LncRNA AWPPH promotes osteosarcoma progression via activation of Wnt/β-catenin pathway through modulating miR-93-3p/FZD7 axis

https://doi.org/10.1016/j.bbrc.2019.04.203Get rights and content

Highlights

  • AWPPH up-regulation is associated with poor prognosis in OS.

  • AWPPH sponges miR-93-3p in OS.

  • AWPPH promotes OS progression through suppressing miR-93-3p.

  • FZD7 up-regulation by AWPPH-caused miR-93-3p suppression induces activation of Wnt/β-catenin signaling.

Abstract

Long noncoding RNAs (lncRNAs) have important regulatory roles in osteosarcoma (OS) progression. Recent researches have shown lncRNA AWPPH promotes lung cancer progression and bladder cancer development. Yet, the function of AWPPH in OS is unknown. In this research, results indicated AWPPH levels were increased in OS tissues in contrast to paracancerous controls. Up-regulated AWPPH was associated with advanced stage, tumor size and metastasis. Besides, AWPPH up-regulation indicated a low survival rate in OS patients. Silencing of AWPPH suppressed proliferation, migration and invasion of OS cells. Mechanistically, AWPPH was demonstrated to sponge miR-93-3p and promote FZD7 expression, causing activation of Wnt/β-catenin. Inhibition of miR-93-3p effectively reversed the effects of AWPPH knockdown on OS cells. Collectively, our findings suggested AWPPH may be a prognostic biomarker and potential therapeutic target. AWPPH enhances FZD7-mediated activation of Wnt/β-catenin by sponging miR-93-3p to promote OS progression.

Introduction

Osteosarcoma (OS) is an aggressive bone cancer and usually occurs among adolescents [1]. High rate of metastasis contributes to rapid progression of OS and leads to large numbers of deaths [2]. The 5-year survival rate in OS patients is quite low [3]. To date, no effective strategy is developed for radical cure of OS. Especially, the mechanism of OS development remains obscure. Today, there is an urgent need to investigate novel targets for OS therapy and improve patients’ outcomes.

Long noncoding RNA (lncRNA) has been defined as a new member of noncoding RNA as miRNA [4]. LncRNA is featured by a length of over 200 nucleotides and limited protein-coding potential [5]. Growing studies show lncRNAs play crucial regulatory functions in various pathological processes, including cancer [6]. Reports have indicated that cell-cycle, metastasis, differentiation or stemness of tumor cells is regulated by lncRNAs [7,8]. For example, Guo et al. showed that lncRNA AB073614 regulates growth and apoptosis in cervical cancer [9]. Sun et al. reported that SNHG12 promotes proliferation and invasiveness in ovarian cancer by up-regulating SOX4 [10]. Wang et al. found that lncRNA IUR regulates tumorigenesis by modulating STAT5-CD71 signaling [11]. Additionally, Huang et al. proved that lncRNA ADPGK-AS1 promotes gastric cancer development and may be a therapeutic target [12]. Thus, functional lncRNAs may be important for OS prognosis and potential therapeutic target.

AWPPH was first reported to regulate hepatocellular carcinoma progression [13]. Recent studies also indicated that AWPPH participates in regulation of bladder cancer, breast cancer and lung cancer [[14], [15], [16]]. Nevertheless, its function in OS remains obscure. In our study, we found AWPPH expression was upregulated in OS tissues. AWPPH level was correlated with advanced stage, tumor size and metastasis. Moreover, AWPPH is a prognostic biomarker of OS patients. AWPPH knockdown caused inhibition on proliferation, migration and invasion. Mechanistically, AWPPH could sponge miR-93-3p to upregulate FZD7 and trigger activation of Wnt/β-catenin signaling. Collectively, our data provides a novel insight on the function and mechanism of AWPPH in OS development.

Section snippets

Human samples

OS tissues and normal tissues were collected from our hospital and stored in liquid nitrogen after surgery. No patient was treated by chemotherapy or radiotherapy before surgery. Singed inform content was obtained from each involved patient. This study was approved by the Ethic Committee of our hospital.

Cell culture and transfection

OS cell lines and normal cell line hFOB 1.19 were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). Cells were cultured with DMEM medium containing 10% FBS (Gibco,

AWPPH up-regulation is associated with poor prognosis in OS

To determine potential roles of AWPPH in OS, its levels were analyzed by qRT-PCR in collected OS tissues and corresponding normal controls. AWPPH expression was up-regulated in OS tissues (Fig. 1A). Notably, AWPPH levels were higher in OS tissues with advanced stages (Fig. 1B) and metastasis (Fig. 1C). We further analyzed the correlation between AWPPH and clinical features. As shown in Table 1, AWPPH expression was positively correlated with tumor size, advance stage and metastasis. There was

Discussion

Elucidating the molecular mechanism of OS development is still urgently required. In this study, we showed AWPPH was up-regulated in OS tissues and correlated with tumor size, advanced stage and metastasis. Moreover, AWPPH high expression indicated a low survival rate. Knockdown of AWPPH inhibited proliferation, migration and invasion of OS cells. We demonstrated AWPPH activated Wnt/β-catenin pathway through modulating miR-93-3p/FZD7 axis. Thus, our findings revealed a novel mechanism

Conflicts of interest

None.

Acknowledgements

None.

References (31)

  • L. Mirabello et al.

    International osteosarcoma incidence patterns in children and adolescents, middle ages and elderly persons

    Int. J. Cancer

    (2009)
  • J.J. Quinn et al.

    Unique features of long non-coding RNA biogenesis and function

    Nat. Rev. Genet.

    (2016)
  • Y. Wang et al.

    Long non-coding RNA DSCR8 acts as a molecular sponge for miR-485-5p to activate Wnt/beta-catenin signal pathway in hepatocellular carcinoma

    Cell Death Dis.

    (2018)
  • L.Y. Guo et al.

    LncRNA AB073614 promotes the proliferation and inhibits apoptosis of cervical cancer cells by repressing RBM5

    Eur. Rev. Med. Pharmacol. Sci.

    (2019)
  • D. Sun et al.

    LncRNA SNHG12 accelerates the progression of ovarian cancer via absorbing miRNA-129 to upregulate SOX4

    Eur. Rev. Med. Pharmacol. Sci.

    (2019)
  • Cited by (28)

    • Long noncoding RNA LINC00978 acts as a potential diagnostic biomarker in patients with colorectal cancer

      2021, Experimental and Molecular Pathology
      Citation Excerpt :

      Li et al. showed that LINC00978 promotes the Wnt/β–catenin signaling through modulating miR-93-3p/FZD7 axis. Moreover, they demonstrated that LINC00978 increased invasion, migration and proliferation in osteosarcoma cell lines (Li et al., 2019). Researchers found that LINC00978 increased invasion and migration in glioma and lung cancer by promoting TGF-β signaling (Dai et al., 2019; Huo et al., 2019).

    • The critical roles of lncRNAs in the development of osteosarcoma

      2021, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      This lncRNA acts as a ceRNA and inhibit miR-93-3p. Down-regulation of miR-93-3p enhances protein levels of FZD7, thus increases the activity of Wnt/β-catenin [35]. In addition, expression of SNHG1 is up-regulated in this cancer.

    • LncRNA PCAT6 promotes tumor progression in osteosarcoma via activation of TGF-β pathway by sponging miR-185-5p

      2020, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Osteosarcoma (OS), one of the most common primary bone tumors around world, usually occurs among children and young adults [1]. The high rate of OS cells metastasizing to distinct organs causes the rapid progression of OS and contributes to the majority of cancer-associated death [2]. In the last decades, great progress has been made in the aspects of treatments for OS [3].

    • Potential regulatory role of lncRNA-miRNA-mRNA axis in osteosarcoma

      2020, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      These lncRNAs, known as competing endogenous RNAs (ceRNAs), served as ‘sponges’ for miRNAs, inhibited the corresponding miRNAs and contributed to enhanced translations of their target mRNAs [23]. The development of osteosarcoma was significantly related with OS cell proliferation, apoptosis and cell cycle, tumor growth, EMT, migration, invasion, metastasis and drug resistance through various signaling pathways, such as the Notch signaling pathway, phosphoinositide 3-kinase (PI3K)/AKT pathway, c-Jun Nterminal kinase (JNK) and Wnt pathways [24–28]. The Notch signaling pathway had been acknowledged to take a great part in cell proliferation, differentiation, migration, invasion, metastasis and survival, and the aberrant activation of Notch signaling was involved in various cancers, including osteosarcoma [24].

    • Emerging roles of long non-coding RNAs in osteosarcoma

      2024, Frontiers in Molecular Biosciences
    View all citing articles on Scopus
    1

    Co-first authors.

    View full text