.M., 2002. Phosphatidylserine dependent

    ingestion of apoptotic cells promotes TGF-1 secretion and resolution of

    inflammation. Journal of Clinical Investigation 109, 4150.

    Janicke, R.U., Sprengart, M.L., Wati, M.R., Porter, A.G., 1998. Caspase-3 is

    required for DNA fragmentation and morphological changes associated with

    apoptosis. Journal of Biological Chemistry 273, 93579360.

    Kallio, A., Zheng, A., Dahllund, J., Heiskanen, K.M., Harkonen, P., 2005. Role

    of mitochondria in tamoxifen-induced rapid death of MCF-7 breast cancer

    cells. Apoptosis 10, 13951410.

    Kamboj, V.P., Setty, B.S., Chandra, H., Roy, S.K., Kar, A.B., 1977. Biological

    profile of centchroman. A new post-coital contraceptive. Indian Journal ofExperimental Biology 15, 11401150.

    Kastan, M.B., Bartek, J., 2004. Cell-cycle checkpoints and cancer. Nature 432,

    316331.

    Kroemer, G., 2002. Introduction: mitochondrial control of apoptosis. Biochimie

    84, 103104.

    Loeffler, M., Daugas, E., Susin, S.A., Zamzami, N., Metivier, D., Nieminen, A.L.,

    Brothers, G.M., Peninger, J.M., Kroemer, G., 2001. Dominant cell death

    induction by extra mitochondrially targeted apoptosis-inducing factor. FASEB

    Journal 15, 758767.

    Macgregor, J.J., Jordan, V.C., 1998. Basic guide to the mechanisms of

    antiestrogen action. Pharmacological Reviews 50, 151196.

    Mandlekar, S., Yu, R., Tan, T.H., Kong, A.N., 2000. Activation of caspase-3

    and c-Jun NH2-terminal kinase-1 signaling pathways in tamoxifen-

    induced apoptosis of human breast cancer cells. Cancer Research 60,

    59956000.

    589M. Nigam et al. / Life Sciences 82 (2008) 577590

  • 8/6/2019 Life Science 2008 Ms

    14/14

    Maria, Uhl, Christoph, Helma, Siegfried, Knasmuller, 2000. Evaluation of the

    single cell gel electrophoresis assay with human hepatoma (Hep G2) cells.

    Mutation Research 468, 213225.

    Mishra, N.C., Nigam, P.K., Gupta, R., Agarwal, A.K., Kamboj, V.P., 1989.

    Centchromana nonsteroidal anticancer agent for advanced breast cancer:

    phase II study. International Journal of Cancer 43, 781783.

    Mukerjee, S.S., Sethi, N., Srivastava, G.N., Roy, A.K., Nityanand, S., Mukherjee,

    S.K., 1997. Chronic toxicity studies of centchroman in rats and rhesusmonkeys. Indian Journal of Experimental Biology 15, 11621166.

    Muss, H.B., 1992. Endocrine therapy for advanced breast cancer: a review.

    Breast Cancer Research and Treatment 21, 1526.

    Obrero, M., Yu, D.V., Shapiro, D.J., 2002. Estrogen receptor-dependent and

    estrogen receptor-independent pathways for tamoxifen and 4-hydroxyta-

    moxifen induced programmed cell death. Journal of Biological Chemistry

    277, 4569545703.

    Paliwal, J.K., Gupta, R.C., 1996. Tissue distribution and pharmacokinetics of

    centchroman a new nonsteroidal post-coital contraceptive agent and its 7-

    desmethyl metabolite in female rats after a single oral dose. Drug

    Metabolism and Disposition 24, 148155.

    Park, B.W., Kim, K.S., Heo, M.K., Ko, S.S., Lee, K.S., Hong, S.W., Yang, W.I.,

    Kim, J.H., Kim, G.E., 2003. Expression of estrogen receptor- in normal

    mammary and tumor tissues: is it protective in breast carcinogenesis? Breast

    Cancer Research and Treatment 80, 7985.Perry, R.R., Kang, Y., Greaves, B.R., 1995a. Relationship between tamoxifen

    induced transforming growth factor1 expression, cytostasis and apoptosis

    in human breast cancer cells. British Journal of Cancer 72, 14411446.

    Perry, R.R., Kang, Y., Greaves, B., 1995b. Effects of tamoxifen on growth and

    apoptosis of estrogen-dependent and -independent human breast cancer

    cells. Annals of Surgical Oncology 2, 238245.

    Rajan, R., 1996a. Contraceptive and non-contraceptive benefits of centchroman.

    Asian Journal of Obstetrics Gynaecology 1, 6571.

    Rajan, R., 1996b. Contraceptive and non-contraceptive benefits of centchroman.

    Proceedings of the International Meet on Safe Motherhood, Guwahati, India,

    pp. 9499.

    Roger, P., Sahla, M.E., Makela, S., Gustafsson, J.A., Baldet, P., Rochefort, H.,

    2001. Decreased expression of estrogen receptor beta protein in proliferative

    preinvasive mammary tumors. Cancer Research 61, 25372541.

    Saxena, A.K., Ramchandani, S., Diwedi, A., Sharma, R., Bajpai, V.K.,Bhardwaj, K.R., Balapure, A.K., 1995. Simplified cryopreservation of

    mammalian cell lines. In Vitro Cellular & Developmental Biology. (Animal)

    31, 326329.

    Sellins, K.S., Cohen, J.J., 1987. Gene induction by gamma irradiation leads to

    DNA fragmentation in lymphocytes. Journal of Immunology 139,

    31993206.

    Shaaban, A.M., O'Neill, P.A., Davies, M.P., Sibson, R., West, C.R., Smith, P.H.,

    Foster, C.S., 2003. Declining estrogen receptor-beta expression defines

    malignant progression of human breast neoplasia. American Journal of

    Surgical Pathology 27, 15021512.

    Shagufta, Srivastava, A.K., Sharma, R., Mishra, R., Balapure, A.K., Murthy, P.S.,

    Panda, G., 2006. Substituted phenantherenes with basic amino side chains: a

    new series of antibreast cancer agents. Bioorganic & Medicinal Chemistry 14,

    14971505.

    Sharma, R., Srivastava, S., Bajpai, V.K., Balapure, A.K., 2002. Histological and

    ultrastructural regulation in rabbit endometrial explants by estrogen in

    serum-free culture. In Vitro Cellular & Developmental Biology. (Animal)

    38, 293297.

    Shiau, A.K., Barstad, D., Loria, P.M., Cheng, L., Kushner, P.J., Agard, D.A.,

    Greene, G.L., 1998. The structural basis of estrogen receptor/coactivatorrecognition and the antagonism of this interaction by tamoxifen. Cell 95,

    927937.

    Singh, M.M., 2001. Centchroman, a selective estrogen receptor modulator, as a

    contraceptive and for the management of hormone-related clinical disorders.

    Medicinal Research Reviews 21, 302347.

    Skliris, G.P., Munot, K., Bell, S.M., Carder, P.J., Lane, S., Horgan, K.,

    Lansdown, M.R., Parkes, A.T., Hanby, A.M., Markham, A.F., Speirs, V.,

    2003. Reduced expression of oestrogen receptor beta in invasive breast

    cancer and its re-expression using DNA methyl transferase inhibitors in a

    cell line model. Journal of Pathology 201, 213220.

    Soto, A.M., Sonnenschein, C., 1985. The role of estrogens on the proliferation

    of human breast tumor cells (MCF-7). Journal of Steroid Biochemistry 23,

    8794.

    Sridar, C., Kent, U.M., Notley, L.M., Gilliam, E.M., Hollenberg, P.F., 2002. Effect

    of tamoxifen on the enzymatic activity of human cytochrome CYP2B6.Journal of Pharmacology and Experimental Therapeutics 301, 945952.

    Srivastava, S., Sharma, R., Balapure, A.K., 2004. Morphological and

    biochemical basis of centchroman as a novel antineoplastic agent in MCF-

    7 human breast cancer cells. Indian Journal of Pharmacology 36, 238243.

    Srivastava, A., Tiwari, M., Sinha, R.A.,Kumar, A., Balapure, A.K., Bajpai, V.K.,

    Sharma, R., Mitra, K., Tandon, A., Godbole, M.M., 2006. Molecular iodine

    induces caspase-independent apoptosis in human breast carcinoma cells

    involving mitochondria-mediated pathway. Journal of Biological Chemistry

    281, 1976219771.

    Strohmeier, R., Roller, M., Sanger, N., Knecht, R., Kuhl, H., 2002. Modulation

    of tamoxifen induced apoptosis by peripheral benzodiazepine receptor

    ligands in breast cancer cells. Biochemical Phamacology 64, 99107.

    Susin, S.A.,Lorenzo, H.K.,Zamzami,N., Marzo, I., Snow, B.E.,Brothers, G.M.,

    Mangion,J., Jacotot, E., Costantini,P.,Loeffler, M., Larochette, N., Goodlett,

    D.R., Aebersold, R., Siderovski, D.P., Peninger, J.M., Kroemer, G., 1999.Molecular characterization of mitochondrial apoptosis-inducing factor.

    Nature 397, 441446.

    Taylor, C.M., Blanchard, B., Zava, D.T., 1984. Estrogen receptor-mediated and

    cytotoxic effects of antiestrogens tamoxifen and 4-hydroxytamoxifen.

    Cancer Research 44, 14091414.

    Vladusic, E.A., Hornby, A.E., Guerra-Vladusic, F.K., Lupu, R., 1998.

    Expression of estrogen receptor b messenger RNA variant in breast cancer.

    Cancer Research 58, 210214.

    WHO, 1993. Proposed international nonproprietary names: list 69. World

    Health Organization Drug Information 7, 87.

    Zhivotovsky, B., Kroemer, G., 2004. Apoptosis and genomic instability. Nature

    Reviews. Molecular Cell Biology 5, 752762.

    590 M. Nigam et al. / Life Sciences 82 (2008) 577590