TY - JOUR AU - Jiang, Fengjie AU - Tang, Xiaozhu AU - Tang, Chao AU - Hua, Zhen AU - Ke, Mengying AU - Wang, Chen AU - Zhao, Jiamin AU - Gao, Shengyao AU - Jurczyszyn, Artur AU - Janz, Siegfried AU - Beksac, Meral AU - Zhan, Fenghuang AU - Gu, Chunyan AU - Yang, Ye PY - 2021 DA - 2021/04/01 TI - HNRNPA2B1 promotes multiple myeloma progression by increasing AKT3 expression via m6A-dependent stabilization of ILF3 mRNA JO - Journal of Hematology & Oncology SP - 54 VL - 14 IS - 1 AB - N6-methyladenosine (m6A) modification is the most prevalent modification in eukaryotic RNAs while accumulating studies suggest that m6A aberrant expression plays an important role in cancer. HNRNPA2B1 is a m6A reader which binds to nascent RNA and thus affects a perplexing array of RNA metabolism exquisitely. Despite unveiled facets that HNRNPA2B1 is deregulated in several tumors and facilitates tumor growth, a clear role of HNRNPA2B1 in multiple myeloma (MM) remains elusive. Herein, we analyzed the function and the regulatory mechanism of HNRNPA2B1 in MM. We found that HNRNPA2B1 was elevated in MM patients and negatively correlated with favorable prognosis. The depletion of HNRNPA2B1 in MM cells inhibited cell proliferation and induced apoptosis. On the contrary, the overexpression of HNRNPA2B1 promoted cell proliferation in vitro and in vivo. Mechanistic studies revealed that HNRNPA2B1 recognized the m6A sites of ILF3 and enhanced the stability of ILF3 mRNA transcripts, while AKT3 downregulation by siRNA abrogated the cellular proliferation induced by HNRNPA2B1 overexpression. Additionally, the expression of HNRNPA2B1, ILF3 and AKT3 was positively associated with each other in MM tissues tested by immunohistochemistry. In summary, our study highlights that HNRNPA2B1 potentially acts as a therapeutic target of MM through regulating AKT3 expression mediated by ILF3-dependent pattern. SN - 1756-8722 UR - https://doi.org/10.1186/s13045-021-01066-6 DO - 10.1186/s13045-021-01066-6 ID - Jiang2021 ER -