Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2021

Open Access 01-12-2021 | Multiple Myeloma | Letter to the Editor

HNRNPA2B1 promotes multiple myeloma progression by increasing AKT3 expression via m6A-dependent stabilization of ILF3 mRNA

Authors: Fengjie Jiang, Xiaozhu Tang, Chao Tang, Zhen Hua, Mengying Ke, Chen Wang, Jiamin Zhao, Shengyao Gao, Artur Jurczyszyn, Siegfried Janz, Meral Beksac, Fenghuang Zhan, Chunyan Gu, Ye Yang

Published in: Journal of Hematology & Oncology | Issue 1/2021

Login to get access

Abstract

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation. Cell. 2017;169(7):1187–200.CrossRef Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation. Cell. 2017;169(7):1187–200.CrossRef
2.
go back to reference Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7(12):885–7.CrossRef Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7(12):885–7.CrossRef
3.
go back to reference Deng X, Su R, Feng X, Wei M, Chen J. Role of N(6)-methyladenosine modification in cancer. Curr Opin Genet Dev. 2018;48:1–7.CrossRef Deng X, Su R, Feng X, Wei M, Chen J. Role of N(6)-methyladenosine modification in cancer. Curr Opin Genet Dev. 2018;48:1–7.CrossRef
4.
go back to reference Alarcon CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 2015;162(6):1299–308.CrossRef Alarcon CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 2015;162(6):1299–308.CrossRef
5.
go back to reference Tauler J, Zudaire E, Liu H, Shih J, Mulshine JL. hnRNP A2/B1 modulates epithelial-mesenchymal transition in lung cancer cell lines. Cancer Res. 2010;70(18):7137–47.CrossRef Tauler J, Zudaire E, Liu H, Shih J, Mulshine JL. hnRNP A2/B1 modulates epithelial-mesenchymal transition in lung cancer cell lines. Cancer Res. 2010;70(18):7137–47.CrossRef
6.
go back to reference Han N, Li W, Zhang M. The function of the RNA-binding protein hnRNP in cancer metastasis. J Cancer Res Ther. 2013;9(Suppl):S129-134.PubMed Han N, Li W, Zhang M. The function of the RNA-binding protein hnRNP in cancer metastasis. J Cancer Res Ther. 2013;9(Suppl):S129-134.PubMed
7.
go back to reference Soung NK, Kim HM, Asami Y, et al. Mechanism of the natural product moracin-O derived MO-460 and its targeting protein hnRNPA2B1 on HIF-1alpha inhibition. Exp Mol Med. 2019;51(2):1–14.CrossRef Soung NK, Kim HM, Asami Y, et al. Mechanism of the natural product moracin-O derived MO-460 and its targeting protein hnRNPA2B1 on HIF-1alpha inhibition. Exp Mol Med. 2019;51(2):1–14.CrossRef
8.
go back to reference Moran-Jones K, Grindlay J, Jones M, Smith R, Norman JC. hnRNP A2 regulates alternative mRNA splicing of TP53INP2 to control invasive cell migration. Cancer Res. 2009;69(24):9219–27.CrossRef Moran-Jones K, Grindlay J, Jones M, Smith R, Norman JC. hnRNP A2 regulates alternative mRNA splicing of TP53INP2 to control invasive cell migration. Cancer Res. 2009;69(24):9219–27.CrossRef
9.
go back to reference Kwon S, Barbarese E, Carson JH. The cis-acting RNA trafficking signal from myelin basic protein mRNA and its cognate trans-acting ligand hnRNP A2 enhance cap-dependent translation. J Cell Biol. 1999;147(2):247–56.CrossRef Kwon S, Barbarese E, Carson JH. The cis-acting RNA trafficking signal from myelin basic protein mRNA and its cognate trans-acting ligand hnRNP A2 enhance cap-dependent translation. J Cell Biol. 1999;147(2):247–56.CrossRef
10.
go back to reference Griffin ME, Hamilton BJ, Roy KM, et al. Post-transcriptional regulation of glucose transporter-1 by an AU-rich element in the 3’UTR and by hnRNP A2. Biochem Biophys Res Commun. 2004;318(4):977–82.CrossRef Griffin ME, Hamilton BJ, Roy KM, et al. Post-transcriptional regulation of glucose transporter-1 by an AU-rich element in the 3’UTR and by hnRNP A2. Biochem Biophys Res Commun. 2004;318(4):977–82.CrossRef
11.
go back to reference Fu Y, Dominissini D, Rechavi G, He C. Gene expression regulation mediated through reversible m(6)A RNA methylation. Nat Rev Genet. 2014;15(5):293–306.CrossRef Fu Y, Dominissini D, Rechavi G, He C. Gene expression regulation mediated through reversible m(6)A RNA methylation. Nat Rev Genet. 2014;15(5):293–306.CrossRef
Metadata
Title
HNRNPA2B1 promotes multiple myeloma progression by increasing AKT3 expression via m6A-dependent stabilization of ILF3 mRNA
Authors
Fengjie Jiang
Xiaozhu Tang
Chao Tang
Zhen Hua
Mengying Ke
Chen Wang
Jiamin Zhao
Shengyao Gao
Artur Jurczyszyn
Siegfried Janz
Meral Beksac
Fenghuang Zhan
Chunyan Gu
Ye Yang
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2021
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-021-01066-6

Other articles of this Issue 1/2021

Journal of Hematology & Oncology 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine