Elsevier

Biochemical Pharmacology

Volume 66, Issue 8, 15 October 2003, Pages 1537-1545
Biochemical Pharmacology

The histone deacetylase inhibitor suberic bishydroxamate: a potential sensitizer of melanoma to TNF-related apoptosis-inducing ligand (TRAIL) induced apoptosis

https://doi.org/10.1016/S0006-2952(03)00509-4Get rights and content

Abstract

TRAIL appears to be a promising anticancer agent in that it induces apoptosis in a wide range of cancer cells but not normal tissues. Sensitivity of melanoma cells to TRAIL-induced apoptosis varied considerably because of their development of various resistance mechanisms against apoptosis. We discuss in this report the potential effect of a histone deacetylase inhibitor SBHA on TRAIL-induced apoptosis. Histone deacetylase (HDAC) inhibitors regulate histone acetylation and thereby modulate the transcriptional activity of certain genes leading to cell growth arrest, cellular differentiation, and apoptosis. Suberic bishydroxamate (SBHA) is a relatively new HDAC inhibitor that induced apoptosis in the majority of melanoma cell lines through a mitochondrial and caspase-dependent pathway. This was due to its regulation of the expression of multiple proteins that are involved in either the mitochondrial apoptotic pathway (Bcl-2 family members) or the final phase of apoptosis (caspase-3 and XIAP). Co-treatment with SBHA at nontoxic doses and TRAIL resulted in a marked increase in TRAIL-induced apoptosis of melanoma, but showed no toxicity to melanocytes. SBHA appeared to sensitize melanoma to TRAIL-induced apoptosis by up-regulation of pro-apoptotic proteins in the TRAIL-induced apoptotic pathway such as caspase-8, caspase-3, Bid, Bak, and Bax, and up-regulation of the BH3 domain only protein, Bim. This, together with activated Bid, may have acted synergistically to cause changes in mitochondria. Treatment with SBHA also resulted in down-regulation of antiapoptotic members of the Bcl-2 family, Bcl-XL and Mcl-1, and the IAP member, XIAP. These changes would further facilitate apoptotic signaling. SBHA appeared therefore to be a potent agent in overcoming resistance of melanoma to TRAIL-induced apoptosis.

Introduction

Histones are basic proteins that, by complexing with DNA, form nucleosomes leading to the compact structure of chromatin. Basic amino acids of the histones can be post-translationally modified with methyl, acetyl or phosphate groups. The balance between histone acetylation and deacetylation is regulated by histone acetyltransferase (HAT) and HDACs, and plays an important role in transcriptional regulation of genes [1], [2], [3], [4], [5]. Acetylation of lysine residues of histones results in more open chromatin structure and therefore activation of transcription, whereas hypoacetylation of histones is associated with a condensed chromatin structure resulting in the repression of gene transcription [4], [5]. Deregulation of HAT and HDACs has been implicated in the formation and development of certain human cancers by changing the expression pattern of various genes [6], [7], [8], [9].

HDACs are a family of enzymes that regulate histone acetylation by catalyzing the removal of acetyl groups on lysine residues of the nucleosomal histones [4], [5]. HDAC inhibitors are members of a class of agents that modulate the expression of genes by inhibiting the HDAC activity, resulting in an increase in histone acetylation [4], [5]. DNA microarray studies using malignant cell lines cultured in the presence of a HDAC inhibitor showed that only a small number (1–2%) of genes were regulated [10]. The altered gene expression after exposure to a HDAC inhibitor has been demonstrated to arrest cell growth [11], [12], [13] and to reverse neoplastic characteristics by inducing differentiation [14], [15], [16], [17]. In addition, HDAC inhibitors can induce apoptotic cell death in a variety of tumor cell lines [14], [18], [19], [20]. HDAC inhibitors are therefore considered to be promising chemotherapeutic agents for treatment of malignant tumors. The potential significance of HDAC inhibitors as anticancer agents has been supported by studies in animal models and clinical trials showing antitumor activity with minor toxicity to normal tissues in vivo[21], [22].

Although a number of HDAC inhibitors have been shown to induce apoptosis of cultured tumor cells, the mechanism(s) underlying this appear to vary. For example, explanations for the induction of apoptosis by the nonspecific HDAC inhibitor sodium butyrate include alterations in Bcl-2 family protein expression [23], [24], [25], [26], increased caspase activity [20], increased sensitivity to Fas–Fas ligand interaction [27], [28] and changes in the expression of genes such as c-myc and k-ras[29]. Moreover, caspase-independent mechanisms have also been suggested as causative [25]. It appeared that sodium butyrate-induced apoptosis was mediated through the mitochondrial pathway in that apoptosis was inhabitable by overexpression of Bcl-XL[25]. Studies with another HDAC inhibitor, apicidin, showed that this was associated with translocation of Bax to mitochondria and subsequent release of cytochrome c[30]. The HDAC inhibitor suberoylanilide (SAHA) appeared to induce apoptosis by direct noncaspase-dependent activation of Bid [31].

Much interest has been given to the role of HDAC inhibitors in regulation of p21WAF1/CIP1[19], [32], [33], which is one of the key regulators of the cell cycle. The transcription of p21WAF1/CIP1 is up-regulated in response to HDAC inhibitors as a consequence of increased acetylation of the chromatin at the Sp1 binding sites in the promoter region of p21WAF1/CIP1[33], [34]. Up-regulation of p21WAF1/CIP1 by HDAC inhibitors plays a critical role in cell cycle arrest by inhibiting cyclin-dependent kinase (cdk) activity [13], [32]. It may also participate in the induction of apoptosis in that overexpression of p21WAF1/CIP1 leads to induction of cytochrome c release, activation of caspases, and apoptosis [33], [34]. However, cell lines that did not have increased p21WAF1/CIP1 protein levels in response to the HDAC inhibitor acelaic bishydroxamic (ABHA) were more sensitive to apoptosis than cell lines in which p21WAF1/CIP1 protein levels were increased following treatment with ABHA [19].

TNF-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor (TNF) family that can induce apoptosis in a wide range of cultured malignant cells including melanoma [35], [36], [37], [38]. The potential significance of TRAIL as an anticancer agent has been supported by studies in animal models showing selective toxicity to human tumor xenografts but not normal tissues. Traditionally, two principal pathways to apoptosis have been recognized, the transmembrane “extrinsic” pathway and the mitochondrial “intrinsic” pathway. Both depend on activation of cysteine proteases that cleave at aspartate residues (caspases). These enzymes are synthesized as proenzymes that become activated by adaptor proteins. Initiator caspases once activated can activate so-called effector caspases, which act on a wide range of substrates to cause apoptosis. The initiator caspases for the extrinsic pathway are caspase-8 and -10, whereas caspase-9 and -2 are initiator caspases for the intrinsic pathway. The effector caspases are believed to be similar for both pathways, i.e. 3, 6 and 7 [39], [40]. TRAIL-induced apoptotic signaling is believed to be initiated by ligand-induced aggregation of death domains that reside on the cytoplasmic sides of the death receptors, TRAIL-R1 and -R2. This in turn orchestrates the assembly of adaptor proteins such as Fas-associated death domain (FADD) that activate initiator caspases, caspase-8 and -10 leading eventually to activation of effector caspases such as caspase-3 [37], [41], [42].

TRAIL-induced apoptosis of melanoma is largely dependent on induction of changes in mitochondrial membrane permeability [43], [44]. Release of Smac/DIABLO from mitochondria to the cytosol mediates TRAIL-induced apoptosis by binding to members of the inhibitors of apoptosis protein (IAP) family. This prevents the IAP proteins binding to the effector caspase-3 and -4, and to caspase-9, so allowing apoptosis to proceed [39], [40], [43]. TRAIL induces low levels of Smac/DIABLO release into the cytosol in TRAIL resistant melanoma cell lines but high levels in TRAIL sensitive cell lines [43]. The basis for this variation in Smac/DIABLO release remains largely unknown but did not appear to be due to differences in activation of caspase-8 and Bid in that they appeared to be cleaved to the same extent in both TRAIL sensitive and resistant melanoma cell lines [43]. It was however suggested that Bcl-2 family members play a pivotal role in regulating mitochondrion-mediated apoptosis. The antiapoptotic members of the Bcl-2 family, such as Bcl-2, Bcl-XL and Mcl-1, appear to preserve the integrity of the mitochondrial outer membrane by binding to BH3 domains of the pro-apoptotic BH3-only proteins such as Bid, Bim, and Noxa [45], [46]. In the absence or deficiency of the antiapoptotic Bcl-2 family members, the pro-apoptotic BH3-only proteins cause the oligomerization of the multi-BH3 domain proteins, Bax and Bak, in the mitochondrial outer membrane and formation of pores which allow release of mitochondrial proteins such as Smac/DIABLO and cytochrome c[47]. A schematic illustration of TRAIL-induced apoptosis pathway in melanoma is shown in Fig. 1.

SBHA is a relatively new HDAC inhibitor that can induce apoptosis in the majority of melanoma cell lines through a mitochondrial and caspase-dependent pathway [48]. Several key antiapoptotic proteins are down-regulated in SBHA-treated cells [49]. These include X-linked IAP (XIAP) and the Bcl-2 family proteins, Mcl-1 and Bcl-XL. In contrast, SBHA induces up-regulation of the Bcl-2 family pro-apoptotic proteins, Bid, Bim, Bax, and Bak. Moreover, the expression levels of pro-caspase-8 and pro-caspase-3 are also up-regulated by SBHA treatment. This combination of events strongly suggests that SBHA may be a valuable agent in sensitizing melanoma to other apoptosis inducers. In this report, we discuss the possible mechanisms by which SBHA induces apoptosis and the potential effect of SBHA on TRAIL-induced apoptosis of melanoma.

Section snippets

SBHA-induced apoptosis of melanoma is caspase-dependent

Studies in a panel of melanoma cell lines have shown that SBHA induces apoptosis in the majority of the cell lines with relatively slow kinetics [49]. The percentage of apoptotic cells peaked between 24 and 48 hr after SBHA treatment. There was a wide variation in sensitivity of melanoma cells to SBHA-induced apoptosis, with negligible cell death in a few cell lines but nearly 50% apoptotic cells in some others. SBHA did not induce apoptosis in melanocytes even when the cells were treated for

SBHA-induced apoptosis of melanoma is dependent on changes in mitochondria

The involvement of capsase-9 but not caspase-8 in SBHA-induced apoptosis suggested that SBHA may be acting to induce changes in mitochondrial membrane permeability (MMP) leading to the release of mitochondrial apoptotic proteins [49]. This was demonstrated by reduction of mitochondrial membrane potential (ΔΨm) and release of cytochrome c and Smac/DIABLO from mitochondria into the cytosol after exposure to SBHA. Further evidence in support of the role of mitochondria in SBHA-induced apoptosis

SBHA down-regulates the expression of the antiapoptotic Bcl-2 family members and XIAP, but up-regulates the expression of pro-apoptotic Bcl-2 family members

Examination of Bcl-2 family proteins and XIAP expression in melanoma cells with or without exposure to SBHA showed that SBHA down-regulates the levels of expression of XIAP and the antiapoptotic Bcl-2 family members, Bcl-XL and Mcl-1, but up-regulates the multi-domain pro-apoptotic proteins, Bax and Bak, and BH-3 only proteins, Bid and Bim [49]. Studies by real time PCR analysis showed that alterations in Bcl-2 family protein expression may be due to the regulation of transcription by SBHA in

Possible initiating factor(s) in SBHA-induced apoptosis of melanoma

It is generally accepted that apoptosis acting via the mitochondrial pathway is dependent on the balance between pro-apoptotic BH3-only Bcl-2 family members and the antiapoptotic Bcl-2 family members. The pro-apoptotic BH3 proteins include Bid (activated by caspase-8), Bim or Bmf (transcriptionally regulated or released from damage to microtubules) [57] or the actin myoskeleton [58], and Noxa and PUMA that are up-regulated by gene transcription resulting from activation of p53 [59], [60]. The

Relation of regulation of p21 by SBHA to SBHA-induced apoptosis of melanoma

Previous studies have shown an association between treatment with HDAC inhibitors, p21 expression, cell cycle arrest and apoptosis. Studies on melanoma cell lines treated with SBHA showed that p21 was up-regulated in all the cell lines with nonmutated p53 up-regulation of p21 by HDAC inhibitors however may be independent of p53 and result from increased acetylation of the promoter regions of 21 [33]. The role of p21 in induction of apoptosis is controversial. Up-regulation of p21 was found to

SBHA sensitizes melanoma to TRAIL-induced apoptosis

Given that SBHA appears to regulate a large number of apoptosis mediators involved in either the mitochondrial apoptotic pathway (Bcl-2 family members), the death receptor pathway (caspase-8 and Bid), or the final common pathway to apoptosis (caspase-3 and XIAP), it could be predicted to sensitize melanoma cells to other apoptosis inducers, such as TRAIL which acts via these pathways. This proved correct in that co-treatment of melanoma cell lines with SBHA and TRAIL at optimal doses for

Possible mechanisms underlying sensitization of melanoma cells to TRAIL-induced apoptosis by SBHA

We examined whether SBHA sensitized melanoma cells to TRAIL-induced apoptosis by up-regulation of TRAIL-death receptor expression [37]. Studies before and after treatment with SBHA at various doses for different time periods failed to show any significant difference in the levels of TRAIL-R1 and TRAIL-R2 death receptor expression [49]. We next examined whether SBHA may overcome intracellular resistance mechanisms against TRAIL-induced apoptosis [43]. One such mechanism is inhibition of

Conclusion and prospects

The anticancer potential of HDAC inhibitors has been widely acknowledged [11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22]. This is believed to be at least partially due to their ability to induce apoptosis [14], [18], [19], [20]. Although a wide range of mechanisms has been suggested, it remains controversial as to how HDAC inhibitors induce apoptotic cell death [14], [18], [19], [20]. The studies presented here suggest that the HDAC inhibitor SBHA induces apoptosis of

Acknowledgements

This work was supported by the New South Wales State Cancer Council, New South Wales, the Melanoma and Skin Cancer Research Institute, Sydney, New South Wales, and the Hunter Melanoma Foundation, Newcastle, New South Wales, Australia.

References (81)

  • Y.T. Hsu et al.

    Bax in murine thymus is a soluble monomeric protein that displays differential detergent-induced conformations

    J. Biol. Chem.

    (1998)
  • K. Nakano et al.

    PUMA, a novel proapoptotic gene, is induced by p53

    Mol. Cell

    (2001)
  • E.H. Cheng et al.

    BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis

    Mol. Cell

    (2001)
  • P.F. Dijkers et al.

    Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1

    Curr. Biol.

    (2000)
  • J. Whitfield et al.

    Dominant-negative c-Jun promotes neuronal survival by reducing BIM expression and inhibiting mitochondrial cytochrome c release

    Neuron

    (2001)
  • Q. Wu et al.

    Transcriptional regulation during p21WAF1/CIP1-induced apoptosis in human ovarian cancer cells

    Biol. Chem.

    (2002)
  • B.F. Hinnebusch et al.

    The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation

    J. Nutr.

    (2002)
  • A.P. Bird et al.

    Methylation-induced repression—belts, braces, and chromatin

    Cell

    (1999)
  • A. Csordas

    On the biological role of histone acetylation

    Biochem. J.

    (1990)
  • T. Jenuwein et al.

    Translating the histone code

    Science

    (2001)
  • M. Grunstein

    Histone acetylation in chromatin structure and transcription

    Nature

    (1997)
  • P.A. Marks et al.

    Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells

    J. Natl. Cancer Inst.

    (2000)
  • A. Brehm et al.

    The E7 oncoprotein associates with Mi2 and histone deacetylase activity to promote cell growth

    EMBO J.

    (1999)
  • S.A. Gayther et al.

    Mutations truncating the EP300 acetylase in human cancers

    Nat. Genet.

    (2000)
  • R.J. Lin et al.

    Role of the histone deacetylase complex in acute promyelocytic leukemia

    Nature

    (1998)
  • C. Van Lint et al.

    The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation

    Gene Exp.

    (1996)
  • A. Saito et al.

    A synthetic inhibitor of histone deacetylase inhibitor, MS-27-275, with marked in vivo antitumour activity against human tumours

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

    (1999)
  • S.Y. Archer et al.

    p21(WAF1) is required for butyrate-mediated growth inhibition of human colon cancer cells

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

    (1998)
  • W. Janson et al.

    Butyrate modulates DNA damage-induced p53 response by induction of p53-independent differentiation and apoptosis

    Oncogene

    (1997)
  • P.G. Parson et al.

    Tumor selectivity and transcriptional activation by azelaic bishydroxamic acid in human melanocytic cells

    Biochem. Pharmacol.

    (1997)
  • N. Saunders et al.

    Histone deacetylase inhibitors as potential antiskin cancer agents

    Cancer Res.

    (1999)
  • P.N. Munster et al.

    The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces differentiation of human breast cancer cells

    Cancer Res.

    (2001)
  • D. Bernhard et al.

    Apoptosis induced by the histone deacetylase inhibitor sodium butyrate in human leukemic lymphoblasts

    FASEB J.

    (1999)
  • A.J. Burgess et al.

    Up-regulation of p21WAF1/CIP1 by histone deacetylase inhibitors reduces their cytotoxicity

    Mol. Pharmacol.

    (2001)
  • V. Medina et al.

    Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway

    Cancer Res.

    (1997)
  • D.C. Coffey et al.

    The histone deacetylase inhibitor, CBHA, inhibits growth of human neuroblastoma xenografts in vivo, along and synergistically with all-trans retinoic acid

    Cancer Res.

    (2001)
  • V. Sandor et al.

    Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms

    Clin. Can. Res.

    (2002)
  • M. Mandel et al.

    Bcl-2 expression regulates sodium butyrate-induced apoptosis in human MCF-7 breast cancer cells

    Cell Growth Differ.

    (1996)
  • A. Hague et al.

    Bcl-2 and bak may play a pivotal role in sodium bytyrate-induced apoptosis in colonic epithelial cells however, overexpression of Bcl-2 does not protect against bak-mediated apoptosis

    Int. J. Cancer

    (1997)
  • X. Cao et al.

    Histone deacetylase inhibitor down-regulation of bcl-xl gene expression leads to apoptotic cell death in mesothelioma

    Am. J. Respir. Cell Mol. Biol.

    (2001)
  • Cited by (97)

    • Real time detection of glutathionse in chemotherapy squamous-cell carcinoma cells of a fluorescent probe

      2019, Clinica Chimica Acta
      Citation Excerpt :

      Glutathione (GSH) offers crucial antioxidant protection from reactive oxygen species (ROS) arising from radiotherapy and chemotherapy and GSH mediates resistance to these types of treatment [1,2]. Also, GSH is associated with diseases including cancer and malformations such as cleft palate [3–6]. Reliable methods for measuring GSH will advance understanding of GSH-mediated detoxification mechanisms [7] as well as help diagnose potential disease at a molecular level [8–15].

    • Anti-cancer activity of ZnO chips by sustained zinc ion release

      2016, Toxicology Reports
      Citation Excerpt :

      Selective IAP down-regulation by pharmacological inhibition has been recently suggested to enhance the sensitization of leukemia cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis [47]. Because anti-cancer compounds can potentiate TRAIL-mediated death of cancer cells accompanied by induction of ROS and the down-regulation of anti-apoptotic molecules including survivin, cIAPs, and XIAP [20], ROS-generating ZnO materials might be used as a potent TRAIL sensitizer for cancer treatment [32,45]. In addition, claspin might be used to determine the sensitivity of cancer cells to zinc ions [21].

    View all citing articles on Scopus
    View full text