Cancer Letters

Cancer Letters

Volume 424, 28 June 2018, Pages 30-45
Cancer Letters

Original Articles
15-Deoxy-Δ12,14-prostaglandin J2 activates PI3K-Akt signaling in human breast cancer cells through covalent modification of the tumor suppressor PTEN at cysteine 136

https://doi.org/10.1016/j.canlet.2018.03.016Get rights and content

Highlights

  • PTEN is inactivated through covalent modification by 15-deoxy-Δ12,14-prostaglandin J2 in breast cancer MCF-7 cells.

  • 15-Deoxy-Δ12,14-prostaglandin J2 directly binds to the cysteine 136 residue of the recombinant PTEN.

  • PTEN interaction with 15-deoxy-Δ12,14-prostaglandin J2 stimulates Akt phosphorylation.

  • 15-Deoxy-Δ12,14-prostaglandin J2 stimulates growth of MDA-MB-231 cells when inoculated to the athymic nude mice.

Abstract

15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), one of the terminal products of cyclooxygenase-2-catalized arachidonic acid metabolism, has been shown to stimulate breast cancer cell proliferation and migration through Akt activation, but the underlying mechanisms remain poorly understood. In the present study, we investigated the effects of 15d-PGJ2 on the activity of PTEN, the inhibitor of the phosphoinositide 3-kinase (PI3K)-Akt axis, in human breast cancer (MCF-7) cells. Since the α,β-unsaturated carbonyl moiety in the cyclopentenone ring of 15d-PGJ2 is electrophilic, we hypothesized that 15d-PGJ2-induced Akt phosphorylation might result from the covalent modification and subsequent inactivation of PTEN that has several critical cysteine residues. When treated to MCF-7 cells, 15d-PGJ2 bound to PTEN, and this was abolished in the presence of the thiol-reducing agent dithiothreitol. A mass spectrometric analysis by using recombinant and endogenous PTEN protein revealed that the cysteine 136 residue (Cys136) of PTEN is covalently modified upon treatment with 15d-PGJ2. Notably, the ability of 15d-PGJ2 to covalently bind to PTEN as well as to induce Akt phosphorylation was abolished in the cells expressing a mutant form of PTEN in which Cys136 was replaced by serine (C136S-PTEN). The present study demonstrates for the first time that electrophilic 15d-PGJ2 directly binds to cysteine 136 of PTEN and provides new insight into PTEN loss in cancer progression associated with chronic inflammation. These observations suggest that 15d-PGJ2 can undergo nucleophilic addition to PTEN, presumably at Cys136, thereby inactivating this tumor suppressor protein with concomitant Akt activation.

Introduction

Breast cancer is one of the leading causes of cancer-related deaths in women [1, 2]. As in the case of the majority of other malignancies, inflammation is associated with pathogenesis of breast cancer. Cyclooxygenase-2 (COX-2), a key enzyme involved in inflammatory response, is frequently overexpressed in various cancer types including breast cancer [[3], [4], [5], [6]]. Abnormally elevated COX-2 expression or activity causes stimulation of cancer cell proliferation, resistance to cancer cell apoptosis, and enhancement of angiogenesis and invasiveness which account for its oncogenic function [7]. COX-2 catalyzes the conversion of arachidonic acid to a series of prostaglandins (PGs), some of which have been reported to play roles in carcinogenesis [4, 5, 8]. Of note, aberrant induction of COX-2 in breast cancer is correlated with an increased production of 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) [8]. 15d-PGJ2 has been known to have oncogenic characteristics [9]. 15d-PGJ2 contains a chemically reactive α,β-unsaturated carbonyl moiety in its cyclopentenone ring. Because of its electrophilic nature, 15d-PGJ2 can form a covalent adduct with free thiols of cysteine residues present in various proteins via the Michael addition reaction. Proteins which are known to be covalently modified by 15d-PGJ2 include nuclear factor-kappa B (NF-κB) [10], activator protein-1 (AP-1) [11], H-Ras [12], the tumor suppressor protein p53 [13], eukaryotic initiation factor 4A [14], Rac1 [15], c-Jun [16] and Hu antigen R [17].

Our previous study demonstrated that 15d-PGJ2 induced COX-2 expression in human breast cancer (MCF-7) cells via the Akt-AP-1 signaling pathway [8]. Akt activation by 15d-PGJ2 was considered to be responsible for pro-inflammatory and plausible tumor-promoting effects of this cyclopentenone PG [8]. However, the molecular mechanism underlying 15d-PGJ2-induced Akt activation remains to be clarified. The tumor suppressor, phosphatase and tensin homolog (PTEN) has been known to counteract the function of PI3K and consequently repress the Akt activation via its lipid phosphatase activity [18]. In multiple tumor types, the functional inactivation of PTEN has been observed [[19], [20], [21], [22]]. In line with this notion, the loss of PTEN gene expression is associated with poor outcome in breast cancer [23]. Notably, overexpression of COX-2 inactivates signaling molecules including PTEN [24, 25]. Upon treatment with arachidonic acid, Akt phosphorylation was induced through oxidation and concurrent inactivation of PTEN in COX-2-overexpressing pancreatic cancer (BxPC3) cells [24]. The principal catalytic function of PTEN is to dephosphorylate phosphatidylinositol-3,4,5-triphosphate (PIP3), which is a potent activator of Akt and 3-phosphoinositide-dependent kinase [26]. The unique PIP3 lipid phosphatase activity of PTEN makes it one of the most important tumor suppressors. Loss of PTEN function leads to increased accumulation of PIP3 and sequential reactivation of the PI3K-Akt signaling that stimulates cell growth and survival [27].

Alteration of PTEN functional activity by post-translational modifications has been proposed as a therapeutically attractive strategy [[28], [29], [30], [31]]. Several studies have convinced that the catalytic activity of PTEN is modulated by reactive oxygen species (ROS) and reactive nitrogen species [29, 32, 33]. PTEN contains several conserved cysteine residues that are susceptible to oxidation or nitrosylation. These catalytic cysteine residues might be also potential targets for Michael addition by electrophilic species. Previous studies from this laboratory and others demonstrated the intracellular production of ROS by 15d-PGJ2. As 15d-PGJ2 has both pro-oxidant and electrophilic properties, this prompted us to investigate whether 15d-PGJ2 could induce Akt phosphorylation through oxidation and/or covalent modification and subsequent inactivation of PTEN in human breast cancer cells.

Section snippets

Materials

15d-PGJ2 was obtained from Cayman Chemical Co. (Ann Arbor, MI). Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid), dithiothreitol (DTT), l-ascorbic acid (vitamin C) and N-acetyl cysteine (NAC) were purchased from Sigma-Aldrich (St. Louis, MO). RPMI 1640 medium and fetal bovine serum were purchased from Gibco BRL (Grand Island, NY). Antibodies against Akt, phospho-Akt, PTEN, and hemagglutinin (HA) tag were products of Cell Signaling Technology (Danvers, MA). Antibody against

15d-PGJ2 induces the activation of Akt in breast cancer cells

To explore the effect of 15d-PGJ2 on activation of the oncoprotein Akt in breast cancer cells, we utilized three different subtypes of breast cancer cell lines [estrogen receptor (ER)-positive, MCF-7; ER-negative, MDA-MB-231; Her-2 enriched: SKBr-3], all of which harbor the wild-type PTEN gene [35]. At first, we intended to determine whether the effect of 15d-PGJ2 on Akt activation is a general phenomenon in breast cancer cells which contain functional PTEN gene. However, while the level of

Discussion

Chronic inflammation is one of the representative risk factors for cancer development and progression [3]. Abnormally increased COX-2 expression has been frequently observed in breast carcinogenesis [43, 44]. Thus, overexpression of COX-2 is commonly associated with poor prognosis and also linked to metastasis and angiogenesis in breast cancer [43, 45]. Mice genetically engineered to overexpress COX-2 in mammary glands developed malignancies, while COX-2 knockout mice were less susceptible to

Conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgements

This study was supported by the Global Core Research Center (GCRC) grant (No. 2011-0030001) from the National Research Foundation (NRF) of Republic of Korea.

References (83)

  • S.R. Lee et al.

    Reversible inactivation of the tumor suppressor PTEN by H2O2

    J. Biol. Chem.

    (2002)
  • J.O. Lee et al.

    Crystal structure of the PTEN tumor suppressor: implications for its phosphoinositide phosphatase activity and membrane association

    Cell

    (1999)
  • H.K. Na et al.

    Peroxisome proliferator-activated receptor γ (PPARγ) ligands as bifunctional regulators of cell proliferation

    Biochem. Pharmacol.

    (2003)
  • Y. Kase et al.

    Preventive effects of Hange-shashin-to on irinotecan hydrochloride-caused diarrhea and its relevance to the colonic prostaglandin E2 and water absorption in the rat

    Jpn J. Pharmacol.

    (1997)
  • T. Shibata et al.

    15-deoxy-Δ12,14-prostaglandin J2. A prostaglandin D2 metabolite generated during inflammatory processes

    J. Biol. Chem.

    (2002)
  • E.H. Kim et al.

    The role of 15-deoxy-Δ12,14-prostaglandin J2, an endogenous ligand of peroxisome proliferator-activated receptor γ, in tumor angiogenesis

    Biochem. Pharmacol.

    (2008)
  • E.H. Kim et al.

    15-deoxy-Δ12,14-prostaglandin J2 as a potential endogenous regulator of redox-sensitive transcription factors

    Biochem. Pharmacol.

    (2006)
  • Y. Azuma et al.

    Possible involvement of p38 in mechanisms underlying acceleration of proliferation by 15-deoxy-Δ12,14-prostaglandin J2 and the precursors in leukemia cell line THP-1

    J. Pharmacol. Sci.

    (2004)
  • M. Emi et al.

    The biphasic effects of cyclopentenone prostaglandins, prostaglandin J2 and 15-deoxy-Δ12,14-prostaglandin J2 on proliferation and apoptosis in rat basophilic leukemia (RBL-2H3) cells

    Biochem. Pharmacol.

    (2004)
  • M. Kondo et al.

    Cyclopentenone prostaglandins as potential inducers of intracellular oxidative stress

    J. Biol. Chem.

    (2001)
  • S.M. Marino et al.

    Analysis and functional prediction of reactive cysteine residues

    J. Biol. Chem.

    (2012)
  • R.L. Siegel et al.

    Cancer statistics, 2015

    CA A Cancer J. Clin.

    (2015)
  • K.W. Jung et al.

    Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2012

    Canc. Treat Res. Off. J. Kor. Canc. Assoc.

    (2015)
  • A. Mantovani et al.

    Cancer-related inflammation

    Nature

    (2008)
  • B. Singh et al.

    COX-2 involvement in breast cancer metastasis to bone

    Oncogene

    (2007)
  • A. Greenhough et al.

    The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment

    Carcinogenesis

    (2009)
  • C. Sobolewski et al.

    The role of cyclooxygenase-2 in cell proliferation and cell death in human malignancies

    Int. J. Cell Biol.

    (2010)
  • E.H. Kim et al.

    15-Deoxy-Δ12,14-prostaglandin J2 induces COX-2 expression through Akt-driven AP-1 activation in human breast cancer cells: a potential role of ROS

    Carcinogenesis

    (2008)
  • L.J. Marnett

    Aspirin and the potential role of prostaglandins in colon cancer

    Canc. Res.

    (1992)
  • A. Rossi et al.

    Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IκB kinase

    Nature

    (2000)
  • J.L. Oliva et al.

    The cyclopentenone 15-deoxy-Δ12,14-prostaglandin J2 binds to and activates H-Ras

    Proc. Natl. Acad. Sci. USA

    (2003)
  • D.H. Kim et al.

    15-Deoxy-Δ12,14-prostaglandin J2 stabilizes, but functionally inactivates p53 by binding to the cysteine 277 residue

    Oncogene

    (2010)
  • W.J. Kim et al.

    Anti-inflammatory lipid mediator 15d-PGJ2 inhibits translation through inactivation of eIF4A

    EMBO J.

    (2007)
  • F.J. Sanchez-Gomez et al.

    Protein thiol modification by 15-deoxy-Δ12,14-prostaglandin J2 addition in mesangial cells: role in the inhibition of pro-inflammatory genes

    Mol. Pharmacol.

    (2004)
  • M. Keniry et al.

    The role of PTEN signaling perturbations in cancer and in targeted therapy

    Oncogene

    (2008)
  • G. Li et al.

    Conditional loss of PTEN leads to precocious development and neoplasia in the mammary gland

    Development

    (2002)
  • T. Daikoku et al.

    Conditional loss of uterine Pten unfailingly and rapidly induces endometrial cancer in mice

    Canc. Res.

    (2008)
  • J.M. Garcia et al.

    Promoter methylation of the PTEN gene is a common molecular change in breast cancer

    Genes Chromosomes Cancer

    (2004)
  • T.M. Covey et al.

    Akt activation by arachidonic acid metabolism occurs via oxidation and inactivation of PTEN tumor suppressor

    Oncogene

    (2007)
  • A. Silva et al.

    PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability

    J. Clin. Invest.

    (2008)
  • F. Fouladkou et al.

    The ubiquitin ligase Nedd4-1 is dispensable for the regulation of PTEN stability and localization

    Proc. Natl. Acad. Sci. USA

    (2008)
  • Cited by (0)

    View full text