PRMT6 promotes endometrial cancer via AKT/mTOR signaling and indicates poor prognosis

https://doi.org/10.1016/j.biocel.2019.105681Get rights and content

Highlights

  • PRMT6 expression is upregulated in EMC and correlated with poor outcomes in two independent cohorts of 564 patients.

  • PRMT6 is targeted and downregulated by miR-372-3p in EMC cells.

  • PRMT6 promotes EMC cell proliferation and migration.

  • PRMT6 exerts oncogenic effects via activation of AKT/mTOR pathway.

Abstract

Arginine methylation plays essential roles in post-transcriptional modification and signal transduction. Dysregulation of protein arginine methyltransferases (PRMTs) has been reported in human cancers, yet the expression and biological function of PRMT6 in endometrial cancer (EMC) remains unclear. Here, we show that PRMT6 is upregulated in EMC and exhibits oncogenic activities via activation of AKT/mTOR pathway. The expression of PRMT6 in EMC is much higher than that in the adjacent nontumorous tissues. Elevated PRMT6 expression is significantly associated with higher histological tumor grade and unfavorable prognosis in two independent cohorts consisting of a total of 564 patients with EMC. In vitro data demonstrate that PRMT6 expression was identified as a downstream target of miR-372-3p. Ectopic expression of miR-372-3p downregulates PRMT6. Overexpression of PRMT6 promotes EMC cell proliferation and migration, whereas knockdown of PRMT6 leads to opposite phenotypes. Mechanistically, PRMT6 induces the phosphorylation of AKT and mTOR in EMC cells. Inhibition of AKT/mTOR signaling by MK2206 or rapamycin attenuates the PRMT6-mediated EMC progression. In clinical samples, high expression of PRMT6 was correlated to low expression of miR-372-3p and high expression of phosphorylated AKT. Collectively, our findings suggest PRMT6 may function as an oncogene to promote tumor progression, and be of prognostic value to predict disease-free survival of patients with EMC. The newly identified miR-372-3p/PRMT6/AKT/mTOR axis represents a new promising target for EMC management.

Introduction

Endometrial cancer (EMC), representing the most common pelvic gynecological malignancy in industrialized countries, ranks the fourth most common malignancy among women in the United States. In 2017, 61,380 newly diagnosed cases and 10,920 EMC-related deaths were estimated (Bray et al., 2018). The mortality of EMC is much higher than that of cervical and ovarian cancers (Siegel et al., 2017). The 5-year survival rate is over 95 % for patients diagnosed at early stage, but decreases to 68 % and 17 % for patients with regional spread and distant metastasis, respectively (Colombo et al., 2016). Historically, EMC is classified into two main clinicopathological types: Type I is the much more common endometrioid adenocarcinoma (80–90 %) and Type II comprises non-endometrioid subtypes such as serous, clear cell and undifferentiated carcinomas, as well as carcinosarcoma/malignant-mixed Müllerian tumor (10–20 %) (Halkia et al., 2012). In China, most of EMC patients are diagnosed with advanced-stage and high-grade tumors that spread beyond the uterus and experience tumor progression within 1 year (He et al., 2018). To date, surgery is the primary option for the early-stage patients and chemotherapy is used to treat advanced and recurrent patients. However, tumor heterogeneity and chemoresistance are major obstacles in EMC therapy. As a result, it is essential to obtain more information that would be helpful in understanding the biological underpinnings of tumor progression to generate promising strategies for the clinical management of EMC.

Post-translational modification participates in essential biological processes, such as signal transduction and cell fate decision. Arginine methylation plays important roles in DNA repair, alternative splicing and RNA metabolism that are involved in the initial and progression in human diseases (Blanc and Richard, 2017). Protein arginine methyltransferases (PRMTs), including nine members, consists of two groups (type I and II). Type I PRMTs (PRMT1, 2, 3, 4, 6, and 8) are responsible for asymmetric dimethylarginine (ADMA), while type II PRMTs (PRMT5 and 7) generates symmetric dimethylarginine (SDMA) (Bedford and Clarke, 2009). Specifically, PRMT6, lying on chromosome 1p13.3, transfers methyl from S-adenosylmethionine to guanidino nitrogen to produce asymmetrical dimethylated arginine (Hamamoto and Nakamura, 2016). PRMT6 is the major PRMT responsible for histone H3R2 methylation and is universally expressed in human normal tissues (Bedford and Clarke, 2009). During neural differentiation, PRMT6 interacts with polycomb repressive complex (PRC) subunits, such as CBX8 and EZH2, to repress the differentiation-associated transcriptional activation of rostral HOXA genes (Stein et al., 2016). In human cancer cells, PRMT6 is differently expressed. Decreased PRMT6 expression has been reported in melanoma (Limm et al., 2013) and hepatocellular carcinoma (Chan et al., 2018), whereas increased expression of PRMT6 is found in lung (Bouchard et al., 2018), breast (Kim et al., 2013), gastric (Okuno et al., 2019) and prostate (Almeida-Rios et al., 2016) cancers. Therefore, the biological function of PRMT6 in human cancers may be contradictory, dependent on the cellular content.

In this study, we intended to examine the expression of PRMT6 and its clinical significance in EMC, and to investigate its role in the progression of EMC. Our data show that PRMT6 is overexpressed and correlates with poor outcomes, and that PRMT6 exhibits oncogenic activity via AKT/mTOR signaling pathway.

Section snippets

Patients

Twenty-eight fresh EMC specimens were collected for determination of mRNA and protein levels of PRMT6 in The First Affiliated Hospital of Sun Yat-sen University (FAHSYSU) and The Affiliated Hexian Memorial Hospital of Southern Medical University. A total of 232 paraffin-embedded EMC cases diagnosed between Jan 2010 to Dec 2012 at FAHSYSU was recruited (known as FAHSYSU cohort) to examine the expression and clinical significance of PRMT6. None of the patients had received radiotherapy or

PRMT6 expression is increased and correlated with poor outcomes in EMC

TCGA data showed that genomic alterations frequently occurred in PRMT members (Supplementary Fig. 1A). Current literatures reported that PRMT6 was differently expressed in human cancers (Supplementary Fig. 1B). In this study, we first intended to investigate the expression of PRMT6 and its clinical significance in EMC. Using qRT-PCR and western blot, the mRNA and protein levels of PRMT6 were determined in 28 pairs of fresh EMC tissues and the corresponding adjacent endometrium tissues

Discussion

EMC represents one of the life risks that cause plenty of death in women (Siegel et al., 2017). Although literatures have unveiled aberrant signaling pathways that contribute to EMC progression, the molecular mechanism of EMC invasion and metastasis remain unclear. In the present study, we present data that PRMT6 expression is upregulated in clinical samples and correlates with unfavorable outcomes. in vitro and in vivo data demonstrate that PRMT6 exerts oncogenic activity to promote cell

Data availability statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Declaration of Competing Interest

All authors declare no conflict of interest.

CRediT authorship contribution statement

Nan Jiang: Conceptualization, Data curation, Writing - review & editing. Qiu-Li Li: Data curation, Methodology, Resources. Wenwei Pan: Data curation, Methodology. Jinhui Li: Data curation. Mei-Fang Zhang: Data curation. Tiefeng Cao: Data curation. Shu-Guang Su: Conceptualization, Supervision, Writing - review & editing. Huimin Shen: Conceptualization, Supervision, Writing - review & editing.

References (34)

  • D. Almeida-Rios et al.

    Histone methyltransferase PRMT6 plays an oncogenic role of in prostate cancer

    Oncotarget

    (2016)
  • F. Bray et al.

    Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

    CA Cancer J. Clin.

    (2018)
  • L.H. Chan et al.

    PRMT6 regulates RAS/RAF binding and MEK/ERK-mediated cancer stemness activities in hepatocellular carcinoma through CRAF methylation

    Cell Rep.

    (2018)
  • X. Chen et al.

    miR-372 regulates glioma cell proliferation and invasion by directly targeting PHLPP2

    J. Cell. Biochem.

    (2015)
  • S. Choi et al.

    Skeletal muscle-specific Prmt1 deletion causes muscle atrophy via deregulation of the PRMT6-FOXO3 axis

    Autophagy

    (2019)
  • S. Choi et al.

    Skeletal muscle-specific Prmt1 deletion causes muscle atrophy via deregulation of the PRMT6-FOXO3 axis

    Autophagy

    (2019)
  • A. Di Lorenzo et al.

    A gain-of-function mouse model identifies PRMT6 as a NF-kappaB coactivator

    Nucleic Acids Res.

    (2014)
  • Cited by (25)

    • Design, synthesis and evaluation of antitumor activity of selective PRMT6 inhibitors

      2023, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Forming a complex with LEF1 to perform unmethylated functions upregulates CCND1 and promotes tumor cell proliferation (Fig. 1) [15]. Studies have shown that the high expression of PRMT6 is closely related to the poor prognosis of various cancers, including lung cancer, breast cancer, colorectal cancer, glioblastoma, endometrial cancer, and prostate cancer [14,16–18]. Therefore, PRMT6 is a promising therapeutic target for cancer.

    • Regulation of the aryl hydrocarbon receptor in cancer and cancer stem cells of gynecological malignancies: An update on signaling pathways

      2022, Seminars in Cancer Biology
      Citation Excerpt :

      It was also reported that chemoresistance of uterine cancer to platinum- and taxane-based chemotherapy is mediated through DNA repair mechanisms, efflux pumps, and survival pathways such as PI3K/AKT pathway and mitogen-activated protein kinase (MAPK) pathway [245]. In addition, several CSC-related signaling pathways were found to be expressed in endometrial CSCs, including, Wnt/β-Catenin [246–248], Notch, and PI3K/AKT [249–252]. In endometrial cancer cells, it has been reported that administration of ITE, an endogenous AhR ligand, inhibited cell proliferation and migration in vitro in AN3CA and HEC-1B cell lines and in xenograft growth in mice, where this effect was associated with a significant increase in CYP1A1 and CYP1B1 mRNA and protein levels [115].

    • The crucial mechanism and therapeutic implication of RNA methylation in bone pathophysiology

      2022, Ageing Research Reviews
      Citation Excerpt :

      Cheng Cheng et al. found that METTL14 increases PTPN6 mRNA stability in an m6A-dependent manner, thereby enhancing cell proliferation and osteogenic differentiation of BMMSCs (Cheng, 2021). Previous studies demonstrated that PRMT6 activates the PI3K/AKT pathway, which plays an important role in regulating osteogenesis of MSCs (Liu, 2007; Jiang, 2020). ALKBH5 could accelerate the degradation of PRMT6 mRNA in an m6A-dependent manner, thereby inhibiting PI3K/AKT pathway and suppressing osteogenesis of BMMSCs (Li, 2021).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text