bak compensated for bax in p53-null cells to release cytochrome c for the initiation of...

11
Bak Compensated for Bax in p53-null Cells to Release Cytochrome c for the Initiation of Mitochondrial Signaling during Withanolide D-Induced Apoptosis Susmita Mondal , Kaushik Bhattacharya 1 , Asish Mallick 1 , Rajender Sangwan 2 , Chitra Mandal 1 * 1 Cancer and Cell Biology Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India, 2 Metabolic and Structural Biology Division, Council of Scientific and Industrial Research - Central Institute of Medicinal and Aromatic Plants, Lucknow, India Abstract The goal of cancer chemotherapy to induce multi-directional apoptosis as targeting a single pathway is unable to decrease all the downstream effect arises from crosstalk. Present study reports that Withanolide D (WithaD), a steroidal lactone isolated from Withania somnifera, induced cellular apoptosis in which mitochondria and p53 were intricately involved. In MOLT-3 and HCT116p53+/+ cells, WithaD induced crosstalk between intrinsic and extrinsic signaling through Bid, whereas in K562 and HCT116p532/2 cells, only intrinsic pathway was activated where Bid remain unaltered. WithaD showed pronounced activation of p53 in cancer cells. Moreover, lowered apoptogenic effect of HCT116p532/2 over HCT116p53+/+ established a strong correlation between WithaD-mediated apoptosis and p53. WithaD induced Bax and Bak upregulation in HCT116p53+/+, whereas increase only Bak expression in HCT116p532/2 cells, which was coordinated with augmented p53 expression. p53 inhibition substantially reduced Bax level and failed to inhibit Bak upregulation in HCT116p53+/+ cells confirming p53-dependent Bax and p53-independent Bak activation. Additionally, in HCT116p53+/+ cells, combined loss of Bax and Bak (HCT116Bax2Bak2) reduced WithaD-induced apoptosis and completely blocked cytochrome c release whereas single loss of Bax or Bak (HCT116Bax2Bak+/HCT116Bax+Bak2) was only marginally effective after WithaD treatment. In HCT116p532/2 cells, though Bax translocation to mitochondria was abrogated, Bak oligomerization helped the cells to release cytochrome c even before the disruption of mitochondrial membrane potential. WithaD also showed in vitro growth-inhibitory activity against an array of p53 wild type and null cancer cells and K562 xenograft in vivo. Taken together, WithaD elicited apoptosis in malignant cells through Bax/Bak dependent pathway in p53-wild type cells, whereas Bak compensated against loss of Bax in p53-null cells. Citation: Mondal S, Bhattacharya K, Mallick A, Sangwan R, Mandal C (2012) Bak Compensated for Bax in p53-null Cells to Release Cytochrome c for the Initiation of Mitochondrial Signaling during Withanolide D-Induced Apoptosis. PLoS ONE 7(3): e34277. doi:10.1371/journal.pone.0034277 Editor: Boris Zhivotovsky, Karolinska Institutet, Sweden Received November 4, 2011; Accepted February 25, 2012; Published March 29, 2012 Copyright: ß 2012 Mondal et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Funding: Council of Scientific and Industrial Research (CSIR) under IAP-0001, Systems Biology (HCP004), New Millennium Indian Technology leadership Initiative (NMITLI, TLP-004), CSIR - Indian Institute of Chemical Biology (IICB), Department of Biotechnology (DBT) under cancer Biology (GAP 235), Government of India supported this work. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Competing Interests: The authors have declared that no competing interests exist. * E-mail: [email protected] ¤ Current address: Department of Microbiology, Sammilani Mahavidyalaya, Baghajatin, Kolkata, India Introduction The primary goal of cancer chemotherapy is to trigger tumor- selective cell death [1] and the response of tumors to therapy to undergo cell death mainly depends on how fast tumor cell gets the signal to accomplish their programmed suicide. In this scenario, the ideal target of an anti-cancer agent might be mitochondria because perforation of mitochondrial membrane results in release of several death-promoting factors which ultimately either caspase dependently or independently execute cell death [2]. Therefore, regardless of the pathways involved, it is undoubtedly accepted that mitochondrial permeabilization is a central event in apoptosis and is undisputedly regulated by members of Bcl-2 family. Therefore, in a way, Bcl-2 family members and mitochondria are important targets of p53 [3]. When p53 encounter cellular stress, it restricts tumor development by responding to diverse signals for the ultimate benefit of the organism [4–5]. To achieve this cellular fate, p53 differentially activate or suppress definite sets of target genes and for this selection, multiple molecular mechanisms were involved. For example, p53 sensibly repress important anti-apoptotic proteins like Bcl-2, Bcl-xl and survivin whose ultimate outcome was identical to that of the activation of pro-apoptotic genes [6–8]. Simultaneously, p53 transactivates and upregulates different pro-apoptotic genes like Bid, Bax, Bak and Noxa [9–11], which mainly helped in mitochondrial membrane permeabilization. Besides that, Bid is a pro-apoptotic BH3-only protein which is cleaved and activated by caspase-8 or truncated by Granzyme B [12]. This truncated Bid (tBid) then interacts with Bax or ANT for the permeabilization of mitochondrial membrane and release cytochrome c along with Smac/DIABLO. Addition- ally, Bcl-xL has also been shown to inhibit tBid-induced cytochrome c release [13]. Therefore, Bid plays crucial role by combining receptor-mediated and mitochondria-mediated path- ways through cross talk. Hence, chemotherapeutic agents targeting mitochondrial death are of immense importance, because this type of agents can enforce death in cells in which upstream signals normally leading to apoptosis have been disabled. PLoS ONE | www.plosone.org 1 March 2012 | Volume 7 | Issue 3 | e34277

Upload: independent

Post on 16-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Bak Compensated for Bax in p53-null Cells to ReleaseCytochrome c for the Initiation of MitochondrialSignaling during Withanolide D-Induced ApoptosisSusmita Mondal1¤, Kaushik Bhattacharya1, Asish Mallick1, Rajender Sangwan2, Chitra Mandal1*

1 Cancer and Cell Biology Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India, 2 Metabolic

and Structural Biology Division, Council of Scientific and Industrial Research - Central Institute of Medicinal and Aromatic Plants, Lucknow, India

Abstract

The goal of cancer chemotherapy to induce multi-directional apoptosis as targeting a single pathway is unable to decreaseall the downstream effect arises from crosstalk. Present study reports that Withanolide D (WithaD), a steroidal lactoneisolated from Withania somnifera, induced cellular apoptosis in which mitochondria and p53 were intricately involved. InMOLT-3 and HCT116p53+/+ cells, WithaD induced crosstalk between intrinsic and extrinsic signaling through Bid, whereasin K562 and HCT116p532/2 cells, only intrinsic pathway was activated where Bid remain unaltered. WithaD showedpronounced activation of p53 in cancer cells. Moreover, lowered apoptogenic effect of HCT116p532/2 over HCT116p53+/+established a strong correlation between WithaD-mediated apoptosis and p53. WithaD induced Bax and Bak upregulationin HCT116p53+/+, whereas increase only Bak expression in HCT116p532/2 cells, which was coordinated with augmentedp53 expression. p53 inhibition substantially reduced Bax level and failed to inhibit Bak upregulation in HCT116p53+/+ cellsconfirming p53-dependent Bax and p53-independent Bak activation. Additionally, in HCT116p53+/+ cells, combined loss ofBax and Bak (HCT116Bax2Bak2) reduced WithaD-induced apoptosis and completely blocked cytochrome c releasewhereas single loss of Bax or Bak (HCT116Bax2Bak+/HCT116Bax+Bak2) was only marginally effective after WithaDtreatment. In HCT116p532/2 cells, though Bax translocation to mitochondria was abrogated, Bak oligomerization helpedthe cells to release cytochrome c even before the disruption of mitochondrial membrane potential. WithaD also showed invitro growth-inhibitory activity against an array of p53 wild type and null cancer cells and K562 xenograft in vivo. Takentogether, WithaD elicited apoptosis in malignant cells through Bax/Bak dependent pathway in p53-wild type cells, whereasBak compensated against loss of Bax in p53-null cells.

Citation: Mondal S, Bhattacharya K, Mallick A, Sangwan R, Mandal C (2012) Bak Compensated for Bax in p53-null Cells to Release Cytochrome c for the Initiationof Mitochondrial Signaling during Withanolide D-Induced Apoptosis. PLoS ONE 7(3): e34277. doi:10.1371/journal.pone.0034277

Editor: Boris Zhivotovsky, Karolinska Institutet, Sweden

Received November 4, 2011; Accepted February 25, 2012; Published March 29, 2012

Copyright: � 2012 Mondal et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permitsunrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Funding: Council of Scientific and Industrial Research (CSIR) under IAP-0001, Systems Biology (HCP004), New Millennium Indian Technology leadership Initiative(NMITLI, TLP-004), CSIR - Indian Institute of Chemical Biology (IICB), Department of Biotechnology (DBT) under cancer Biology (GAP 235), Government of Indiasupported this work. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Competing Interests: The authors have declared that no competing interests exist.

* E-mail: [email protected]

¤ Current address: Department of Microbiology, Sammilani Mahavidyalaya, Baghajatin, Kolkata, India

Introduction

The primary goal of cancer chemotherapy is to trigger tumor-

selective cell death [1] and the response of tumors to therapy to

undergo cell death mainly depends on how fast tumor cell gets the

signal to accomplish their programmed suicide. In this scenario,

the ideal target of an anti-cancer agent might be mitochondria

because perforation of mitochondrial membrane results in release

of several death-promoting factors which ultimately either caspase

dependently or independently execute cell death [2]. Therefore,

regardless of the pathways involved, it is undoubtedly accepted

that mitochondrial permeabilization is a central event in apoptosis

and is undisputedly regulated by members of Bcl-2 family.

Therefore, in a way, Bcl-2 family members and mitochondria

are important targets of p53 [3]. When p53 encounter cellular

stress, it restricts tumor development by responding to diverse

signals for the ultimate benefit of the organism [4–5]. To achieve

this cellular fate, p53 differentially activate or suppress definite sets

of target genes and for this selection, multiple molecular

mechanisms were involved. For example, p53 sensibly repress

important anti-apoptotic proteins like Bcl-2, Bcl-xl and survivin

whose ultimate outcome was identical to that of the activation of

pro-apoptotic genes [6–8]. Simultaneously, p53 transactivates and

upregulates different pro-apoptotic genes like Bid, Bax, Bak and

Noxa [9–11], which mainly helped in mitochondrial membrane

permeabilization. Besides that, Bid is a pro-apoptotic BH3-only

protein which is cleaved and activated by caspase-8 or truncated

by Granzyme B [12]. This truncated Bid (tBid) then interacts with

Bax or ANT for the permeabilization of mitochondrial membrane

and release cytochrome c along with Smac/DIABLO. Addition-

ally, Bcl-xL has also been shown to inhibit tBid-induced

cytochrome c release [13]. Therefore, Bid plays crucial role by

combining receptor-mediated and mitochondria-mediated path-

ways through cross talk. Hence, chemotherapeutic agents

targeting mitochondrial death are of immense importance,

because this type of agents can enforce death in cells in which

upstream signals normally leading to apoptosis have been disabled.

PLoS ONE | www.plosone.org 1 March 2012 | Volume 7 | Issue 3 | e34277

Withanolide D (C4b-C5b,C6b-epoxy-1-oxo-,20b, dihydroxy-

20S,22R-witha-2,24-dienolide; WithaD) is a steroidal lactone

isolated from the leaves of Ashwagandha (Withania somnifera Dunal,

Solanaceae), one of the most reputed medicinal plant of Ayurveda

[14]. The herb forms essential constituent of more than 100

traditional medicine formulations [15–19]. Earlier we have

demonstrated that WithaD effectively induced apoptosis in

leukemia (MOLT-4 and K562) and in primary cells from patients

irrespective of their lineages. Also, we had shown that WithaD-

induced apoptosis was through the early accumulation of ceramide

by the activation of neutral-sphingomyelinase [20]. Here we

wanted to explore the mitochondrial pathway as targeting a single

pathway is unable to decrease all the downstream effect arises from

signal cross talk.

We identified differences in activation pattern of intrinsic and

extrinsic pathways in MOLT-3 and K562 cells, which was

correlated with p53 status as revealed by HCT116p53+/+ and

HCT116p532/2 cells. WithaD robustly enhanced p53 expres-

sion and also induced p53-dependent Bax and independent Bak

upregulation. Additionally, WithaD elicited apoptosis through a

Bax/Bak dependent way in p53-proficient cells, whereas Bak

compensated against loss of Bax in p53-null cells. Moreover,

WithaD induced in vitro growth-inhibitory activity against an array

of p53 wild type (wt) and null cancer cells and inhibits tumor

growth in athymic nude mice. Hence, we suggest that WithaD is a

potent anti-cancer agent that induced mitochondria-mediated

apoptosis both in p53wt and null cells.

Results

WithaD-mediated apoptosis commence through theinvolvement of mitochondria

To specify the role of mitochondria in WithaD-induced

apoptosis, we first investigated the expression of pro- and anti-

apoptotic molecules in leukemic cells (MOLT-3 and K562).

Results showed that in both the cells, expression of Bcl-xl and Bcl-

2 were reduced dose dependently after WithaD treatment.

However, in MOLT-3 cells, WithaD showed a prominent increase

in Bax and Bak levels. In contrast, Bax level remains unchanged

after increasing WithaD treatment in K562, whereas Bak was

significantly upregulated (Fig. 1B). These results suggested that in

leukemia, mitochondria related Bcl-2 family proteins were

differentially involved in WithaD-mediated cell death.

To locate the specific death cascade through which WithaD

exerts its action, we investigated the key molecules of intrinsic and

extrinsic pathways. Results showed that WithaD proteolytically

cleaved inactive pro-caspase-9 (47 kDa) after 15 hr of treatment at

0.5 mM dose to form the active 35–37 kDa fragment in MOLT-3.

Moreover, WithaD induced the proteolytic processing of execu-

tioner caspases-7 and -3 and also stimulated a dose-dependent

hydrolysis of the 116 kDa PARP to 85 kDa fragment. In contrast,

in K562 cells, the activation of pro-caspase 9, -7, -3 and PARP

cleavage were only observed at higher concentration of WithaD

(2 mM), suggesting the involvement of intrinsic pathway in both

the cells, only the amount of WithaD required to activate the

pathway was different (Fig. 1C).

Next, we tested the possibility of involvement of extrinsic

pathway in WithaD-mediated apoptosis. In MOLT-3, proteolytic

cleavage of pro-caspase 8 to its active 43 kDa fragment was

observed within 0.5–1 mM WithaD treatment. Moreover, results

showed significant reduction in total Bid expression with increased

dose of WithaD. In contrast, in K562 cells, we did not detect any

active caspase 8 fragments, only the reduced level of pro-caspase 8

was observed. Additionally, Bid level was also remain unaltered

(Fig. 1D). These results suggest that, possibly caspase 9-mediated

intrinsic pathway playing the central role in both the cells in

WithaD-mediated apoptosis.

To confirm the possibility of involvement of intrinsic pathway in

WithaD-induced cell death, we specifically inhibit the caspase -9, -

8 and -3 and measured the apoptosis in MOLT-3 and K562 cells.

Caspase -9 inhibition by LEHD-FMK significantly reduced

WithaD-induced apoptosis both in MOLT-3 and K562, while

caspase-8 inhibition by IETD-FMK only marginally affects

(Fig. 1E). Additionally, caspase-3 inhibition by DEVD-CHO

markedly reduced WithaD-induced apoptosis suggested WithaD-

mediated specific activation of caspase cascade, which ultimately

executed through caspase-3 activation. In summary, these results

confirmed that the contribution of mitochondria-mediated

pathway executed the WithaD induced apoptosis both in

MOLT-3 and K562, although the accomplishment was different.

p53 is a critical mediator of WithaD-induced apoptosisA consistent difference in the activation of intrinsic and caspase-

8-mediated death receptor pathway in MOLT-3 and K562 cells

along with difference in Bax activation prompted us to find the

reason(s) behind this discrepencie(s). In intrinsic pathway, p53

target crucial subset of Bcl-2 family genes including Bax, Bid, Bcl-

xl etc. [21] or induce the oligomerization of Bak at mitochondrial

level. Therefore, we envisioned that the discrepancies between

MOLT-3 and K562 may be due to the p53 status, as K562 are

p53-null, whereas MOLT-3 is p53 wild type (wt). Therefore, we

next assessed the effect of WithaD on the expression of p53 in

MOLT-3 and K562 along with two other p53 expressing cells

(HCT116 and U87MG). WithaD dose-dependently enhanced p53

expression in MOLT-3, HCT116 and U87MG whereas in K562

there was no p53 expression as expected (Fig. 2A).

Next we used HCT116p53+/+ and a stably p53 knockdown

HCT116p532/2 cells and checked their p53 status (Fig. 2B). We

then investigated the intrinsic and caspase-8 mediated death

receptor pathways in these cells to specifically demonstrate

whether p53 status really made any differences. In

HCT116p53+/+ cells, WithaD induced the activation of cas-

pase-9, caspase-3, caspase-7 and also stimulated the processing of

PARP in similar manner as was observed in MOLT-3.

Interestingly, activation of caspase-9 was only occurred at higher

dose and caspase-3, caspase-7 and PARP cleavage proceed

subsequently in HCT116p532/2 cells as was in K562 (Fig. 2C).

In case of caspase-8-mediated death receptor pathway, active

caspase-8 fragment was formed at 2.0 mM in HCT116p53+/+cells, while in HCT116p532/2 cells, only the level of pro-

caspase-8 was reduced. Additionally, reduction of total Bid

expression was observed merely in the highest dose in

116p532/2 cells (Fig. 2D). Therefore, these results suggested

that disparities in the activation of intrinsic and extrinsic pathway

might be due to the variation in p53 status.

Next, we evaluated whether presence of p53 really made a

difference in WithaD-induced apoptosis. Results showed that only

26.1% HCT116p53+/+ cells were viable at 24 hr at 5 mM dose

whereas at identical conditions 43.8% HCT116p532/2 cells were

viable (Figure S1). Similar differences were observed after 48 hr of

WithaD treatment. Moreover, this trend of differences i.e. lower cell

death in HCT116p532/2 compared to HCT116p53+/+ were

also observed in annexinV-PI staining (Fig. 2E) which was further

reflected in the changes of cell morphology (Fig. 2F). With

increasing dose of WithaD, HCT116p53+/+ cells lost their

adherent property, detached from the substratum and also rounded

up. In contrast, HCT116p532/2 cells showed more adherences to

its niche with extended normal cellular morphology. These results

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 2 March 2012 | Volume 7 | Issue 3 | e34277

altogether confirmed that p53 crucially regulate WithaD-mediated

apoptosis.

WithaD induced p53-dependent Bax and p53-independent Bak activation

Having established that p53 is a crucial mediator and

mitochondria playing important role in WithaD-induced apopto-

sis, we next attempt to find the missing link between these two

events. Hence, efforts were made to identify the role of different

p53 downstream effector molecules related to mitochondrial

apoptosis along with Bcl-2. Results showed that in

HCT116p532/2 and HCT116p53+/+ cells, expression of Bcl-

xl and Bcl-2 were reduced dose dependently after WithaD

treatment. Being a p53 target molecule, Bcl-xl’s reduction

irrespective of p53 status could be explained by the fact that

there may be other factors regulating Bcl-xl. Also, an increase in

p21 level was observed in both the cells. However, WithaD

showed a prominent dose-dependent increase in Bax and Bak

levels in constitutive p53 expressing cells. In contrast, in p53-null

cells, Bax level remains unchanged even at higher doses as was

observed in K562, whereas under identical conditions Bak was

significantly upregulated (Fig. 3A). These results suggested that

possibly Bax was upregulated p53 dependently, while Bak

upregulation was p53 independent. To scrutinized the p53

dependency of Bax, we specifically inhibited p53 expression with

pifithrin-a in HCT116p53+/+ cells. A significant reduction of Bax

expression was observed, while Pifithrin-a failed to affect the

enhancement of Bak expression confirming p53-dependent Bax

and p53-independent Bak activation (Fig. 3B).

Bak functionally harmonizes for Bax in p53-null cells torelease cytochrome c

To this end, we have established comparable different time scan

for transmitting the intrinsic apoptotic signal and differential

upregulation of Bax and Bak in p53 wt and null cells. These

prompted us to further define the role of Bax and Bak in WithaD-

induced mitochondrial apoptosis. Therefore, we further investi-

gated Bax and cytochrome c level in both mitochondria and

Figure 1. WithaD-induced apoptosis occurs mainly through intrinsic pathway. (A) Chemical structure of WithaD isolated from leaves ofWithania somnifera. MOLT-3 and K562 cells were treated with (0–4 mM) WithaD for 15 hr and subsequently evaluated for the (B) expression of pro-survival (Bcl-xl, Bcl-2) and pro-apoptotic (Bak, Bax) Bcl-2 family members; (C) proteolytic processing of pro-caspase-9, -3 and -7 and the cleavage ofPARP and (D) activation of caspase-8 and reduction of total Bid. In each experiment, b-actin served as the loading control. (E) For blocking assay,MOLT-3 and K562 cells were pre-incubated with or without IETD-FMK (20 mM), LEHD-FMK (20 mM) and DEVD-CHO (100 mM) for 1 hr followed byincubation for an additional 48 hr in presence of WithaD (2 mM) and % of apoptotic cells (annexinV+/PI+) were measured by flow cytometry.‘*’ indicates the difference was statistically significant (P,0.005) in WithaD+z-LEHD-FMK and WithaD treated cells.doi:10.1371/journal.pone.0034277.g001

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 3 March 2012 | Volume 7 | Issue 3 | e34277

cytosol and Bak oligomerization in mitochondria (Fig. 4A).

Release of cytochrome c in cytosol was enhanced along with

decrease in Bax level in cytosolic fraction of HCT116 p53+/+cells. In mitochondrial fraction, we observed significant accumu-

lation of Bax and reduced cytochrome c level. Interestingly, we

observed WithaD-induced dose-dependent oligomerization of Bak

in mitochondria (Fig 4A). Initially at lower dose (0.5 mM), the level

of monomeric (16) and oligomerized (26 and 36) Bak were

minimal. Consequently, generation of 16, 26 and 36oligomeric

Bak were enhanced as we have increased the dose of WithaD to 1–

2 mM. However, at higher doses (3–4 mM), significant production

of 36 oligomeric Bak and concurrently reduction of 16 and 26form was observed.

In HCT116 p532/2 cells, cytochrome c level was enhanced in

cytosolic fraction although Bax level remains unaltered. However,

in mitochondrial fraction only basal level of Bax and reduced

cytochrome c level was observed. Interestyingly, in HCTp532/2

cells a robust upregulation and generation of oligomeric Bak (36)

Figure 2. p53 is crucial in WithaD-induced apoptosis. (A) Cells were treated with WithaD (0–4 mM) for 15 hr and p53 expression was evaluatedin the lysate of MOLT-3, HCT116, U87MG and K562 cells by Immunoblot. (B) Status of p53 in HCT116 p53+/+ and stably knockdown HCT116p532/2cells. (C) WithaD (0–4 mM) treatment led to proteolytic processing of pro-caspase-9, -3 and -7 and the cleavage of PARP as determined byimmunoblot analysis after 15 hr in HCT116p53+/+ and HCT116p532/2 cells. (D) Effect of WithaD (0–4 mM) on activation of caspase-8 and reductionof total Bid after 15 hr of treatment in HCT116 p53+/+ and HCT116p532/2 cells. In each immunoblot, b-actin was served as the loading control. (E)Difference in WithaD (0–4 mM) mediated apoptosis induction in HCT116 p53+/+ and HCT116p532/2 cells for 24 hr. (F) Morphological changes asevaluated by phase contrast microscopy in HCT116 p53+/+ and HCT116p532/2 cells after 24 hr WithaD (2 and 4 mM) treatment.doi:10.1371/journal.pone.0034277.g002

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 4 March 2012 | Volume 7 | Issue 3 | e34277

Figure 3. WithaD induced p53-dependent Bax and p53-independent Bak activation. (A) Expression of p53 downstream effector moleculesincluding Bcl-xl, Bcl-2, p21, Bak and Bax were evaluated by immunoblot assay after WithaD (0–4 mM) for 15 hr in HCT116p53+/+ and HCT116p532/2cells. (B) HCT116p53+/+ cells were pre-incubated with pifithrin a (30 mM) for 1 hr followed by 15 hr WithaD (0–2 mM) treatment and the protein levelof p53, Bax and Bak were evaluated by Western blot. In each blot, b-actin served as the loading control.doi:10.1371/journal.pone.0034277.g003

Figure 4. Cytochrome c was released before mitochondrial depolarization. (A) To detect cytochrome c release, cytosolic and mitochondrialfraction of HCT116p53+/+ and HCT116p532/2 cells were separated as mentioned in materials and methods and electrophoresed on 15% SDS-PAGEand immunoblotted using anti-cytochrome c antibody. Cytosolic Bax and cytochrome c were evaluated by Western blot analysis and b-actin servedas the loading control. Mitochondrial Bak oligomerization, Bax and cytochrome c were detected by Western blot and Cox IV served as mitochondrialloading control. (B) Mean fluorescence intensity (MFI) of FL1 was evaluated in HCT116p53+/+ and HCT116p532/2 after JC1 staining. Dosedependent treatment of WithaD (0–4 mM) revealed no significant changes in MFI value at15 hr. # considered not significant difference (P = 0.125)between untreated and WithaD (3 mM) treated cells.doi:10.1371/journal.pone.0034277.g004

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 5 March 2012 | Volume 7 | Issue 3 | e34277

was observed in mitochondrial fraction which was higher when

compared to HCT p53+/+ cells. These results suggested that both

Bax and Bak were involved in cytochrome c release to activate

intrinsic pathway in p53 wt cells, while only Bak was responsible

for the same.

Cytochrome c release and mitochondrial membrane depolar-

ization are two important events in intrinsic pathway mediated

apoptosis. These two events are differentially regulated and

induction of cytochrome c release is either dependent or

independent on the detectable loss of the negative electrical

gradient across the mitochondrial membrane [22]. Hence, we

tested the possibility whether WithaD-induced cytochrome c

release was through Bax and/Bak or due to mitochondrial

membrane depolarization (Fig. 4B). We observed no detectable

change in the MFI value of green fluorescence upto 2 mM,

whereas even at 0.5 mM considerable release of cytochrome c in

cyotsol was detected in both the cells. However, mitochondrial

membrane potential loss may also be resulted due to the

generation of ROS into the mitochondrial lumen. A recent study

reports that production of ROS in mitochondria requires Bak in a

Bax-independent manner. Additionally, the activation of Bak

responsible for ROS production is dependent on the cytosolic

presence of tBid [23]. Interestingly, although WithaD activated

Bak, it was unable to produce early ROS either dose or time

dependant manner (Figure S2). Therefore, it may be suggested

that Bax and/Bak are responsible for cytochrome c release in

cytosol rather than mitochondrial membrane depolarization.

To further delineate the role of Bax and Bak in WithaD-

induced cell death, we took HCT116p53+/+ cells either wild

type (HCT116Bax+) or homozygously deleted for the bax

(HCT116Bax2) generating four sublines HCT116Bax+/Bak+,

HCT116 Bax2/Bak+, HCT116Bax+/Bak2 and HCT116Bax2/

Bak2. Results showed p53 level enhanced dose dependently in all

the cells and also Bax and Bak expression satisfy their sub-cell line

status (Fig. 5A). Most importantly, WithaD dose dependently

augmented the level of cytochrome c in cytosol in HCT116 Bax+/

Bak+ whereas totally abolished the cytochrome c release from

mitochondria in HCT116Bax2/Bak2 cells having complete loss of

Bax and Bak. Interestingly, single loss of Bax or Bak could release the

cytochrome c in HCT116 Bax2/Bak+ and HCT116 Bax+/Bak2,

only the intensity was lower than that of HCT116 Bax+/Bak+ cells.

Next, we wanted to explore the role of Bax and Bak with

WithaD-mediated mitochondrial apoptosis. Hence using all four

cell lines, we determined the cellular viability and results showed

that after 24 hr of WithaD (2 mM) treatment, HCT116Bax2/

Bak2 showed 68.12% viability, whereas HCT116Bax+/Bak+showed only 42.65%. Interestingly, in HCT116 Bax2/Bak+ or

HCT116Bax+/Bak2 viabilities were 55.02 and 59.11% respec-

tively (Fig. 5B). Similar trend was observed in 48 hr viability

(Fig. 5C) and 7-AAD staining assays (Fig. 5D). These results

suggested that WithaD elicited apoptosis through Bax/Bak

dependent way in p53-functional cells, whereas in Bak dependent

way in p53-null cells as was observed in K562.

WithaD inhibited in vitro and in vivo tumor growth innude mice model

To check the contribution of p53 in WithaD-induced cell death,

we have used a few p53wt (SiHa, HCT116, U87MG) cell lines

along with MOLT-3 and p53-null (K562, H1299) cells of different

cancer origins. Continuous exposure of different doses of WithaD

for 24 and 48 hr revealed dose-dependent growth inhibition of

p53wt cells, IC50 values being 0.75 mM, 0.9 mM, 0.75 mM and

1.0 mM for MOLT-3, HCT116, U87MG and SiHa respectively

after 48 hr, whereas in p53-null cells reduction of cell growth was

lower for K562 [19] and H1299 than that of the p53wt cells

(Figure S3). This discrimination in sensitivity of p53wt and null

cells were also reflected by the total disintegration of cell

morphology and reduced cell density. Flow cytometric study

revealed significant increase in numbers of annexinV-positive and

both annexinV-PI- positive (Figure S4) cells in all the cancer cells

indicating higher apoptosis in p53wt cells.

We have also examined the in vivo efficacy of WithaD against

K562 xenograft in athymic nude mice. Tumor growth inhibition

was most evident in mice treated with WithaD at 10 mg/kg/day,

where ,80% reduction in tumor size was observed, in contrast

with mice treated with vehicle (Fig. 6A). The average body weights

of the control and WithaD-treated mice did not differ significantly

throughout the study (data not shown). Moreover, the WithaD-

treated mice seemed healthy and did not exhibit signs of distress

such as impaired movement or posture and indigestion. The

average tumor volume in WithaD-treated mice was significantly

lower compared with control mice on every day of tumor

measurement (Fig. 6B). For example, on 6th day, the average

tumor volume in control mice (1,080 mm3) was ,5 fold higher

compared with WithaD-treated mice (200 mm3). Consistent with

tumor volume data, the average weight of the wet tumor was

significantly lower in WithaD-treated mice compared with control

mice (data not shown). To test whether WithaD-mediated

inhibition of K562 xenograft growth in vivo was associated with

reduced cell proliferation and/or increased cell death, tumor

tissues from control and WithaD-treated mice were processed for

PI positivity. Data from a representative mouse of each group were

shown in Fig. 6C. The tumor cells from the WithaD-treated mice

exhibited a significantly higher PI positivity compared with control

tumors. Collectively, these results indicated that WithaD admin-

istration caused suppression of cellular proliferation and increased

cellular death in the tumor. The histological data indicated that

the minimal toxic effects over non-specific tissues (section of lungs,

liver and spleen) of WithaD-treated nude mice. Results showed

almost no toxic patches in the histological sections of spleen and

lungs (Fig. 6D) after WithaD treatment. Only the liver was

underwent some stress condition, which was identified by less

packed density of the liver cells. Thus, WithaD administration

significantly inhibited K562 xenograft growth in female nude mice

minimally affecting the normal tissue.

Discussion

The discovery of anti-leukemic activity and a novel ceramide

signaling of WithaD encouraged us to explore its ability as a multi-

signal inducing anti-cancer agent and decipher the molecular

mechanism of this natural product. Accordingly, the major

findings of the present investigation in WithaD-induced cell death

include (a) involvement of mitochondrial pathway, (b) p53 as

critical mediator, (c) role of Bak and Bax in p53-null and wt cells

and (d) demonstration of in vitro and in vivo growth-inhibitiory

activity of WithaD fulfilling the criteria of a potent multi-faceted

anti-cancer agent.

Apoptosis can be triggered through multiple signaling pathways,

but the ultimate event by which physiological or chemotherapy

induced cell death occurred is permeabilization of mitochondrial

membrane. This ‘point-of-no-return’ in the cell death machinery is

a complicated process and regulated mainly by the anti- and pro-

apoptotic members of Bcl-2 family proteins. Hence the exploration

of the role of Bcl-2 family members after WithaD treatment

revealed significant upregulation of both Bax and Bak in MOLT-3

but Bak was upregulated only in K562, while Bcl-2 and Bcl-xl was

downregulated in both the cells. Additionally, activation of

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 6 March 2012 | Volume 7 | Issue 3 | e34277

caspases, cleavage of PARP, enhanced pro-caspase-8 proteolysis

and dose dependent decrease of total Bid clearly indicated the

activation of both intrinsic and extrinsic signaling in WithaD-

mediated apoptosis in MOLT-3. In contrast, in K562 cells

absence of early proteolytic processing of pro-caspase-8 and almost

unchanged total Bid expression indicated towards the inactivation

of death receptor signaling. Therefore, intrinsic pathway plays the

central role in WithaD-mediated apoptosis in both the cells.

Moreover, significant reduction in apoptogenic effect of WithaD

after inhibition of caspase 9 further confirmed that WithaD-

induced cell death commence mainly through mitochondrial

pathway. However, a consistent difference exists in the com-

mencement of intrinsic and caspase-8-mediated death receptor

pathways in MOLT-3 and K562 cells. K562 being p53-null cell

hinted us about the role of p53 in WithaD-induced cell death. p53

is not just a tumor suppressor protein that singly decide cells’ fate,

instead it is a central component which intricate network of signals

and molecular interactions [24]. It has the ability to activate both

the extrinsic and intrinsic apoptotic pathways. Extrinsic pathway is

activated through the induction of Fas, DR5 and PERP [25–26]

whereas in case of intrinsic pathway, p53 target Bcl-2 family

proteins at mitochondrial level thus ultimately releasing cyto-

chrome c [27].

WithaD have been shown to induce robust upregulation of p53

in MOLT-3 and two other cancer cells including HCT116 and

U87MG having functional wild type p53. To exclude the

variations in results due to different cell lines (MOLT-3 and

K562) and to evaluate the role of p53 in WithaD-induced

apoptosis, we used HCT116p53+/+ and HCT116p532/2 cells.

Here we identified similar activation of intrinsic and death

receptor signaling in HCT116p53+/+ cells as was in MOLT-3.

Bid is a unique protein, playing the crucial role of maintaining the

flow of death signal from cell surface to mitochondria. Activation

of Bid mainly depends on either truncation by activated caspase 8

or transcriptional regulation by p53 [28]. Activated Bid then

translocates to mitochondria and activates Bax and Bak to initiate

intrinsic signal leading to apoptosome formation. Hence, p53

appears to promote the convergence of intrinsic and extrinsic

pathways through Bid regulation [29]. Dose dependent decrease

of total Bid in HCT116p53+/+ cells thereby clearly suggests that

functional p53 simultaneously activate both intrinsic as well as

extrinsic pathways intimated through Bid after WithaD treatment.

Strikingly, in HCT116p532/2 cells, early processing of pro-

caspase-8 was totally absent, although activation of caspase 9, -3,

-7 and cleavage of PARP was observed. Moreover, almost

unchanged Bid expression indicated towards the inactivation of

Figure 5. Bax and Bak were critical mediator of WithaD-mediated apoptosis. (A) Expression of p53, Bax, Bak and Cytochrome c wereevaluated in the lysate of HCT116 cells proficient for Bax and Bak (Bax+/Bak+), deficient for either Bax (Bax2/Bak+) or Bak (Bax+/Bak2) or deficient forboth Bax and Bak (Bax2/Bak2) after WithaD (0–2 mM) treatment for 15 hr by Western blot analysis. Each sub-lines were treated with WithaD (0–4 mM) for (B) 24 and (C) 48 hr and viabilities were determined by MTT assay. (D) Apoptosis induction was assessed by flow cytometric detection ofeach sub-lines by 7-AAD staining. ‘*’ indicates the difference was statistically significant (P,0.005) between HCT116Bax+/Bak+ and HCT116Bax2/Bak2 cells.doi:10.1371/journal.pone.0034277.g005

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 7 March 2012 | Volume 7 | Issue 3 | e34277

death receptor signaling. In extrinsic pathway, the cell-surface

receptor Fas (CD95/Apo-1) is a key component and in turn

promotes cell death through caspase-8 [30]. However, Fas appears

to be dispensable for p53-dependent apoptosis [31]. Therefore, the

plausible explanation of the abrogation of extrinsic pathway is due

to the absence of p53 in HCT116p532/2 as well as in K562.

Furthermore, reduced sensitivity of p53-null (HCT116p532/2)

than p53wt (HCT116p53+/+) cells towards the apoptogenic effect

of WithaD suggested a crucial role of p53 in WithaD-mediated

apoptosis.

During the induction of mitochondrial apoptosis by a death

stimulus, the role of p53 is manifold and therefore considerably

difficult to follow [5]. p53 can target itself to the mitochondrial

compartment or transactivate or trans-repress specific genes

rendering its effect of mitochondrial death [4,6]. Among them,

Bak and Bax play the pivotal role of gatekeepers of mitochondrial

integrity and cytochrome c release [32]. After WithaD treatment,

significant upregulation of both Bax and Bak in HCT116 p53+/+cells but upregulated Bak only in HCT116p532/2 cells suggested

p53-independent Bak activation. Inhibition of p53 substantially

reduced Bax but not Bak in HCT116 p53+/+ cells further

confirming p53-dependent Bax and independent Bak activation.

Therefore, activation of mitochondrial pathway in p53-null cells

could be explained through the differential upregulation of Bax

and Bak which is well correlated with p53 status.

Bax and Bak both can be activated either p53 dependently or

independently. p53 can bind directly to Bak and induce a

conformational change in the N-terminus encouraging the

oligomerization thus allowing the release of cytochrome c and

other pro-apoptotic proteins [27]. Similar function of p53 with

Bax was also evident [33]. Alternatively, Degenhardt et al reported

p53 independent role of Bax and Bak in tumor suppression [34].

Bax and Bak are also reported to have necessary function in

staurosporin, UV radiation, etoposide, thapsigargin, and tunica-

mycin-induced apoptosis [31]. Here, reduced apoptosis in

HCT116Bax2Bak2 cells over HCT116Bax+Bak+ cells and

marginal effect of single loss of Bax or Bak on the cell death

clearly suggests that Bak can functionally complement for the loss

of Bax and vice versa. Therefore when both Bax and Bak were

present, WithaD-induced mitochondrial apoptosis was most

potent as was observed in MOLT-3. In agreement with differential

role of Bax and Bak, Bax translocation along with Bak

oligomerization revealed perfect coordination with cytochrome c

release in HCT116 p53+/+ cells. In striking contrast, no Bax

translocation was found in mitochondria of HCT116p532/2

cells, although release of cytochrome c did not differ from

HCT116 p53+/+ cells. A robust Bak upregulation and oligomer-

ization further indicates toward the fact that in HCT116p532/2

cells, WithaD triggered mitochondrial apoptotic pathway that

predominantly depends on Bak but not Bax. Loss of DYm only at

Figure 6. WithaD inhibit in vitro and in vivo K562 xenograft growth. (A) WithaD could prolong the survival time of nude mice injected withK562. The tumor load of treated mice (10 mg/kg body weight) was visibly lower than untreated control mice. (B) WithaD significantly reduced tumorvolume. Each point represents mean of three tumors. (C) Status of PI-positive cells in untreated and WithaD-treated tumor cells. PI-positive cells (&)were determined with respect to PI unstained cells (%). (D) Tissue section of spleen, lungs and liver of nude mice (control and WithaD treated)determined by H&E staining and observed both in 206 and 406.doi:10.1371/journal.pone.0034277.g006

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 8 March 2012 | Volume 7 | Issue 3 | e34277

higher doses clearly indicated that cytochrome c release was an

earlier event than mitochondrial DYm dissipation where Bax

and/or Bak were solely responsible for that.

In conclusion, WithaD elicited mitochondria-mediated apopto-

sis in malignant cells through a Bax/Bak dependent way in p53wt

cells, whereas Bak compensated against loss of Bax in p53-null

cells (Fig. 7). Hence, although extrinsic pathway and Bax were

crippled due to absence of crucial p53, WithaD is able to recruit

Bak which p53-independently can induce apoptosis in p53-null

cells. Therefore, this study highlights a new possibility of using

WithaD as alternative anti-cancer agent along with the existing

chemotherapeutic agent which potentially target mitochondria-

mediated apoptosis both in p53wt as well as p53-null malignant

cells.

Materials and Methods

Reagents3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

(MTT) was purchased from Sigma (St Louis, MO). The antibodies

against p53, cytochrome c, caspase-3, FITC-annexin V, propi-

dium iodide (PI), annexin V binding buffer, 7-AAD, BD

Mitoscreen kit (JC 1) were from BD Bioscience (San Diego,

USA). Antibodies against PARP, Bax, Bak, Bcl-2, Bcl-xl, Bid, p21,

caspase 7, caspase-8, caspase-9 and HRP-secondary antibodies

were from Cell Signaling Technology (USA). Cocktail protease

inhibitor, z-VAD-fmk, z-IETD-fmk z-LEHD-fmk, z-DEVD-CHO

were from Calbiochem. RPMI-1640, IMDM and fetal bovine

serum (FBS) were from Gibco/BRL, USA.

Withanolide DWithaD (M.W 470.6) was purified in high yields from the leaves

of a well known medicinal plant Withania somnifera as described

previously [35]. The pure compound was crystallized and

analyzed by IR, mass, 1HNMR and 13C-NMR spectral analysis.

The chemical structure of WithaD has been characterized as C28

steroidal lactone, namely C4b-hydroxyC5b,C6b-epoxy-1-oxo-

,C20b,dihydroxy-20S,22R-witha-2,24-dienolide (Fig. 1A). WithaD

was dissolved in absolute ethanol as 0.5 mM solution and stored at

270uC.

Cell lines and culture conditionsChronic myelogenous leukemia (K562), colorectal carcinoma

(HCT116), cervical carcinoma (SiHa), brain carcinoma (U87MG)

and lung carcinoma (H1299) cells were purchased from ATCC.

K562 cells were cultured in RPMI-1640 medium and rest of the cells

were cultured in IMDM supplemented with 10% FBS and

incubated in 5% CO2-95% air humidified atmosphere at 37uC.

HCT116p532/2 cells were kindly provided by Dr. S. Roychowdh-

ury (CSIR-IICB). HCT116Bax2/Bak2 and HCT116Bax+/Bak2

cell lines were a kind gift from Prof. G. Chinnadurai, Institute for

Molecular Virology, USA. HCT116Bax2/Bak+ cells were a

generous gift from Dr B. Vogelstein, Johns Hopkins University,

USA. All these HCT116 sub-cell lines were cultured according to the

originator [9].

Viability assay by MTTCells (16104/250 ml/well) in log phase were seeded on 96-well

tissue culture plates incubated with WithaD (0–5 mM) for 24 and

48 hr at 37uC. After incubation, MTT (0.1 mg/well) was added,

Figure 7. Probable mechanism of WithaD induced apoptosis. WithaD treatment induces p53 activation and mitochondrial apoptosis inp53wt cells in Bax and Bak dependent manner. However, in p53-deficient cells, lack of Bax function is complemented with Bak in WithaD-inducedmitochondrial apoptosis.doi:10.1371/journal.pone.0034277.g007

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 9 March 2012 | Volume 7 | Issue 3 | e34277

and further incubated for 3–4 hr. After plate centrifugation, the

resultant pellet was dissolved in DMSO. Absorbance of the

resultant formazon was measured at 550 nm using a plate reader

(Multiskan Ex, Thermo Electron Corporation).

Tumor xenograft studyFemale nude mice of 6–7 weeks, having 20–22 gm of body

weight were acclimated for 1 week in pathogen free condition. For

subcutaneous xenograft study, mice were randomized in two

groups; control and experimental, each group containing 5 mice.

Exponentially growing K562 cells were suspended in 1:1 RPMI-

matrigel (BD bioscience) and 0.2 ml suspension containing 16107

cells were injected s.c. on right flank of each mouse above the hind

limb of each mice [36]. Tumor was allowed to develop for 20–25

days and tumor volumes were recorded till it reached 100–

120 mm3. The mice were then injected i.p. with either vehicle

(10% DMSO, 0.15 M NaCl) or vehicle containing 10 mg

WithaD/Kg body weight per day for subsequent 8 days. Tumor

volume was measured in a regular basis by external caliper and

calculated as follows: L6W2/2 (mm3); where L = length,

W = width. On the 9th day, mice were sacrificed and tumor

xenografts were excised from each mouse. By collagenase type II-

DNase I treatment, the tumor cells were isolated from tumor

tissue. Control and experimental cells were then stained with PI

and analyzed by flow cytometry.

Western blot analysisCells (16106) were treated with WithaD (0–4 mM) for 15 hr and

lysate were prepared by sonication (2 watt, 3 pulse). Equal amount

of protein were electrophoresed on SDS-PAGE (10–15%) and

electro-transferred to nitrocellulose membranes. The membrane

was blocked by TBS-BSA, probed with primary antibody

overnight at 4uC, washed with TBS containing 0.1% Tween-20

and incubated with the appropriate HRP-conjugated secondary

antibody. Immunoreactive proteins were detected on X-ray films

using the enhanced chemiluminescence system (Pierce, USA). For

the detection of Bak oligomer, equal amount of mitochondrial

fraction was boiled in sample buffer (-b-Me) and run on 12% SDS-

PAGE and processed [37].

Apoptosis assayCells (16106) were treated with WithaD (0–6 mM). Phosphati-

dyl-serine externalization was analyzed by double staining the cells

with FITC-annexin V and PI (5 mg/ml) [38]. Alternatively, treated

cells were stained with 7-AAD for 30 minutes. Cells were acquired

and analyzed by CellQuest Pro software (BD FACSCalibur). For

blocking assay, cells were separately pre-treated with caspase 8, 9

and 3 inhibitors IETD-FMK (20 mM), LEHD-FMK (20 mM) and

z-DEVD-CHO (100 mM) respectively for 30 minutes at 37uCfollowed by WithaD treatment.

Sub-cellular fractionationWithaD-treated cells (26107) were harvested, washed and

mitochondrial and cytosolic fractions were separated according

to manufacturer’s instructions (Pierce protein research products,

USA). Protein content was measured by Lowry’s method.

Mitochondrial depolarization assayMitochondrial transmembrane potential (Dym) was determined

using JC1 by Mitoscreen kit. Briefly, cells (16106) were washed

with PBS; JC1 (25 mM) was added and incubated in dark for

15 min at 37uC. Subsequently, they were washed with assay buffer

and acquired immediately by flow cytometer [39].

Statement of EthicsAnimal experiments reported in the manuscript were performed

with the approval of ‘Institutional Animal Ethics Committee’ of

the National Institute of Immunology, New Delhi, India following

the guidelines of CPCSEA. No research on humans has been

carried out.

Statistical analysisAll the results were expressed as the mean 6 S.D. of data

obtained from three separate experiments. All statistical analysis

was evaluated using graph pad prism software (San Diego). Data

were analyzed by the paired t test, and P values less than 0.05 was

considered statistically significant.

Supporting Information

Figure S1 HCT116p53+/+ and HCT116p532/2 cellswere treated with WithaD (0–5 mM) for 24 and 48 hrs andviabilities were assessed using MTT.(TIF)

Figure S2 WithaD fails to induce ROS production. (A)

Effect of 1 mM WithaD in different time points (0–2.5 hr) on the

generation of intracellular ROS by H2DCFDA in HCT116 wt

cells. (B) Effect of different doses of WithaD (0–4 mM) after 1 hr on

the generation of intracellular ROS by H2DCFDA in HCT116 wt

cells.

(TIF)

Figure S3 Anti-proliferative effect of WithaD (0–5 mM)and vehicle treated control for 24 and 48 hr againstcancer cell lines determined by MTT assay.(TIF)

Figure S4 (A) WithaD (2 mM) treatment for 24 hr induced

morphological changes and detachment of cancer cells from its

substratum. (B) Flow cytometric analysis of annexin-V/PI in

cancer cells treated with WithaD (2 mM) for 24 hr. WithaD

treatment increased the percentage of annexin V+/PI2 (down

right quadrant) and annexin V+/PI+ (upper right quadrant).

(TIF)

Acknowledgments

We earnestly thank Director, Dr. C. Shaha, National Institute of

Immunology, New Delhi, India for providing us the nude mice facility.

We sincerely thank Prof. G. Chinnadurai, Institute for Molecular Virology,

USA, Dr. B. Vogelstein, Johns Hopkins University, USA and Dr. S.

Roychowdhury (CSIR-IICB) for their generous gift of cell lines.

Author Contributions

Conceived and designed the experiments: SM CM. Performed the

experiments: SM KB. Analyzed the data: SM KB CM. Contributed

reagents/materials/analysis tools: RS. Wrote the paper: SM CM. Cell

culture and preliminary screening of WithaD: AM.

References

1. Kasibhatla S, Tseng B (2003) Why Target Apoptosis in Cancer Treatment? Mol

Cancer Ther 2: 573–580.

2. Gogvadze V, Orrenius S, Zhivotovsky B (2009) Mitochondria as targets for

cancer chemotherapy. Semin Cancer Biol 19: 57–66.

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 10 March 2012 | Volume 7 | Issue 3 | e34277

3. Vaseva AV, Moll UM (2009) The mitochondrial p53 pathway. Biochim Biophys

Acta 1787: 414–20.4. Oren M (2003) Decision making by p53: life, death and cancer. Cell Death

Differ 10: 431–442.

5. Vousden KH, Prives C (2009) Blinded by the light: the growing complexity ofp53. Cell 137: 413–431.

6. Miyashita T, Harigai M, Hanada M, Reed JC (1994) Identification of a p53-dependent negative response element in the bcl-2 gene. Cancer Res 54:

3131–3135.

7. Sugars KL, Budhram-Mahadeo V, Packham G, Latchman DS (2001) Aminimal Bcl-x promoter is activated by Brn-3a and repressed by p53. Nucleic

Acids Res 29: 4530–4540.8. Hoffman WH, Biade S, Zilfou JT, Chen J, Murphy M (2002) Transcriptional

repression of the anti apoptotic survivin gene by wild type p53. J Biol Chem 277:3247–3257.

9. Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B (2000) Role of BAX in the

apoptotic response to anticancer agents. Science 290: 989–992.10. Pietsch EC, Perchiniak E, Canutescu AA, Wang G, Dunbrack RL, et al. (2008)

Oligomerization of BAK by p53 utilizes conserved residues of the p53 DNAbinding domain. J Biol Chem 283: 21294–21304.

11. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, et al. (2000) Noxa, a BH3-

only member of the Bcl-2 family and candidate mediator of p53-inducedapoptosis. Science 288: 1053–1058.

12. Billen LP, Shamas-Din A, Andrews DW (2008) Bid: a Bax-like BH3 protein.Oncogene 27 Suppl 1: S93–104.

13. Madesh M, Antonsson B, Srinivasula SM, Alnemri ES, Hajnoczky G (2002)Rapid kinetics of tBid-induced cytochrome c and Smac/DIABLO release and

mitochondrial depolarization. J Biol Chem 277: 5651–9.

14. Mondal S, Bandyopadhyay S, Ghosh MK, Mukhopadhyay S, Roy S, et al.(2012) Natural Products: Promising Resources For Cancer Drug Discovery.

Anticancer Agents Med Chem 12: 49–75.15. Ziauddin M, Phansalkar N, Patki P, Diwanay S, Patwardhan B (1996) Studies

on the immunomodulatory effects of Ashwagandha. J Ethnopharmacol 50:

69–76.16. Agarwal R, Diwanay S, Patki P, Patwardhan B (1999) Studies on immuno-

modulatory activity of Withania somnifera (Ashwagandha) extracts in experi-mental immune inflammation. J Ethnopharmacol 67: 27–35.

17. Rasool M, Varalakshmi P (2007) Protective effect of Withania somnifera rootpowder in relation to lipid peroxidation, antioxidant status, glycoproteins and

bone collagen on adjuvant-induced arthritis in rats. Fundam Clin Pharmacol 21:

157–164.18. Jayaprakasam B, Zhang Y, Seeram NP, Nair MG (2003) Growth inhibition of

human tumor cell lines by withanolides from Withania somnifera leaves. Life Sci74: 125–132.

19. Iuvone T, Esposito G, Capasso F, Izzo AA (2003) Induction of nitric oxide

synthase expression by Withania somnifera in macrophages. Life Sci 72:1617–1625.

20. Mondal S, Mandal C, Sangwan R, Chandra S, Mandal C (2010) Withanolide Dinduces apoptosis in leukemia by targeting the activation of neutral

sphingomyelinase-ceramide cascade mediated by synergistic activation of c-JunN-terminal kinase and p38 mitogen-activated protein kinase. Mol Can 9: 239.

21. Tokino T, Nakamura Y (2000) The role of p53-target genes in human cancer.

Crit Rev Oncol Hematol 33: 1–6.

22. Kluck RM, Wetzel EB, Green DR, Newmeyer DD (1997) The release of

cytochrome c from mitochondria: a primary site for Bcl-2 regulation of

apoptosis. Science 275: 1132–6.

23. Madesh M, Zong WX, Hawkins BJ, Ramasamy S, Venkatachalam T, et al.

(2009) Execution of superoxide-induced cell death by the proapoptotic Bcl-2-

related proteins Bid and Bak. Mol Cell Biol 29: 3099–112.

24. Vogelstein B, Lane D, Levine AJ (2000) Surfing the p53 network. Nature 408:

307–310.

25. Muller M, Wilder S, Bannasch D, Israeli D, Lehlbach K, et al. (1998) p53

activates the CD95 (APO-1/Fas) gene in response to DNA damage by

anticancer drugs. J Exp Med 188: 2033–2045.

26. Wu GS, Burns TF, McDonald ER, 3rd, Jiang W, Meng R, et al. (1997)

KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene.

Nat Genet 17: 141–143.

27. Leu JI, Dumont P, Hafey M, Murphy ME, George DL (2004) Mitochondrial

p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol 6:

443–50.

28. Li H, Zhu H, Xu CJ, Yuan J (1998) Cleavage of BID by caspase 8 mediates the

mitochondrial damage in the Fas pathway of apoptosis. Cell 94: 491–501.

29. Haupt S, Berger M, Goldberg Z, Haupt Y (2003) Apoptosis – the p53 network.

J Cell Sci 116: 4077–85.

30. Nagata S, Golstein P (1995) The Fas death factor. Science 267: 1449–1456.

31. O’Connor L, Harris AW, Strasser A (2000) CD95 (Fas/APO-1) and p53 signal

apoptosis independently in diverse cell types. Cancer Res 60: 1217–1220.

32. Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, et al. (2001)

Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction

and death. Science 292: 727–30.

33. Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD, et al.

(2004) Direct activation of Bax by p53 mediates mitochondrial membrane

permeabilization and apoptosis. Science 303: 1010–4.

34. Degenhardt K, Chen G, Lindsten T, White E (2002) BAX and BAK mediate

p53-independent suppression of tumorigenesis. Cancer Cell 2: 193–203.

35. Chaurasiya ND, Uniyal GC, Lal P, Misra L, Sangwan NS, et al. (2008) Analysis

of withanolides in root and leaf of Withania somnifera by HPLC with

photodiode array and evaporative light scattering detection. Phytochem Anal

19: 148–54.

36. Bhattacharya K, Samanta SK, Tripathi R, Mallick A, Chandra S, et al. (2010)

Apoptotic effects of mahanine on human leukemic cells are mediated through

crosstalk between Apo-1/Fas signaling and the Bid protein and via

mitochondrial pathways. Biochem Pharmacol 79: 361–72.

37. Dewson G, Kratina T, Sim HW, Puthalakath H, Adams JM, et al. (2008) To

trigger apoptosis, Bak exposes its BH3 domain and homodimerizes via

BH3:groove interactions. Mol Cell 30: 369–80.

38. Mandal C, Tringali C, Mondal S, Anastasia L, Chandra S, et al. (2010) Down

regulation of membrane-bound Neu3 constitutes a new potential marker for

childhood acute lymphoblastic leukemia and induces apoptosis suppression of

neoplastic cells. Int J Cancer 126: 337–49.

39. Mandal C, Dutta A, Mallick A, Chandra S, Misra L, et al. (2008) Withaferin A

induces apoptosis by activating p38 mitogen-activated protein kinase signaling

cascade in leukemic cells of lymphoid and myeloid origin through mitochondrial

death cascade. Apoptosis 13: 1450–64.

WithaD Induced Bax and/or Bak Dependent Apoptosis

PLoS ONE | www.plosone.org 11 March 2012 | Volume 7 | Issue 3 | e34277