Methyl methanesulfonate induces apoptosis in p53-deficient H1299 and Hep3B cells through a caspase 2- and mitochondria-associated pathway

https://doi.org/10.1016/j.etap.2012.09.019Get rights and content

Abstract

Methyl methanesulfonate (MMS) has been shown to induce apoptosis in various cell types through p53-dependent pathways. Nevertheless, pharmacological and genetic blockade of p53 functions results in similar or delayed sensitivity to MMS treatment, suggesting the presence of p53-independent apoptotic mechanisms. To understand the p53-independent mechanisms that are engaged during MMS-induced apoptosis, we established MMS-induced apoptotic cell models using p53-deficient H1299 and Hep3B cells. Our results demonstrated that MMS at concentrations of 50, 100, 200, 400 and 800 μM induced the formation of gammaH2AX foci, and that at higher concentrations, 400 and 800 μM, MMS treatment led to apoptosis in the two cell lines. This apoptotic cell death was concurrent with the loss of mitochondrial membrane potential, nuclear-cytosolic translocation of active caspase 2, release of cytochrome c from mitochondria, and the cleavage of caspase 9, caspase 3 and PARP. However, MMS-induced DNA damage failed to stabilize the p53 family members TAp73 and DNp73. These results demonstrated a p53- and p73-independent mechanism for MMS-induced apoptosis that involves the nuclear-cytosolic translocation of active caspase 2 as well as the mitochondria-mediated pathway.

Highlights

MMS treatment induced apoptosis in p53-deficient H1299 and Hep3B cells. ► MMS-induced apoptosis involves the nuclear-cytosolic translocation of cleaved caspase 2. ► MMS-induced apoptosis takes mitochondria-dependent pathways.

Introduction

Methyl methanesulfonate (MMS) is a typical methylating agent which can be used as an experimental research chemical and as a solvent catalyst in polymerization, alkylation, and esterification reactions (Wyatt and Pittman, 2006). MMS has also been tested as a cancer chemotherapeutic agent, and the monoester of methanesulfonic acid can be used as a human male contraceptive and an insect attractant and repellent (Kunz et al., 2010). MMS is a known genotoxic compound that can directly react with guanine and adenine bases of DNA to generate interstrand and intrastrand cross-links (Hosseinimehr et al., 2010). In dividing cells, a replication fork can be stalled at the sites of DNA cross-links, resulting in the formation of DNA double strand breaks (DSBs), which are regarded as one of the most detrimental forms of DNA damage (Yu et al., 2006). By disrupting the dynamic and structural properties of DNA, DSBs affect many aspects of DNA metabolism, including DNA replication, transcription, recombination and repair (Shanbhag et al., 2010). DSBs can also activate several signal transduction pathways that can eventually lead to cell tumorigenesis or, depending on the situation, to apoptosis (Suwaki et al., 2011). This DSB-induced apoptosis is a key event for many conventional chemotherapeutic agent applications (Waxman and Schwartz, 2003).

p53 plays a pivotal role in DSB-induced apoptosis, which is central to its function as a tumor suppressor (Chen et al., 2011). In response to DSB formation, p53 is rapidly phosphorylated by the ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3 related (ATR) protein kinases, resulting in its activation (Shiloh, 2006). Upon activation, p53 can activate a series of pro-apoptotic proteins such as BCL2-associated X protein (BAX), p53 upregulated modulator of apoptosis (PUMA), p53-induced protein with a death domain (PIDD), and FAS receptor (Berube et al., 2005, Meulmeester and Jochemsen, 2008, Yee et al., 2009). However, the high frequency of p53 mutations in human cancer cells makes the exploration of p53-independent cell signaling pathways become increasingly urgent (Kashiwazaki et al., 1997). Previous studies have also demonstrated important roles for p53 in MMS-induced apoptosis (Tian et al., 2009, Ziv et al., 2006). Despite these findings, it is recognized that p53 may only be partially responsible for MMS-induced cytotoxicity. For instance, after treatment with MMS, the p53 mutant human lymphoblastoid cells (WTK1) exhibited a delayed apoptosis, as compared to the p53 proficient human lymphoblastoid cells (TK6) (Greenwood et al., 1998). In another case, both p53 knockout fibroblasts and the corresponding wild-type cells displayed a similar sensitivity to MMS-induced chromosomal aberrations and apoptosis (Lackinger et al., 2001). Thus, the apoptotic response to MMS-induced DNA damage involves both p53-dependent and -independent mechanisms. However, very little is known about the p53-independent mechanisms.

p73, as a p53 family member, is also responsive to the DNA damage that leads to apoptosis (Roos and Kaina, 2006). In contrast to p53, p73 is rarely mutated in human cancers, thus, p73 as a new candidate tumor suppressor is getting more and more interest (Ozaki and Nakagawara, 2005). The p73 gene encodes two isoforms that differ in their N-termini, and which can be functionally classified as trans-activation competent (TAp73) and dominant negative (DNp73) proteins (Moll and Slade, 2004). The TAp73 shares similarities with p53 with regards to DNA damage-induced apoptosis through its activation of p53 responsive genes (Nieto-Rementeria et al., 2009). The DNp73 has a very important regulatory role, as it exerts a dominant-negative effect on p53 and TAp73 by inhibiting their transactivation activity. Thus, DNp73 confers drug resistance to tumor cells harboring p53 and/or TAp73 (Stiewe et al., 2002). Its inhibitory function is exerted either at the oligomerization level, or by competing for binding to the p53/TAp73 DNA target sequence (Vossio et al., 2002). In addition, the DNp73 promoter contains an efficient p53/TAp73 responsive element that can be transactivated by p53 and/or TAp73, and therefore creates a dominant-negative feedback loop that regulates the function of p53 and/or TAp73 (Concin et al., 2004). Several lines of evidences indicate that the presence and participation of different spliced forms of p73 might be one reason for the different cell fates observed in response to DNA damage (Sayan et al., 2010). Moreover, previous studies have also demonstrated in response to different DNA-damaging stimuli, p73 isoforms can contribute to p53-dependent or -independent cell apoptotic signaling pathways (Costanzo et al., 2002, Murray-Zmijewski et al., 2006). However, it is still unknown whether TAp73 and/or DNp73 participate in MMS-induced apoptosis.

Caspase 2 is the only pro-caspase constitutively present in the nucleus, and it is unique among the caspases in that it has features of both upstream and downstream caspases (van Loo et al., 2002). In recent years, accumulating evidence indicates that in response to DNA damage, caspase 2 can act as an initiator regulatory enzyme upstream of the mitochondria-dependent apoptosis pathway (Vakifahmetoglu et al., 2008). For example, treatment of cells with 2,3-dimethoxy-1,4-naphthoquinone (DMNQ), a well-known oxidative agent that induces DNA damage, resulted in the release of cytochrome c from mitochondria, and this activity can be inhibited by the caspase 2 selective inhibitor (zVD-VAD-fmk) (Tamm et al., 2008). In addition, during cisplatin-induced apoptosis, using siRNA to inhibit caspase 2 expression caused decreased cytochrome c release, and reduced caspase 3 and caspase 9 activities (Vakifahmetoglu et al., 2008). Moreover, several observations suggested in some cell lines, caspase 2 is required for p53-mediated apoptosis induced by genotoxic agents (Ren et al., 2005). But the link between p73 isoforms and caspase 2 activation remains to be elusive. Despite the above evidence supporting roles for caspase 2 in stress-induced apoptosis, there is currently no model that links a given type of DNA damage with nuclear caspase 2 activation and mitochondria-dependent apoptosis. Furthermore, most reports have only investigated either the role of caspase 2 on the activation of down-stream genes, or the effect of genotoxic agents on the expression of caspase 2 in whole-cell lysates, rather than in specific sub-cellular compartments (Guo et al., 2002, Sidi et al., 2008).

In this current study, we sought to analyze the p53-independent mechanisms induced during MMS-induced apoptosis. Using the p53-deficient H1299 and Hep3B cells, we first established an MMS-induced apoptotic model. Then, we carried out a set of analyses assessing cell viability, percentages of apoptotic cells, extent of DNA damage, and mitochondrial membrane potentials (MMP). Finally, we analyzed the protein levels of TAp73, DNp73, pro-caspase 2, cleaved caspase 2, pro-caspase 9, cleaved caspase 9, pro-caspase 3, cleaved caspase 3, pro-PARP, cleaved PARP, and cytochrome c. Our results, as detailed below, suggest that there exists a caspase 2-associated, but p53- and p73-independent, apoptotic response to MMS-induced DNA damage, which is mediated through the mitochondrial pathway.

Section snippets

Materials

RPMI 1640, Dulbecco's Modified Eagle's Medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco Invitrogen Corp. (Gibco Laboratories, Grand Island, NY, USA). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), methyl methanesulfonate (MMS), 4,6-diamidino-2-phenylindole (DAPI) and rhodamine123 were purchased from Sigma–Aldrich (St. Louis, MO, USA). The nucleus/nucleus-free cytosol protein isolation kit was supplied by KeyGEN (Nanjing, China). TAp73, DNp73, caspase

Effect of MMS on cell viability

The cytotoxic effects of different concentrations (0, 50, 100, 200, 400 and 800 μM) of MMS on H1299 and Hep3B cells were assessed using the MTT reduction assay at 2, 4, 8, 12 and 24 h post-treatment. As shown in Fig. 1, viabilities of H1299 (Fig. 1A) and Hep3B (Fig. 1B) cells were both significantly reduced by 400 and 800 μM MMS at 12 h, and by 200, 400 and 800 μM MMS at 24 h (P < 0.01). However, there was no significant change in the viability at 2, 4 and 8 h for all MMS-treated groups in the two cell

Discussion

MMS is an alkylating agent used in cancer therapy, and has been reported to induce apoptosis through the activation of p53-dependent and/or -independent pathways (Lackinger et al., 2001, Ryu et al., 2001). In agreement with these studies, we found that MMS at concentrations of 400 and 800 μM significantly reduced the viability of the p53-deficient H1299 and Hep3B cells 12 h post-treatment. This result was further confirmed by flow cytometry, which demonstrated that MMS at concentrations of 400

Conflict of interest

The authors declare that they have no conflict of interest.

Acknowledgements

This work was supported by grants from National Natural Science Foundation of China (No. 81172692); Zhejiang Provincial Natural Science Foundation (R2100555); Ministry of Science and Technology, China (2009DFB30390); and Post Doctor Science Foundation of China (No. 2011M501356). J. Yang is a recipient of the Zhejiang Provincial Program for the Cultivation of High-level Innovative Health Talents.

References (52)

  • T. Stiewe et al.

    Transactivation-deficient Delta TA-p73 inhibits p53 by direct competition for DNA binding: implications for tumorigenesis

    J. Biol. Chem.

    (2002)
  • N. Suwaki et al.

    RAD51 paralogs: roles in DNA damage signalling, recombinational repair and tumorigenesis

    Semin. Cell Dev. Biol.

    (2011)
  • A. Takahashi et al.

    Does gammaH2AX foci formation depend on the presence of DNA double strand breaks?

    Cancer Lett.

    (2005)
  • Y. Yu et al.

    A comparative study of using comet assay and gammaH2AX foci formation in the detection of N-methyl-N′-nitro-N-nitrosoguanidine-induced DNA damage

    Toxicol In Vitro

    (2006)
  • C. Berube et al.

    Apoptosis caused by p53-induced protein with death domain (PIDD) depends on the death adapter protein RAIDD

    Proc. Natl. Acad. Sci. U.S.A.

    (2005)
  • X. Chen et al.

    G-protein-coupled receptor kinase 5 phosphorylates p53 and inhibits DNA damage-induced apoptosis

    J. Biol. Chem.

    (2011)
  • N. Concin et al.

    Transdominant deltaTAp73 isoforms are frequently up-regulated in ovarian cancer, Evidence for their role as epigenetic p53 inhibitors in vivo

    Cancer Res.

    (2004)
  • S. Cuenin et al.

    p53-Induced protein with a death domain (PIDD) isoforms differentially activate nuclear factor-kappaB and caspase-2 in response to genotoxic stress

    Oncogene

    (2008)
  • S. Emmrich et al.

    Antisense gapmers selectively suppress individual oncogenic p73 splice isoforms and inhibit tumor growth in vivo

    Mol. Cancer

    (2009)
  • Y.H. Han et al.

    Arsenic trioxide inhibits growth of As4.1 juxtaglomerular cells via cell cycle arrest and caspase-independent apoptosis

    Am. J. Physiol. Renal Physiol.

    (2007)
  • T. Helleday et al.

    DNA repair pathways as targets for cancer therapy

    Nat. Rev. Cancer

    (2008)
  • S.J. Hosseinimehr et al.

    Protective effect of hawthorn extract against genotoxicity induced by methyl methanesulfonate in human lymphocytes

    Toxicol. Ind. Health

    (2010)
  • P.A. Jeggo et al.

    DNA double-strand breaks: their cellular and clinical impact?

    Oncogene

    (2007)
  • Y. Jiang et al.

    Benzo(a)pyrene induces p73 mRNA expression and necrosis in human lung adenocarcinoma H1299 cells

    Environ. Toxicol.

    (2012)
  • H. Kashiwazaki et al.

    High frequency of p53 mutations in human oral epithelial dysplasia and primary squamous cell carcinoma detected by yeast functional assay

    Oncogene

    (1997)
  • A. Kumari et al.

    p53 protects from replication-associated DNA double-strand breaks in mammalian cells

    Oncogene

    (2004)
  • View full text