Bcl-2 blocks 2-methoxyestradiol induced leukemia cell apoptosis by a p27Kip1-dependent G1/S cell cycle arrest in conjunction with NF-κB activation

https://doi.org/10.1016/j.bcp.2009.03.017Get rights and content

Abstract

2-Methoxyestradiol (2-ME2) induces leukemia cells to undergo apoptosis in association with Bcl-2 inactivation but the mechanisms whereby Bcl-2 contributes to protection against programmed cell death in this context remain unclear. Here we showed that 2-ME2 inhibited the proliferation of Jurkat leukemia cells by markedly suppressing the levels of cyclins D3 and E, E2F1 and p21Cip1/Waf1 and up-regulating p16INK4A. Further, 2-ME2 induced apoptosis of Jurkat cells in association with down-regulation and phosphorylation of Bcl-2 (as mediated by JNK), up-regulation of Bak, activation of caspases-9 and -3 and PARP-1 cleavage. To determine the importance and mechanistic role of Bcl-2 in this process, we enforced its expression in Jurkat cells by retroviral transduction. Enforcing Bcl-2 expression in Jurkat cells abolished 2-ME2-induced apoptosis and instead produced a G1/S phase cell cycle arrest in association with markedly increased levels of p27Kip1. Bcl-2 and p27Kip1 were localized mainly in the nucleus in these apoptotic resistant cells. Interestingly, NF-κB activity and p50 levels were increased by 2-ME2 and suppression of NF-κB signaling reduced p27Kip1 expression and sensitized cells to 2-ME2-induced apoptosis. Importantly, knocking-down p27Kip1 in Jurkat Bcl-2 cells sensitized them to spontaneous and 2-ME2-induced apoptosis. Thus, Bcl-2 prevented the 2-ME2-induced apoptotic response by orchestrating a p27Kip1-dependent G1/S phase arrest in conjunction with activating NF-κB. Thus, we achieved a much better understanding of the penetrance and mechanistic complexity of Bcl-2 dependent anti-apoptotic pathways in cancer cells and why Bcl-2 inactivation is so critical for the efficacy of apoptosis and anti-proliferative inducing drugs like 2-ME2.

Graphical abstract

2-ME2-activates JNK mediating phosphorylation and inactivation of Bcl-2 and induction of apoptosis. Overexpression of Bcl-2 enhances NF-κB activity and up-regulates p27kip1 leading to G1/S arrest and inhibition of apoptosis. Supppression of NF-κb or p27Kip1 sensitizes Bcl-2-espressing cells to apoptosis.

  1. Download : Download full-size image

Introduction

2-Methoxyestradiol (2-ME2), a catechol estrogen, is a natural metabolic by-product of 17β-estradiol that acts independently of estrogen receptors to inhibit angiogenesis and tumor cell proliferation and to induce apoptosis in vitro and in vivo[1], [2], [3], [4], [5], [6], [7], [8].

Multiple discrete mechanisms are involved in the specific anti-proliferative action of 2-ME2 in tumor cells including both G1/S and G2/M cell cycle phase arrest. 2-ME2 was shown to arrest the growth of many human cancer cell lines in vitro, including, Jurkat cells [9], multiple myeloma [10], epithelial [11], [12], [13], [14], [15], [16], melanoma [17] and medulloblastoma cancer cells [18] and transformed fibroblasts [19] in G2/M phase. G2/M cell cycle arrest was characterized by the induction of cyclin B and Cdc2 kinase activity [9], [11], [13], [17]. Others, however, showed that 2-ME2 inhibited the growth of pancreatic cancer cells by prolonging S-phase [20] or by inducing both G1/S and G2/M arrest of human osteosarcoma cells [21] or of pancreatic cell lines [22]. In contrast, 2-ME2 had no effect on the growth of normal cells [13], [15], [17], [19], [23] including lymphocytes [24]. The induction of apoptosis by 2-ME2 in tumor cells involves different molecular mechanisms. While several studies suggested that 2-ME2 can induce apoptosis both by p53-dependent and p53-independent mechanisms in various tumor cell types [8], [13], [15], [17], [18], [19], [23], [25], [26], scant evidence exists implicating NF-κB in 2-ME2-induced apoptosis [18], [27]. While p38/JNK-dependent NF-κB activation was required for 2-ME2-induced apoptosis in prostate cancer cells [27], in contrast a reduction in NF-κB transcriptional and DNA binding activity was observed in 2-ME2-induced apoptosis of medulloblastoma cells [18].

Further studies have implicated the anti-apoptotic members of the Bcl-2 family in 2-ME2-induced apoptosis [15], [27], [28], [29], [30]. The Bcl-2 family comprises two mutually opposing groups of proteins including: anti-apoptotic Bcl-2 and Bcl-XL and pro-apoptotic Bak and Bax. While several models have been proposed to explain the mechanism by which Bcl-2 family members regulate apoptosis, the ratio of anti-apoptotic:pro-apoptotic Bcl-2 family members is one key factor dictating the relative sensitivity or resistance of cells to a wide variety of apoptotic stimuli [31], [32], [33]. In addition, Bcl-2 and Bcl-XL are regulated by phosphorylation in their flexible loop between the BH4 and BH3 domains, which determines their cytoprotective function in response to cellular stresses as well as growth and survival factors [34]. Bcl-2 phosphorylation by ERK1/2 and PKCα kinases, either at the unique Ser70 residue or at multiple Thr69, Ser70, and Ser89 sites, positively regulates Bcl-2 anti-apoptotic function [35]. However, c-jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK)-mediated phosphorylation of Bcl-2 at multiple-sites hinders Bcl-2 survival function in paclitaxel-induced apoptosis [36], [37]. Thus, the type of stimulus, the regulatory pathways involved, and the degree and duration of phosphorylation at specific Bcl-2 residues produce different outcomes.

In response to 2-ME2, both Bcl-2 [15], [27], [28], [29] and Bcl-XL[22], [30] are inactivated by phosphorylation at Ser70 and Ser62, respectively, mediated by JNK but not ERK1/2 [27], [29], [30], [37]. Whether Bcl-2 phosphorylation induced by microtubule destabilizing agents such as taxol [37], [38], [39] or 2-ME2 [15], [27], [28], [29] interferes with the heterodimerization of Bcl-2 to Bax remains elusive [37], [38], [39]. However, JNK-mediated phosphorylation of Bcl-2 leading to its inactivation in response to 2-ME2 will allow the pro-apoptotic members of the Bcl-2 family, to drive the cell towards death [31], [32], [33].

2-ME2-induced phosphorylation of Bcl-2, mediated by JNK/SAPK, has been correlated with apoptosis of prostate [15], [27] and leukemia cells [29]. Activation of JNK by 2-ME2 appears to be due to its ability to potently inhibit superoxide dismutase [40] resulting in enhanced formation of ROS [7], [24], [25], [26], [40] and Akt inhibition [26] to selectively kill tumor cells [24], [25], [26], [40].

Although available data point to Bcl-2 phosphorylation as a key executing signal for 2-ME2-induced apoptosis, Bcl-2's mechanisms of action in this context and its ability to protect cells from 2-ME2-induced apoptosis both remain undefined. We show here that 2-ME2 treatment of leukemia cells promoted a p53-independent apoptotic response characterized by Bcl-2 down-regulation and phosphorylation mediated by JNK/SAPK, Bak up-regulation, proteolytic cleavages of caspases-9, -3 and PARP-1. Moreover, ectopic over-expression of Bcl-2 in leukemia cells prevented all of these aspects of the 2-ME2-induced apoptotic response by orchestrating a p27Kip1-dependent G1/S phase arrest in conjunction with activating NF-κB.

Section snippets

Cell culture

Human Jurkat T lymphoma cells (clone E6-1) were cultured in RPMI-1640 and amphotropic Phoenix cells in DMEM supplemented with 10% FCS (all from Biochrom AG, Germany), 2 mM l-glutamine, 100 units/ml penicillin and 100 μg/ml streptomycin (PAA Laboratories, Germany) at 37 °C, 5% CO2.

Retroviral vectors and generation of infected Jurkat cells

The retroviral vectors pBabe-Puro and pBabe-Puro/Bcl-2 carrying human Bcl-2 cDNA [41] pSR-Puro (referred to as Vector thereafter) and pSR-Puro carrying p27Kip1 shRNA (referred to as p27KD, thereafter) [42] have been

Induction of Jurkat cell apoptosis by 2-ME2

Actively proliferating Jurkat cells were treated with different concentrations of 2-ME2 ranging from 0.5 μM to 10 μM or ethanol as a negative control for 24 h and 48 h and low molecular weight DNA was extracted and analyzed by agarose gel electrophoresis (Fig. 1A). Whereas ethanol-treated cells did not undergo apoptosis, 2-ME2-treated Jurkat cells exhibited DNA fragmentation, producing a DNA ladder characteristic of cells undergoing apoptosis even with 2-ME2 as low as 0.5–1.0 μM. Based on the

Discussion

2-ME2 has been shown to inhibit the growth and induce apoptosis of tumor cells but not of normal cells, through several mechanisms studied in different cellular systems including phosphorylation and inactivation of Bcl-2 [7], [15], [27], [28], [29]. However, the mechanism(s) involved in 2-ME2-induced apoptosis in a particular setting and more specifically the role(s) of Bcl-2 in the effects of 2-ME2 remain unclear. In this report, the effects of 2-ME2 on human Jurkat T-leukemic cells and the

Conflict of interest

The authors have no conflicting financial interests.

Acknowledgements

We are grateful to Dr. D. Tang, University of Montreal, Canada for providing pBabe-Puro/hBcl-2, Dr. J. Chen, H. Lee Moffitt Cancer Center & Research Institute, The University of South Florida, USA, for pSR-Puro and pSR-Puro#p27Kip1, and Drs C.J. Sherr and M. Roussel, Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA, for providing p21Cip1/Waf1 and p27Kip1 retroviral vectors. We also thank Drs S. Georgatos, C. Murphy and P. Pappas, University of Ioannina

References (82)

  • D. Tang et al.

    Akt is activated in response to an apoptotic signal

    J Biol Chem

    (2001)
  • C. Wang et al.

    Activation of p27Kip1, expression by E2F1: a negative feedback mechanism

    J Biol Chem

    (2005)
  • J. Li et al.

    Novel NEMO/IκB kinase and NF-κB target genes at the pre-B to immature B cell transition

    J Biol Chem

    (2001)
  • E. Kolettas et al.

    Bcl-2 but not clusterin/apolipoprotein J protected human diploid fibroblasts and immortalised keratinocytes from ceramide-induced apoptosis: role of p53 in the ceramide response

    Arch Biochem Biophys

    (2006)
  • G.P. Dimri et al.

    Regulation of two E2F-related genes in presenescent and senescent human fibroblasts

    J Biol Chem

    (1994)
  • G.-Q. Wang et al.

    A role for mitochondrial Bak in apoptotic response to anticancer drugs

    J Biol Chem

    (2001)
  • C.A. Massaad et al.

    Inhibition of transcription factor activity by nuclear compartment-associated Bcl-2

    J Biol Chem

    (2004)
  • X. Deng et al.

    Bcl2 retards G1/S cell cycle transition by regulating intracellular ROS

    Blood

    (2003)
  • J.-H. Jang et al.

    Bcl-2 attenuation of oxidative cell death is associated with up-regulation of γ-glutamylcysteine ligase via constitutive NF-κB activation

    J Biol Chem

    (2004)
  • M.J. Crawford et al.

    Bcl-2 overexpression protects photooxidative stress-induced apoptosis of photoreceptor cells via NF-κB preservation

    Biochem Biophys Res Commun

    (2001)
  • H. Albrecht et al.

    Bcl-2 protects from oxidative damage and apoptotic cell death without interfering with activation of NF-κB by TNF

    FEBS Lett

    (1994)
  • J.L. Herrmann et al.

    Bcl-2 suppresses apoptosis resulting from disruption of the NF-κB survival pathway

    Exp Cell Res

    (1997)
  • S. Minami et al.

    Molecular cloning of the human p27kip1 gene promoter

    FEBS Lett

    (1997)
  • E. Sicinska et al.

    Requirement for cyclin D3 in lymphocyte development and T cell leukemias

    Cancer Cell

    (2003)
  • A.B. Gladden et al.

    Cell cycle progression without cyclin E-CDK2: breaking down the walls of dogma

    Cancer Cell

    (2003)
  • R.H. Weiss

    p21Waf1/Cip1 as a therapeutic target in breast and other cancers

    Cancer Cell

    (2003)
  • C. Borner

    Diminished cell proliferation associated with the death protective activity of bcl-2

    J Biol Chem

    (1996)
  • V.L. Johnson et al.

    Effects of differential overexpression of Bcl-2 on apoptosis, proliferation and telomerase activity in Jurkat cells

    Exp Cell Res

    (1999)
  • R.C. Prasad et al.

    Identification of genes, including the gene encoding p27Kip1, regulated by serine 276 phosphorylation of the p65 subunit of NF-κB

    Cancer Lett

    (2009)
  • T. Fotsis et al.

    The endogenous estrogen metabolite 2-methoxyoestradiol inhibits angiogenesis and suppresses tumour growth

    Nature

    (1994)
  • R.J. D’Amato et al.

    2-methoxyestradiol, an endogenous mammalian metabolite, inhibits tubulin polymerization by interacting at the colchicine site

    Proc Natl Acad Sci USA

    (1994)
  • N. Klauber et al.

    Inhibition of angiogenesis and breast cancer in mice by microtubule inhibitors 2-methoxyestradiol and taxol

    Cancer Res

    (1997)
  • V.S. Pribluda et al.

    2-methoxyestradiol: an endogenous antiangiogenic and antiproliferative drug candidate

    Cancer Metastasis Rev

    (2000)
  • N.J. Lakhani et al.

    2-methoxyestradiol, a promising anticancer agent

    Pharmacotherapy

    (2003)
  • S.L. Mooberry

    New insights into 2-methoxyestradiol, a promising antiangiogenic and antitumor agent

    Curr Opin Oncol

    (2003)
  • D. Dingli et al.

    Promising preclinical activity of 2-methoxyestradiol in multiple myeloma

    Clin Cancer Res

    (2002)
  • M.N. Zoubine et al.

    2-methoxyestradiol-induced growth suppression and lethality in estrogen responsive MCF-7 cells may be mediated by downregulation of p34cdc2 and cyclin B1 expression

    Int J Oncol

    (1999)
  • T.M. La Vallee et al.

    2-methoxyestradiol inhibits proliferation and induces apoptosis independently of estrogen receptors α and β

    Cancer Res

    (2002)
  • A.P. Kumar et al.

    2-methoxyestradiol blocks cell cycle progression at G2/M phase and inhibits growth of human prostate cancer cells

    Mol Carcinogen

    (2001)
  • H.L. Lin et al.

    2-methoxyestradiol-induced caspase-3 activation and apoptosis occurs through G2/M arrest dependent and independent pathways in gastric carcinoma cells

    Cancer

    (2001)
  • R. Ghosh et al.

    Cell cycle block and apoptosis induction in a human melanoma cell line following treatment with 2-methoxyestradiol: therapeutic implications

    Melanoma Res

    (2004)
  • Cited by (0)

    View full text