Elsevier

Cellular Signalling

Volume 40, December 2017, Pages 172-182
Cellular Signalling

Diindolylmethane and its halogenated derivatives induce protective autophagy in human prostate cancer cells via induction of the oncogenic protein AEG-1 and activation of AMP-activated protein kinase (AMPK)

https://doi.org/10.1016/j.cellsig.2017.09.006Get rights and content

Highlights

  • DIM and ring-DIMs induce the formation of autophagic vacuoles in prostate cancer cells.

  • AEG-1/AMPK downregulation sensitizes prostate cancer cells to death in presence of subtoxic concentrations of (ring-)DIM.

  • DIM- and ring-DIM-induced protective autophagy is mediated by the induction of AEG-1 and subsequent activation of AMPK.

  • Downregulation of AEG-1 induces senescence in prostate cancer cells.

Abstract

3,3′-Diindolylmethane (DIM) and its synthetic halogenated derivatives 4,4′-Br2- and 7,7′-Cl2DIM (ring-DIMs) have recently been shown to induce protective autophagy in human prostate cancer cells. The mechanisms by which DIM and ring-DIMs induce autophagy have not been elucidated. As DIM is a mitochondrial ATP-synthase inhibitor, we hypothesized that DIM and ring-DIMs induce autophagy via alteration of intracellular AMP/ATP ratios and activation of AMP-activated protein kinase (AMPK) signaling in prostate cancer cells. We found that DIM and ring-DIMs induced autophagy was accompanied by increased autophagic vacuole formation and conversion of LC3BI to LC3BII in LNCaP and C42B human prostate cancer cells. DIM and ring-DIMs also induced AMPK, ULK-1 (unc-51-like autophagy activating kinase 1; Atg1) and acetyl-CoA carboxylase (ACC) phosphorylation in a time-dependent manner. DIM and the ring-DIMs time-dependently induced the oncogenic protein astrocyte-elevated gene 1 (AEG-1) in LNCaP and C42B cells. Downregulation of AEG-1 or AMPK inhibited DIM- and ring-DIM-induced autophagy. Pretreatment with ULK1 inhibitor MRT 67307 or siRNAs targeting either AEG-1 or AMPK potentiated the cytotoxicity of DIM and ring-DIMs. Interestingly, downregulation of AEG-1 induced senescence in cells treated with overtly cytotoxic concentrations of DIM or ring-DIMs and inhibited the onset of apoptosis in response to these compounds. In summary, we have identified a novel mechanism for DIM- and ring-DIM-induced protective autophagy, via induction of AEG-1 and subsequent activation of AMPK. Our findings could facilitate the development of novel drug therapies for prostate cancer that include selective autophagy inhibitors as adjuvants.

Introduction

Prostate cancer is a major health problem worldwide, ranking as the second most common cancer in males [1] and the third leading cause of cancer-related deaths among American and Canadian men [2]. Treatment with drugs targeting androgen receptor (AR) signaling is the main therapy for early-stage androgen-dependent (AD) prostate cancer [3]. Unfortunately, many patients with AD prostate cancer will progress to an androgen-independent (AI) phenotype, which is harder to treat and often fatal [4], [5]. Diindolylmethane (DIM) is a promising anticancer agent derived from the ingestion of Brassica plants (cabbage, broccoli, etc.) [6]. We have shown that several di-halogenated analogs of DIM (ring-DIMs) have anti-androgenic effects in human AD LNCaP prostate cancer cells [7] and induce apoptosis and necrosis in LNCaP and human AI PC-3 prostate cancer cells with greater potencies than DIM [8]. More recently, we have shown that DIM and ring-DIMs induce ER stress, mitochondrial dysfunction, and autophagy in prostate cancer cells [9]. Autophagy is a self-digestion process activated by cellular stress, in which dysfunctional organelles and protein aggregates are sequestered in double-membraned vesicles [10], and then transported to lysosomes for proteolytic degradation and recycling to maintain cellular homeostasis [11].

Autophagy plays an important role in cancer cell progression; its induction in response to stresses following chemotherapy may promote cancer cell survival. However, excessive autophagy could activate a cell death mechanism known as cytotoxic autophagy, which is different from programmed cell death (apoptosis) [12], [13]. Uncontrolled growth of cancer cells is facilitated by the inactivation of cell death pathways, such as apoptosis, and stimulation of cell survival pathways [14]. Thus, the dysregulation of autophagic machinery in cancer cells, leading to imbalances in the activation of cell death- or survival-related pathways, may have a critical influence on either tumor progression or regression. Various Atg (autophagy-related) proteins are involved in the initiation and regulation of autophagy [15]. The conversion of LC3B (Atg3) from its diffuse LC3BI form to the punctuated LC3BII, which associates with the autophagosome [16], is used as a classic marker of autophagy. AMP-activated protein kinase (AMPK) regulates autophagy via phosphorylation of the autophagy-initiating protein ULK1 (Atg1) [17]. The oncogenic protein, astrocyte elevated gene-1 (AEG-1), also known as metadherin (MTDH) or protein LYRIC, is overexpressed in various cancer including that of the prostate [18], [19], where it acts as a mediator of AMPK activity and autophagy in response to cellular metabolic stress [20], [21]. AEG-1 contributes to chemoresistance in hepatocellular carcinoma (HCC) cells [22] and promotes hepato-carcinogenisis through inhibition of senescence in transgenic mice that overexpress hepatocyte-specific AEG-1 [23].

Although DIM is known to be a mitochondrial ATP synthase inhibitor [24] that alters AMP/ATP ratios leading to activation of AMPK [25], the exact mechanism(s) of DIM-induced protective autophagy has not been elucidated. Previous studies have shown that AEG-1 induces protective autophagy via activation of AMPK [21]. Therefore, we wished to determine the possible involvement of AEG-1 in AMPK activation as well as DIM- and ring-DIM-mediated induction of autophagy.

Section snippets

Cell culture and treatment

LNCaP AR-positive and AD human prostate cancer cells were purchased from the American Type Culture Collection (Manassas, VA); LNCaP C4-2B (C42B) AR-positive and AI human prostate cancer cells were purchased from the MD Anderson Cancer Centre (Houston, TX). LNCaP and C42B prostate cancer cells were cultured in RPMI 1640 medium supplemented with 10% FBS, and 1% penicillin/streptomycin (Life Technologies, Gaithersburg, MD) at 37 °C and 5% CO2. Ring-DIMs were synthesized in our laboratories as

DIM and ring-DIMs induce the formation of autophagic vacuoles in prostate cancer cells

An 8-h treatment of androgen-sensitive LNCaP and androgen-insensitive C42B cells with DIM, 4,4′-Br2DIM and 7,7′-Cl2DIM significantly increased the formation of autophagic vacuoles (Fig. 1A–D). The concentrations used in these experiments were based on our previous studies on DIM- and ring-DIM-mediated induction of protective autophagy in the same prostate cancer cells [8], [9]. We also confirmed the formation of autophagosomes by transmission electron microscopy after exposure of C42B cells to

Discussion

Our current findings demonstrate that DIM and ring-DIMs activate the AMPK signaling in LNCaP and C42B cells by increasing AMPK-, ACC-(a substrate uniquely phosphorylated by AMPK) and ULK1 phosphorylation in a time-dependent manner (Fig. 2). Chen and coworkers previously reported that a formulated DIM (B-DIM), which has increased bioavailability in vivo, can activate AMPK signaling as early as 3 h after exposure [25]. DIM has also been shown to activate AMPK signaling in ovarian cancer cells, and

Conclusion

We have identified a novel mechanism of DIM- and ring-DIM-induced protective autophagy, which is due to induction of AEG-1 and subsequent activation of AMPK. Our results suggest that development of novel drug therapies against prostate cancer could include selective autophagy inhibitors as adjuvants. Moreover, targeting DIM- and ring-DIM-mediated induction of AEG-1 and subsequent induction of senescence may be an effective novel therapy for treating prostate cancer.

The following are the

Acknowledgments

This work was funded by an operating grant from the Canadian Institutes of Health Research (CIHR grant no. MOP-115019) to JTS, EG and SS. HD received a scholarship from the Fonds de Recherche du Québec - Santé (FRQS). All authors declare to have no conflicts of interest.

References (39)

  • J. Ferlay et al.

    Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012

    Int. J. Cancer

    (2015)
  • R.L. Siegel et al.

    Cancer statistics, 2016

    CA Cancer J. Clin.

    (2016)
  • D.K. Wysowski et al.

    Fatal and nonfatal hepatotoxicity associated with flutamide

    Ann. Intern. Med.

    (1993)
  • D.G. McLeod

    Tolerability of nonsteroidal antiandrogens in the treatment of advanced prostate cancer

    Oncologist

    (1997)
  • D.G. McLeod et al.

    Combined androgen blockade: the gold standard for metastatic prostate cancer

    Eur. Urol.

    (1997)
  • L.F. Bjeldanes et al.

    Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin

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

    (1991)
  • K. Abdelbaqi et al.

    Antiandrogenic and growth inhibitory effects of ring-substituted analogs of 3,3′-diindolylmethane (ring-DIMs) in hormone-responsive LNCaP human prostate cancer cells

    Prostate

    (2011)
  • A.A. Goldberg et al.

    Ring-substituted analogs of 3,3′-diindolylmethane (DIM) induce apoptosis and necrosis in androgen-dependent and -independent prostate cancer cells

    Investig. New Drugs

    (2014)
  • A.A. Goldberg et al.

    3,3′-Diindolylmethane (DIM) and its ring-substituted halogenated analogs (ring-DIMs) induce differential mechanisms of survival and death in androgen-dependent and -independent prostate cancer cells

    Genes Cancer

    (2015)
  • C.W. Wang et al.

    The molecular mechanism of autophagy

    Mol. Med.

    (2003)
  • D. Glick et al.

    Autophagy: cellular and molecular mechanisms

    J. Pathol.

    (2010)
  • E. White

    The role for autophagy in cancer

    J. Clin. Invest.

    (2015)
  • S.Y. Yang et al.

    Dual role of autophagy in colon cancer cell survival

    Ann. Surg. Oncol.

    (2011)
  • C.B. Blackadar

    Historical review of the causes of cancer

    World J Clin Oncol.

    (2016)
  • N. Mizushima et al.

    The role of Atg proteins in autophagosome formation

    Annu. Rev. Cell Dev. Biol.

    (2011)
  • T.E. Hansen et al.

    Following autophagy step by step

    BMC Biol.

    (2011)
  • J. Kim et al.

    AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1

    Nat. Cell Biol.

    (2011)
  • G. Hu et al.

    The multifaceted role of MTDH/AEG-1 in cancer progression

    Clin. Cancer Res.

    (2009)
  • X. Shi et al.

    The role of MTDH/AEG-1 in the progression of cancer

    Int. J. Clin. Exp. Med.

    (2015)
  • Cited by (30)

    • Autophagy and beyond: Unraveling the complexity of UNC-51-like kinase 1 (ULK1) from biological functions to therapeutic implications

      2022, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      Kinsenoside activates AMPK–ULK1-dependent autophagy to alleviate alcoholic liver injury311. Activation of autophagy also plays a therapeutic role in cancer therapy regulated by AMPK/ULK1 signaling axis, such as baicalein against multiple cancers312,313; narciclasine against TNBC314; raloxifene315 against breast cancer; 3,3′-diindolylmethane against human PC316. As for drug-resistant, temozolomide activates autophagic activity through ATM–AMPK–ULK1 pathway, leading to chemotherapy resistance in glioma317.

    • The absence of cellular glucose triggers oncogene AEG-1 that instigates VEGFC in HCC: A possible genetic root cause of angiogenesis

      2022, Gene
      Citation Excerpt :

      Next, we examined the impact of GD on the formation of autophagolysosomes. Previous studies state that oncogenic protein AEG-1 induces a protective autophagy role in prostate cancer (Draz et al., 2017). Based on this evidence, LC3-a and LC3-b protein expression were used to investigate the relation between AEG-1 and autophagy.

    • Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies

      2022, Biochemical Pharmacology
      Citation Excerpt :

      In another study, in vitro effects of diindolylmethane and its derivatives significantly enhance autophagy via activation of AMPK and induction of AEG-1 towards the suppression of human prostate cancer cells. Besides, diindolylmethane derivatives promoted the phosphorylation of AMPK, acetyl-CoA carboxylase, and ULK-1 [292]. A new derivative of indenone exerted anticancer potentials by enhancing senescence and cell cycle arrest, as well as down-regulation of p53, p21CIP1/WAF1, survivin and p65/NF-κB in the MDA-MB-231 and MDA-MB-468 cells during an in vitro model of breast cancer cells [293].

    • Role of AMPK mediated pathways in autophagy and aging

      2022, Biochimie
      Citation Excerpt :

      Activation of AMPK can inhibit the occurrence of cancer, but may also promote the occurrence and development of cancer, depending on the specific situation. In LNCaP and C42B human prostate cancer cells, indolemethane and its halogenated derivatives induce protective autophagy in cancer cells by inducing phosphorylation of AMPK, ULK-1 and acetyl-CoA carboxylase [142]. Downregulation of AEG-1 or AMPK can inhibit DIM and RING-DIM-induced autophagy.

    View all citing articles on Scopus
    View full text