Akademska digitalna zbirka SLovenije - logo
E-viri
Recenzirano Odprti dostop
  • m6A modification of lncRNA ...
    Lang, Chuandong; Yin, Chi; Lin, Kaiyuan; Li, Yue; Yang, Qing; Wu, Zhengquan; Du, Hong; Ren, Dong; Dai, Yuhu; Peng, Xinsheng

    Clinical and translational medicine, June 2021, Letnik: 11, Številka: 6
    Journal Article

    Background Bone metastasis is the leading cause of tumor‐related death in prostate cancer (PCa) patients. Long noncoding RNAs (lncRNAs) have been well documented to be involved in the progression of multiple cancers. Nevertheless, the role of lncRNAs in PCa bone metastasis remains largely unclear. Methods The expression of prostate cancer‐associated transcripts was analyzed in published datasets and further verified in clinical samples and cell lines by RT‐qPCR and in situ hybridization assays. Colony formation assay, MTT assay, cell cycle analysis, EdU assay, Transwell migration and invasion assays, wound healing assay, and in vivo experiments were carried out to investigate the function of prostate cancer‐associated transcript 6 (PCAT6) in bone metastasis and tumor growth of PCa. Bioinformatic analysis, RNA pull‐down, and RIP assays were conducted to identify the proteins binding to PCAT6 and the potential targets of PCAT6. The therapeutic potential of targeting PCAT6 by antisense oligonucleotides (ASO) was further explored in vivo. Results PCAT6 was upregulated in PCa tissues with bone metastasis and increased PCAT6 expression predicted poor prognosis in PCa patients. Functional experiments found that PCAT6 knockdown significantly inhibited PCa cell invasion, migration, and proliferation in vitro, as well as bone metastasis and tumor growth in vivo. Mechanistically, METTL3‐mediated m6A modification contributed to PCAT6 upregulation in an IGF2BP2‐dependent manner. Furthermore, PCAT6 upregulated IGF1R expression by enhancing IGF1R mRNA stability through the PCAT6/IGF2BP2/IGF1R RNA‐protein three‐dimensional complex. Importantly, PCAT6 inhibition by ASO in vivo showed therapeutic potential against bone metastasis in PCa. Finally, the clinical correlation of METTL3, IGF2BP2, IGF1R, and PCAT6 was further demonstrated in PCa tissues and cells. Conclusions Our study uncovers a novel molecular mechanism by which the m6A‐induced PCAT6/IGF2BP2/IGF1R axis promotes PCa bone metastasis and tumor growth, suggesting that PCAT6 may serve as a promising prognostic marker and therapeutic target against bone‐metastatic PCa. 1. PCAT6 is upregulated in bone metastasis‐positive prostate cancer and PCAT6 upregulation correlates with poor prognosis in patients with prostate cancer. 2. PCAT6 promotes prostate cancer bone metastasis by stabilizing IGF1R mRNA through interacting with IGF2BP2. 3. METTL3‐mediated m6A modification leads to the upregulation of PCAT6 in an IGF2BP2‐dependent manner.