Cancer Letters

Cancer Letters

Volume 416, 1 March 2018, Pages 11-23
Cancer Letters

Original Articles
SEC-induced activation of ANXA7 GTPase suppresses prostate cancer metastasis

https://doi.org/10.1016/j.canlet.2017.12.008Get rights and content

Highlights

  • SEC-activated ANXA7 GTPase inhibits prostate cancer metastasis by AMPK/mTORC1/STAT3 signaling pathway.

  • RKIP-ANXA7 interaction compromises SEC-induced ANXA7 activation and blocks its downstream signaling pathway.

  • ANXA7 functionally interplays with RKIP in prostate cancer metastasis.

Abstract

Annexin A7 (ANXA7) is a suppressor of tumorigenesis and metastasis in prostate cancer. Activated ANXA7 GTPase promotes prostate cancer cell apoptosis. However, the role and underlying mechanism of ANXA7 GTPase in prostate cancer metastasis have not been established. RKIP is a metastatic suppressor and downregulated in prostate cancer metastases. The binding of RKIP and its target proteins could inhibit the activation of its interactive partners. However, the effect of RKIP on ANXA7 GTPase activation is not clear. Here, we report that activation of ANXA7 GTPase by a small molecule SEC ((S)-ethyl 1-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3- (4-methoxyphenyl)-1H-pyrazole-5-carboxylate) effectively inhibited prostate cancer metastasis. Mechanistically, activated ANXA7 promoted AMPK phosphorylation, leading to decreased mTORC1 activity, suppressed STAT3 nuclear translocation, and downregulation of pro-metastatic genes, including CCL2, APLN, and IL6ST. Conversely, RKIP interacted with ANXA7 and impaired activation of ANXA7 GTPase by SEC and its downstream signaling pathway. Notably, SEC treatment suppressed metastasis of prostate cancer cells in in vivo orthotopic analysis. Together, our findings provide a novel insight into how metastasis of prostate cancer with low RKIP expression is suppressed by SEC-induced activation of ANXA7 GTPase via the AMPK/mTORC1/STAT3 signaling pathway.

Introduction

Cancer metastasis is the major cause of cancer mortality, accounting for approximately 90% of deaths. Once cancer cells leave and spread beyond their primary site, metastatic cancers are highly incurable and fatal, whereas most primary tumors are manageable or curable [1]. Prostate cancer is the most frequently diagnosed cancer in men with an estimated 180,000 new cases in U.S. in 2016. Five-year relative survival rate of localized and regional prostate cancer reaches approximately 99%; however, only about 27% of metastatic prostate cancer patients survive in 5 years [2]. Cancer metastasis is a complicated progress that involves four essential steps: detachment, migration, invasion and adhesion [3]. Whereas our understanding of the biology behind mechanisms of androgen receptor pathway activation, tumor-microenvironment interaction, and anti-tumor immunotherapy has advanced therapies for metastatic prostate cancer [4], a better understanding of molecular and cellular mechanisms of suppressor gene and associated signaling pathway in prostate cancer metastasis is critical and in great need to identify new therapeutic targets and agents.

Annexin A7 (ANXA7), a member of annexin superfamily, is a Ca2+- and phospholipid-binding protein and possesses GTPase activity [5,6]. The ANXA7 gene is located on human chromosome 10q21, where multiple potential tumor suppressor genes exist. Homozygous ANXA7 (−/−) knockout mice showed a lethal phenotype at embryonic day 10 [7]. Heterozygous ANXA7 (±) knockout mice have a high frequency of spontaneous tumors (about 20–50%) in multiple organs, including liver, prostate, endometrium, salivery gland and thymus [8]. Moreover, tumor metastasis was observed in 42% of the mutant mice developing spontaneous tumor [8]. These findings suggest that ANXA7 may play an important role in inhibiting tumorigenesis and metastasis. Compared with that in normal and healthy prostate tissue, ANXA7 expression is reduced by approximately 50% in primary prostate tumors and even more downregulated in metastatic prostate cancers [7,9]. However, the mechanism of ANXA7 GTPase in the regulation of metastasis has not been established. Our recent work has demonstrated that activation of ANXA7 GTPase by a small molecule SEC ((S)-ethyl 1-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3-(4-methoxyphenyl)-1 H-pyrazole-5 -carboxylate) induces prostate cancer cell apoptosis [10]. We wondered whether activated ANXA7 GTPase could inhibit prostate cancer metastasis.

Raf kinase inhibitory protein (RKIP) has been associated with natural anti-metastasis property in multiple tumor types [11]. The metastatic suppression role of RKIP was initially identified in prostate cancer [12]. RKIP expression is low in metastatic prostate cancer cell lines PC3 and DU145, while RKIP is relatively high expressed in non-invasive prostate cancer cell line LNCaP [13]. Analysis of clinical samples showed that metastases of human prostate cancer exhibited dramatically decreased RKIP level compared with benign prostate hyperplasia and localized prostate cancer samples [12,13]. It has been proposed that low RKIP expression is a good prognostic marker for the pathogenesis of human prostate cancer [14].

Accumulating evidence suggests that RKIP associates with GTP-binding proteins [15]. RKIP associates with Raf-1 and inhibits the Raf/MEK/ERK/Myc signaling, leading to the inhibition of Snail and TET activity and decreased pro-metastatic gene expression [16]. RKIP interferes with NF-κB signaling by the interaction with several upstream kinases TAK1, NIK and IKK, contributing to metastasis inhibition [17]. The binding of RKIP and its target proteins could inhibit the interactive proteins activation by their activators, leading to the inversion of related signaling and the inhibition of tumor metastasis [18]. In addition, it was reported that nonylphenol induced the upregulation of RKIP in testicular Sertoli cells, accompanied with decreased ANXA7 protein level [19]. However, the role and mechanism of RKIP in the regulation of ANXA7 signaling pathway remains unknown so far. We seek to determine whether RKIP binds to ANXA7 and antagonizes the activation of ANXA7 GTPase by SEC.

In the current study, we used metastatic prostate cancer cell PC3 as well as non-metastatic LNCaP, and constructed HEK293T RKIP−/− cell line, together with HEK293T RKIP+/+ cell line as the cell model, to study the role and mechanism of ANXA7 GTPase in prostate cancer metastasis. Our substantial evidence suggests that SEC treatment increases AMPK phosphorylation via activating ANXA7 GTPase, and that activated AMPK inhibits STAT3 nuclear translocation via the mTORC1/STAT3 pathway, leading to downregulation of pro-metastatic genes and inhibition of prostate cancer metastasis. Notably, RKIP interacts with and compromises the inhibitory effect of SEC on prostate cancer metastasis. Together, our findings indicate that activation of ANXA7 by a small molecule SEC antagonizes prostate cancer metastasis with low RKIP expression via the AMPK/mTORC1/STAT3 signaling pathway, providing a novel insight into how prostate cancer metastasis is suppressed by a small molecule SEC via previously uncharacterized but essential interplay between the two metastasis suppressors ANXA7 and RKIP.

Section snippets

Antibodies

Antibodies to RKIP (CST#5291), AMPKα (CST#5832), phospho-AMPKα (T172) (CST#8208), p70S6K (CST#2708), phospho-p70S6K (CST#9208), 4EBP1 (CST#9644), phospho-4EBP1 (CST#9459), STAT3 (CST#9132), phospho-STAT3 (Tyr705) (CST#9131) and phospho-STAT3 (Ser727) (CST#9134) were purchased from Cell Signaling Technology. Antibodies to E-Cadherin (20874-1-AP) and vimentin (10366-1-AP) were obtained from Proteintech Group. Anti-ANXA7 antibody (sc-17815) used for co-immunoprecipitation and anti-p-Thr antibody

SEC inhibited migration in HEK293T RKIP−/- cells

It is well established that RKIP has an anti-metastatic property. To get an in-depth understanding of underlying mechanism, we constructed HEK293T cell lines carrying RKIP knockout (RKIP−/−) and wild-type RKIP expression (RKIP+/+) (Fig. 1A). RKIP-null HEK293T cells showed higher migration ability than wild-type RKIP expressing cells (Fig. 1B). The small molecule SEC dramatically suppressed HEK293T RKIP−/− cell migration while had no effect on HEK293T RKIP+/+ cells (Fig. 1B). Moreover, SEC

Discussion

Directly modulating specific target proteins and signaling pathways with small-molecule drugs or agonistic antibodies is widely used for cancer metastasis therapies [[39], [40], [41]]. ANXA7 acts as a metastasis suppressor in prostate cancer. However, the mechanism of ANXA7 on inhibiting prostate cancer metastasis is not clear. In the current study, we revealed for the first time that activation of ANXA7 GTPase by SEC increases AMPK phosphorylation to suppress mTORC1/STAT3 signal pathway and to

Disclosure of potential conflicts of interest

The authors declare no conflict of interest.

Acknowledgements

This work was supported by the National Natural Science Foundation of China (No. 91313303, 31570834, 81502948, 31270877), the Major Project of Science and Technology of Shandong Province (NO. 2015ZDJS04001) and Shandong Excellent Young Scientist Award Fund (No. BS2015YY031, BS2013SW001). The Yan lab was supported in part by funds provided by University of North Carolina at Charlotte and grants from the NIH/NIGMS (R15GM101571 and R15GM114713).

References (61)

  • B. Faubert et al.

    AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo

    Cell Metabol.

    (2013)
  • L. Zhang et al.

    Up-regulation of the active form of small GTPase Rab13 promotes macroautophagy in vascular endothelial cells

    Biochim. Biophys. Acta

    (2017)
  • D.M. Gwinn et al.

    AMPK phosphorylation of raptor mediates a metabolic checkpoint

    Mol. Cell

    (2008)
  • J.J. Howell et al.

    Metformin inhibits hepatic mTORC1 signaling via dose-dependent mechanisms involving AMPK and the TSC complex

    Cell Metabol.

    (2017)
  • F. Yang et al.

    Gadd45a suppresses tumor angiogenesis via inhibition of the mTOR/STAT3 protein pathway

    J. Biol. Chem.

    (2013)
  • C. Ward et al.

    Evaluation of carbonic anhydrase IX as a therapeutic target for inhibition of breast cancer invasion and metastasis using a series of in vitro breast cancer models

    Oncotarget

    (2015)
  • R.L. Siegel et al.

    Cancer statistics, 2016

    CA Cancer J. Clin.

    (2016)
  • M. Srivastava et al.

    ANX7, a candidate tumor suppressor gene for prostate cancer

    Proc. Natl. Acad. Sci. USA

    (2001)
  • M. Srivastava et al.

    Haploinsufficiency of Anx7 tumor suppressor gene and consequent genomic instability promotes tumorigenesis in the Anx7(+/-) mouse

    Proc. Natl. Acad. Sci. USA

    (2003)
  • M. Srivastava et al.

    ANXA7 expression represents hormone-relevant tumor suppression in different cancers

    Int. J. Canc.

    (2007)
  • S.Y. Liu et al.

    A small molecule induces integrin beta4 nuclear translocation and apoptosis selectively in cancer cells with high expression of integrin beta4

    Oncotarget

    (2016)
  • K. Lamiman et al.

    Survey of Raf kinase inhibitor protein (RKIP) in multiple cancer types

    Crit. Rev. Oncog.

    (2014)
  • Z. Fu et al.

    Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis

    J. Natl. Canc. Inst.

    (2003)
  • S. Beach et al.

    Snail is a repressor of RKIP transcription in metastatic prostate cancer cells

    Oncogene

    (2008)
  • Z. Fu et al.

    Metastasis suppressor gene Raf kinase inhibitor protein (RKIP) is a novel prognostic marker in prostate cancer

    Prostate

    (2006)
  • S. Bucquoy et al.

    Relationships between molecular interactions (nucleotides, lipids and proteins) and structural features of the bovine brain 21-kDa protein

    Eur. J. Biochem.

    (1994)
  • A.E. Yesilkanal et al.

    Raf kinase inhibitory protein (RKIP) as a metastasis suppressor: regulation of signaling networks in cancer

    Crit. Rev. Oncog.

    (2014)
  • L. Su et al.

    Raf kinase inhibitor protein regulates oxygen-glucose deprivation-induced PC12 cells apoptosis through the NF-kappaB and ERK pathways

    J. Clin. Biochem. Nutr.

    (2016)
  • K. Rajkumar et al.

    Understanding perspectives of signalling mechanisms regulating PEBP1 function

    Cell Biochem. Funct.

    (2016)
  • J. Wu et al.

    Proteomic analysis of changes induced by nonylphenol in Sprague-Dawley rat Sertoli cells

    Chem. Res. Toxicol.

    (2009)
  • Cited by (0)

    1

    These authors are joint first authors.

    View full text