Elsevier

Cellular Signalling

Volume 59, July 2019, Pages 24-33
Cellular Signalling

TMEPAI/PMEPA1 inhibits Wnt signaling by regulating β-catenin stability and nuclear accumulation in triple negative breast cancer cells

https://doi.org/10.1016/j.cellsig.2019.03.016Get rights and content

Highlights

  • TMEPAI/PMEPA1 suppresses β-catenin/TCF4 transcriptional activity on Wnt target genes.

  • TMEPAI regulates β-catenin stability and nuclear accumulation.

  • TMEPAI PY motifs are essential for Wnt signaling suppression.

Abstract

Transmembrane prostate androgen-induced protein (TMEPAI) is a type I transmembrane protein induced by several intracellular signaling pathways such as androgen, TGF-β, EGF, and Wnt signaling. It has been reported that TMEPAI functions to suppress TGF-β and androgen signaling but here, we report a novel function of TMEPAI in Wnt signaling suppression. First, we show that TMEPAI significantly inhibits TCF/LEF transcriptional activity stimulated by Wnt3A, LiCl, and β-catenin. Mechanistically, TMEPAI overexpression prevented β-catenin accumulation in the nucleus and TMEPAI knockout in triple negative breast cancer cell lines promoted β-catenin stability and nuclear accumulation together with increased mRNA levels of Wnt target genes AXIN2 and c-MYC. The presence of TGF-β type I receptor kinase inhibitor did not affect the enhanced mRNA expression of AXIN2 in TMEPAI knockout cells. These data suggest that TMEPAI suppresses Wnt signaling by interfering with β-catenin stability and nuclear translocation in a TGF-β signaling-independent manner.

Introduction

Transmembrane prostate androgen induced protein (TMEPAI), also known as prostate transmembrane protein, androgen induced 1 (PMEPA1), or solid tumor associated gene 1 (STAG1), was originally identified as a prostatic protein induced by testosterone or its derivatives [1,2]. It is a type 1b transmembrane protein consisting of a short extracellular domain, a single-pass transmembrane domain (TM), and intracellular domain, including two PPxY (PY) motifs and a Smad interaction motif (SIM) [3,4].

During tumor progression, cancer cells undergo a number of alterations in cellular signaling pathways which control cell proliferation, survival, motility, and metabolism. Many proteins in these signaling pathways are currently under investigation as cancer therapeutic targets [5,6]. TMEPAI is constitutively and highly expressed in many types of cancers including triple negative breast cancer (TNBC), an aggressive subtype of breast cancer compared with other types of breast cancers, and associated with poor prognoses in TNBC [7,8]. In consistent with these findings, we previously demonstrated that knockdown of TMEPAI in human lung cancer cells reduces tumorigenic activities (such as xenograft tumor formation and sphere formation) [9], TMEPAI is thought to be an oncogenic protein even though its role in tumorigenesis is still not fully understood.

TMEPAI itself is induced by TGF-β [4,7,8,10] and ERK signaling [2,11]. In addition, our previous study showed that TMEPAI is a downstream target of Wnt signaling in which the Wnt/β-catenin/TCF7L2 pathway preferentially activates TMEPAI/PMEPA1 gene transcription together with TGF-β signaling [12]. Since the Wnt signaling pathway controls cell proliferation and stem cell maintenance [13], we investigated the role of TMEPAI in Wnt signaling with respect to mechanistic interaction with Wnt downstream targets. Here, we demonstrate that TMEPAI inhibits Wnt signaling and subsequent AXIN2 and c-MYC transcription by regulating β-catenin protein stability and nuclear accumulation.

Section snippets

Cell culture

MDA-MB-231, Hs578T, human embryonic kidney (HEK) 293, and HEK-293 T cells were cultured in Dulbecco's modified Eagle's medium (DMEM) (Sigma) containing 10% fetal bovine serum (FBS) (Life Technologies) and penicillin-streptomycin solution (Wako). Hs578T cells and Hs578T cells stably expressing TMEPAI isoform A were cultured in the above medium supplemented with 10 μg/ml insulin and 8 μg/ml blasticidin hydrochloride. BT-549 cells were cultured in RPMI-1640 medium (Sigma) supplemented with 10%

TMEPAI inhibits Wnt signal-induced TOP-flash-luciferase reporter activity

Human TMEPAI transcription results in four isoforms (A, B, C and D) (Sup. Fig. 1) and is coordinated by both TGF-β and Wnt signaling [4,12]. First, we confirmed our previous finding of the inhibitory role of TMEPAI in TGF-β/Smad signaling [4]. As expected, overexpression of TMEPAI isoforms A, B, and D in HEK-293 cells inhibited the (CAGA)12-luciferase reporter, which is specifically activated by the TGF-β-induced Smad3 and Smad4 complex (Sup. Fig. 2). We then tested the role of TMEPAI in Wnt

Discussion

TGF-β and Wnt signaling pathways are important for cellular function and activation of both pathways is required for many developmental and patterning events [28]. Multiple studies have described the signaling crosstalk between TGF-β and Wnt pathways by showing that Axin can associate with the TGF-β-regulated Smad2 and Smad3 [[22], [23], [24], [25], [26], [27]]. Mouse models have indicated the cooperation of TGF-β and Wnt signaling in cancer [28], although much remains to be understood on how

Conclusions

Collectively, our data support the inhibitory role of TMEPAI in Wnt signaling. TMEPAI regulates β-catenin stability, nuclear accumulation, and the resultant transcriptional activity that affects the expression of Wnt target genes AXIN2 and c-MYC.

Conflict of interest

The authors declare that there are no conflicts of interest.

Author contributions

Conception and design of the work: R.A., Y.W., M.K. Acquisition of data: R.A., M.A., M.U.P., J.H., F. A., Y.W. Analysis and interpretation of data: R.A., M.A., M.U.P., J.H., F. A., Y.W., M.K. Drafting or correcting the manuscript: R.A., Y.W., M.K. All authors read and approved of the final article.

Acknowledgements

This work was supported by Grant-in-Aid for Young Scientists (B) [JP25870093 (Y.W.), JP16K19100 (Y.W.)], for Scientific Research (B) [JP25293092 (M.K.)], for Scientific Research on Innovative Area [JP26116707 (M.K.)] and Research Grant from the Uehara Memorial Foundation (Y.W.). The authors thank George Church for the kind gifts of hCas9 expression vector (Addgene plasmid #41815) and gRNA_cloning vector (Addgene plasmid #41824). We also thank Dr. Bryan J. Mathis of the Medical English

References (32)

  • D.M. DiRenzo et al.

    A crosstalk between TGF-β/Smad3 and Wnt/β-catenin pathways promotes vascular smooth muscle cell proliferation

    J. Cell. Sig.

    (2016)
  • H. Li et al.

    A feedback loop between the androgen receptor and a NEDD4-binding protein, PMEAP1, in prostate cancer cells

    J. Biol. Chem.

    (2008)
  • P.G.J. Fournier et al.

    The TGF-β signaling regulator PMEPA1 suppresses prostate cancer metastases to bone

    Cancer Cell

    (2015)
  • G. Giannini et al.

    EGF- and cell-cycle-regulated STAG1/PMEPA1/ERG1.2 belongs to a conserved gene family and is overexpressed and amplified in breast and ovarian cancer

    Mol. Carcinog.

    (2003)
  • L.L. Xu et al.

    PMEPA1, an androgen-regulated NEDD4-binding protein, exhibits cell growth inhibitory function and decreased expression during prostate cancer progression

    Cancer Res.

    (2003)
  • A. Saadi et al.

    Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers

    PNAS.

    (2010)
  • Cited by (20)

    • AGO-RBP crosstalk on target mRNAs: Implications in miRNA-guided gene silencing and cancer

      2022, Translational Oncology
      Citation Excerpt :

      Recently, we reported that CSDE1 interacts with AGO2 within miRISC and particularly, in melanoma cells, this association is facilitated by target mRNAs [41,106]. CSDE1 and AGO2 share a total of 93 targets, including Prostate Transmembrane Protein, Androgen Induced 1 (PMEPA1), a protein that enhances tumorigenic properties in breast, lung cancers and glioblastoma whilst displaying anti-tumoral roles in prostate cancer [107–109]. CSDE1 competes with miR-129-5p/AGO2 to bind PMEPA1 mRNA and promote its expression at protein levels.

    • PMEPA1 Stimulates the Proliferation, Colony Formation of Pancreatic Cancer Cells via the MAPK Signaling Pathway

      2021, American Journal of the Medical Sciences
      Citation Excerpt :

      Several studies have defined that PMEPA1 is highly expressed in various types of solid cancers, such as breast carcinoma and hepatocellular carcinoma.14, 15 Furthermore, there is evidence that high PMEPA1 patients with triple-negative breast cancer have a high risk of metastasis and unfavorable survival.15 As is well known, the typical hallmark of cancer is the acquisition of sustained proliferative, invasive and disseminating capacities during the course of carcinogenesis and progression,16 which has been verified as the main factor contributing to the poor prognosis of PC patients.

    • Diagnostic and therapeutic values of PMEPA1 and its correlation with tumor immunity in pan-cancer

      2021, Life Sciences
      Citation Excerpt :

      Interestingly, the correlation between PMEPA1 expression and tumor progression in different subtypes of prostate cancer is very different [29]. Its role in the occurrence and maintenance of different cancers remains controversial; however, its involvement in the pathogenesis of several types of cancers is proven [30,31]. PMEPA1 can be activated by a variety of cytokines that play an important role in the course of cancers, such as androgen, TGF-β, EGF, PDGF, GM-CSF and IGF, confirming PMEPA1 role tumorigenesis [26,32,33].

    • miR-130a-3p regulates steroid hormone synthesis in goat ovarian granulosa cells by targeting the PMEPA1 gene

      2021, Theriogenology
      Citation Excerpt :

      At the same time, PMEPA1 can also mediate the degradation of TGF-β type I receptors, further inhibiting the TGF-β signaling pathway [30]. Hence, both the TGF-β signaling pathway and AR are involved in the proliferation, differentiation, apoptosis, and secretion of ovarian GCs [31] and PMEPA1 can inhibit the further transmission of both. It could be speculated therefore that PMEPA1 has a certain regulatory role in GCs and in follicular development.

    • Prostaglandin F<inf>2α</inf>-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity

      2019, Neoplasia (United States)
      Citation Excerpt :

      Indeed, PMEPA1 was able to elevate functional E-cadherin levels and thus concentrate part of the β-catenin in the intercellular junctions, while other epithelial markers failed to be induced. We demonstrate that β-catenin remains in a cytoplasmic and/or membrane localization in a PMEPA1 dependent manner, in agreement with a very recent publication by Amalia et al., showing how PMEPA1 inhibited Wnt signaling through β-catenin stability and nuclear localization regulation [46]. E-cadherin can be degraded or downregulated upon different stimuli, as TGF-β [47], and thus lose association with β-catenin, that could translocate to the nucleus.

    View all citing articles on Scopus
    View full text