Role of ATF3 in synergistic cancer cell killing by a combination of HDAC inhibitors and agonistic anti-DR5 antibody through ER stress in human colon cancer cells

https://doi.org/10.1016/j.bbrc.2014.01.184Get rights and content

Highlights

  • Six different HDACIs upregulate DR5 by ER stress/UPR pathway in colon cancer cells.

  • Co-treatment with HDACIs and DJR2-2 sensitizes cancer cells to apoptosis.

  • Pro-apoptotic genes ATF3 and CHOP are required to activate DR5 transcription.

  • ATF3 plays role in TRAIL-based cell death by HDACIs.

Abstract

Histone deacetylase inhibitors (HDACIs) are promising agents for cancer therapy. However, the mechanism(s) responsible for the efficacy of HDACIs have not yet to be fully elucidated. Death receptor 5 (DR5) is a transmembrane receptor containing death domain that triggers cell death upon binding to TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) or agonistic anti-DR5 monoclonal antibody, and the combination of TRAIL/agonistic anti-DR5 monoclonal antibody and agents that increase the expression of DR5 is expected as a novel anticancer therapeutic strategy. Here we report that six different HDACIs activated endoplasmic reticulum (ER) stress sensor PERK and eIF2α and induced the ATF4/ATF3/CHOP pathway in p53-deficient human colon cancer cells. This resulted in an increased expression of DR5 on the cell surface and sensitized cells to apoptosis by agonistic anti-DR5 monoclonal antibody. Stress response gene ATF3 was required for efficient DR5 induction by HDACIs, and DR5 reporter assay showed that ATF3 play crucial role for the HDACIs-induced activation of DR5 gene transcription. These provide important mechanistic insight into how HDACIs exhibit pro-apoptotic activity in clinical anti-cancer treatments when they are used in combination with other therapeutic strategies.

Introduction

Histone deacetylase inhibitors (HDACIs) are a class of targeted therapeutics with promising anti-tumor efficacy that reverse aberrant epigenetic changes associated with cancer. HDACIs induce hyper-acetylation of DNA-bound histones or sequence-specific transcription factors, which can either increase or decrease the transcription of cancer-related genes [1], [2], [3]. It has also been shown that the treatment with HDACIs results in hyper-acetylation of chaperones such as HSP90, HSP70 and HSP40, which impairs the chaperone activity and leads to the accumulation of unfolded or misfolded proteins in endoplasmic reticulum (ER) [3], [4], [5]. This causes the activation of a specific cellular process called the unfolded protein response (UPR), which is mediated through at least three major pathways initiated by IRE1, PERK or ATF6 respectively, coordinating a temporal shut-down of protein translation and a complex gene transcriptional program to safeguard cell survival. Upon excessive or prolonged UPR, however, apoptosis is activated to eliminate faulty cells [6], [7].

To date, several structurally distinct HDACIs have been developed and are classified into six groups, including short-chain fatty acids, hydroxamic acids, cyclic peptides, benzamides, electrophilic ketones, and hybrid molecules [8]. At present, an increasing number of HDACIs have been reported as candidate for stand-alone therapeutics or combination therapy with other agents. For instance, suberoylanilide hydroxamic acid (SAHA, also called Vorinostat) is a broad set and non-selective inhibitor of HDACs. After its approval for the treatment of cutaneous T cell lymphoma [9], SAHA is trying to find its value for solid tumors including thyroid cancer [10], breast cancer [11] and mesothelioma [12]. In colon cancer, it is reported that SAHA can induce sub-G1 arrest and apoptosis [13], [14].

Death receptor 5 (DR5) belongs to the TNF receptor family. Upon binding of its ligand TRAIL or agonistic antibody, DR5 mediates TRAIL-induced apoptosis through the formation of a death-inducing signaling complex [15]. DR5 is expressed more strongly in cancer cells than in normal cells [16], and mediates apoptosis selectively in cancer cells both in vitro and in vivo, with little or no effect on normal cells. However, clinical trials have shown that small therapeutic effects are observed when TRAIL or TRAIL agonistic monoclonal antibodies specific for DR5, such as DJR2-2 [17], [18] are used as monotherapy. Thus, it is required to maximize the therapeutic efficacy through drug combination that synergize with TRAIL-based agents. It has been shown that some HDACIs such as Butyrate may increase the expression of DR5 through Sp1 in colon cancer cells [19]. However, it is unclear whether there are other mechanisms at work as well.

Stress response gene ATF3 is a member of the ATF/CREB family. Its messenger RNA (mRNA) level is low or undetectable, but is greatly induced upon exposure of cells to a variety of stress signals including DNA damage, oxidative stress, and cytokines and growth factors [20]. We have previously shown that ATF3 plays a role in p53-dependent DR5 induction upon DNA damage of human colorectal cancer cells [21]. More recently, Wilson et al. have reported that HDACIs sensitized colon cancer cells to apoptosis by a Sp1/Sp3-dependent transcriptional program [22]. However, it still remains elusive whether ATF3 plays role in the transcriptional networks or cell death provoked by HDACIs in human colon cancer cells.

The present study reports that at least six structurally different HDAC inhibitors caused ER stress and activated the PERK-eIF2α pathway of the UPR, leading to up-regulation of DR5 expression on the cell surface of p53-deficient human colon cancer cells. Combined treatment with HDACIs and anti-DR5 monoclonal antibody synergistically sensitized cancer cells to apoptotic cell death. Functional role of ATF3 in HDACIs-induced DR5 gene transcription and cell death was investigated.

Section snippets

Plasmids, antibodies, and reagents

Retroviral expression plasmid pMX-ATF3 encoding human ATF3 gene was constructed by subcloning cDNA of human ATF3 into pMXs-puro provided by Dr. T Kitamura (University of Tokyo, Japan). Antibodies: anti-KDEL from SressGen (Glanford, Victoria, Canada), anti-phosphorylated PERK (Thr981), anti-PERK (C16), anti-eIF2α (FL315), anti-ATF3 (C19), anti-ATF4 (C20), anti-CHOP (R20) and anti-DR5 (N19) from Santa Cruz (Santa Cruz, CA, USA), anti-β-actin (A2228) from Sigma–Aldrich (St. Louis, MO, USA),

HDACIs cause ER stress and increase the expression of ATF4, ATF3, CHOP and DR5 in p53-deficient human colon cancer cells

To understand the effect of HDACIs on human colon cancer cells, we first investigated the effect of SAHA on the expression of ER stress markers and the downstream genes ATF4, ATF3, CHOP and DR5 in human colon cancer cell lines harboring p53 mutations. As shown in Fig. 1A, SAHA significantly increased the expression of ER chaperon GRP78 and elevated the phosphorylation level of RNA-dependent protein kinase-like ER kinase (PERK) and eukaryotic translation initiation factor 2 on the alpha subunit

Discussion

In this study, we have clearly demonstrated that HDACIs including SAHA, TSA, M344, MS-275, But and VPA, all caused the induction of GRP78 and the activation of PERK in ER in human p53-deficient colorectal cancer cells. This is in part consistent with previous reports in which HDACI activated the PERK-eIF2α [4], and supports that HDACIs cause the accumulation of misfolded or unfolded proteins in the ER. It should be noted that six different HDACIs used in this study belong to three structurally

Acknowledgments

This study was supported by Grants from JSPS KAKENHI (#24590373) and the Ministry of Health, Labour and Welfare of Japan (#25134708).

References (28)

  • O. Khan et al.

    Drug insight: histone deacetylase inhibitor-based therapies for cutaneous T-cell lymphomas

    Nat. Clin. Pract. Oncol.

    (2008)
  • E. Borbone et al.

    Histone deacetylase inhibitors induce thyroid cancer-specific apoptosis through proteasome-dependent inhibition of TRAIL degradation

    Oncogene

    (2010)
  • S. Shankar et al.

    Suberoylanilide hydroxamic acid (Zolinza/vorinostat) sensitizes TRAIL-resistant breast cancer cells orthotopically implanted in BALB/c nude mice

    Mol. Cancer Ther.

    (2009)
  • N. Crawford et al.

    SAHA overcomes FLIP-mediated inhibition of SMAC mimetic-induced apoptosis in mesothelioma

    Cell Death Dis.

    (2013)
  • Cited by (22)

    • Emerging roles of activating transcription factor (ATF) family members in tumourigenesis and immunity: Implications in cancer immunotherapy

      2022, Genes and Diseases
      Citation Excerpt :

      In the cellular context of cancer, ATF3 may be a tumour suppressor gene or an oncogene.73 ATF3 is the target gene of multiple signalling pathway networks, including JNK,74–76 p53,77,78 c-Myc79,80 and Smad.81 The stress response gene ATF3 is transcriptionally activated by binding of β-catenin and T-cell specific transcription factor (TCF4) to the redundant TCF4 site at the proximal promoter region of the ATF3 gene, indicating that ATF3 is a direct target of the Wnt/β-catenin pathway.

    • Exploring the role of post-translational modulators of transcription factors in triple-negative breast cancer gene expression

      2020, Meta Gene
      Citation Excerpt :

      There are 13 TFs included in cell proliferation network (Fig. 4B) of which 11 (ATF3, PRRX2, FOXO1, SMAD4, ESR1, ESR2, ETS1, CEBPB, YBX1, NFIB, AR) were important for the regulation of key biological processes involved in carcinogenesis. The gene ATF3 was found to be associated with apoptosis (Chen et al., 1996; Wek et al., 2006; Yin et al., 2008) through upregulating proapoptotic proteins (Liu et al., 2014; Tian et al., 2011; Xu et al., 2012). ETS1 transcription factors may play a role in the disease progression of breast cancer (Lincoln 2nd and Bove, 2005) through the regulation of key cellular events involved in angiogenic and extracellular matrix remodeling factors (Fry and Inoue KJCr, 2018).

    • Activating Transcription Factor 3 as a Novel Regulator of Chemotherapy Response in Breast Cancer

      2018, Translational Oncology
      Citation Excerpt :

      ATF3 functions by upregulating genes involved in alleviating cellular stress; however, when the stress cannot be overcome, enhanced and sustained expression of ATF3 promotes apoptosis [9–11]. Apoptosis can be initiated through the upregulation of the downstream target of ATF3, DDIT3 (CHOP/GADD153), which upregulates proapoptotic proteins [12–14]. Multiple signaling pathways have been demonstrated to regulate ATF3 expression, including the DNA-damage response (DDR), integrated stress response (ISR), and MAPK signaling pathways [15–17].

    • Inhibition of SRC-3 enhances sensitivity of human cancer cells to histone deacetylase inhibitors

      2016, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      HDAC are highexpressed in a wide variety of cancers. HDAC inhibitors have been anticipated to exert substantial anti-tumor activity by selectively upregulating expression of epigenetically silenced tumor suppressors through transcription-dependent mechanisms [13,14]. Notably, combinations of decitabine with HDAC inhibitors have been approved by FDA for treatment of hematological malignancies [15].

    • Induction of Activating Transcription Factor 3 Is Associated with Cisplatin Responsiveness in Non–Small Cell Lung Carcinoma Cells

      2016, Neoplasia (United States)
      Citation Excerpt :

      This suggests a potential role for ATF3 expression in overcoming cisplatin resistance that requires further study. Of interest, vorinostat was identified in both library screens and was previously shown to enhance cisplatin responsiveness [32,33]; however, the mechanism of action of this phenomenon has not been determined. We first confirmed the differential sensitivity of the wild-type and ATF3−/− MEFs to vorinostat treatments employing 0- to 10-μM treatments for 48 hours (Figure 6A), clearly demonstrating a role for ATF3 in regulating vorinostat cytotoxicity.

    View all citing articles on Scopus
    View full text