LncRNA SPRY4‑IT1 promotes progression of osteosarcoma by regulating ZEB1 and ZEB2 expression through sponging of miR‑101 activity

Int J Oncol. 2020 Jan;56(1):85-100. doi: 10.3892/ijo.2019.4910. Epub 2019 Nov 13.

Abstract

Long non‑coding (lnc)RNA sprouty receptor tyrosine kinase signalling antagonist 4‑intronic transcript 1 (SPRY4‑IT1) has been demonstrated to serve a critical role in the tumorigenesis of osteosarcoma (OS); however, the specific underlying mechanism remains unclear. The aim of the present study was to examine the interactions between SPRY4‑IT1 and its downstream effectors, to determine if any of the interactions contributed to SPRY4‑IT1‑mediated proliferation, migration and invasion in cancer cells. A signalling cascade which involved SPRY4‑IT1, miR‑101 and zinc finger E‑box‑binding homeoboxes (ZEBs) was examined in the present study. Intracellular SPRY4‑IT1 and miR‑101 expression levels were altered through transfection to assess their effect on proliferation, cell cycle progression, survival, migration and invasion. A dual‑luciferase assay was utilized to determine the association between SPRY4‑IT1/miR‑101 and ZEBs/miR‑101 and nude mouse xenograft experiments were performed to determine the effect of SPRY4‑IT1 in vivo. The results indicated that the SPRY4‑IT1 levels were negatively associated with miR‑101 expression levels in OS cells, an association which was not observed in the normal osteoblast cells. SPRY4‑IT1 knockdown or miR‑101 overexpression reduced proliferation, cell cycle progression, survival, migration and invasion of MG‑63 and U2OS cells. SPRY4‑IT1 knockdown was accompanied by increased expression of miR‑101 and E‑cadherin levels, as well as decreased expression levels of ZEB1/2 and other epithelial‑mesenchymal transition‑associated proteins. Simultaneous knockdown of SPRY4‑IT1 and inhibition of miR‑101 partially reversed the anti‑tumour effects of SPRY4‑IT1 inhibition in vitro. Consistent with these findings, short hairpin RNA targeting SPRY4‑IT1 also hindered xenograft tumour growth and altered the levels of miR‑101, ZEB1/2 and E‑cadherin in vivo. Dual‑luciferase reporter assays demonstrated that SPRY4‑IT1 may have regulated the expression of ZEB1 and ZEB2 by sponging miR‑101. In conclusion, SPRY4‑IT1 inhibition increased miR‑101 levels, resulting in downregulation of ZEB1/2 expression and thus exerting anti‑tumour effects in OS.

MeSH terms

  • Animals
  • Apoptosis
  • Biomarkers, Tumor / genetics
  • Biomarkers, Tumor / metabolism*
  • Bone Neoplasms / genetics
  • Bone Neoplasms / metabolism
  • Bone Neoplasms / pathology*
  • Cell Proliferation
  • Disease Progression
  • Epithelial-Mesenchymal Transition
  • Gene Expression Regulation, Neoplastic
  • Humans
  • Mice
  • Mice, Inbred BALB C
  • MicroRNAs / genetics*
  • Osteosarcoma / genetics
  • Osteosarcoma / metabolism
  • Osteosarcoma / pathology*
  • RNA, Long Noncoding / genetics*
  • Signal Transduction
  • Tumor Cells, Cultured
  • Xenograft Model Antitumor Assays
  • Zinc Finger E-box Binding Homeobox 2 / genetics
  • Zinc Finger E-box Binding Homeobox 2 / metabolism*
  • Zinc Finger E-box-Binding Homeobox 1 / genetics
  • Zinc Finger E-box-Binding Homeobox 1 / metabolism*

Substances

  • Biomarkers, Tumor
  • MIRN101 microRNA, human
  • MicroRNAs
  • RNA, Long Noncoding
  • ZEB1 protein, human
  • ZEB2 protein, human
  • Zinc Finger E-box Binding Homeobox 2
  • Zinc Finger E-box-Binding Homeobox 1
  • long noncoding RNA SPRY4-IT1, human