File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: hnRNP C modulates MERS-CoV and SARS-CoV-2 replication by governing the expression of a subset of circRNAs and cognitive mRNAs

TitlehnRNP C modulates MERS-CoV and SARS-CoV-2 replication by governing the expression of a subset of circRNAs and cognitive mRNAs
Authors
KeywordscircRNA
Coronavirus
hnRNP C
mRNA
RNA binding protein
Issue Date2022
Citation
Emerging Microbes and Infections, 2022, v. 11, n. 1, p. 519-531 How to Cite?
AbstractHost circular RNAs (circRNAs) play critical roles in the pathogenesis of viral infections. However, how viruses modulate the biogenesis of host proviral circRNAs to facilitate their replication remains unclear. We have recently shown that Middle East respiratory syndrome coronavirus (MERS-CoV) infection increases co-expression of circRNAs and their cognate messenger RNAs (mRNAs), possibly by hijacking specific host RNA binding proteins (RBPs). In this study, we systemically analysed the interactions between the representative circRNA–mRNA pairs upregulated upon MERS-CoV infection and host RBPs. Our analysis identified heterogeneous nuclear ribonucleoprotein C (hnRNP C) as a key host factor that governed the expression of numerous MERS-CoV-perturbed circRNAs, including hsa_circ_0002846, hsa_circ_0002061, and hsa_circ_0004445. RNA immunoprecipitation assay showed that hnRNP C could bind physically to these circRNAs. Specific knockdown of hnRNP C by small interfering RNA significantly (P < 0.05 to P < 0.0001) suppressed MERS-CoV replication in human lung adenocarcinoma (Calu-3) and human small airway epithelial (HSAEC) cells. Both MERS-CoV and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection increased the total and phosphorylated forms of hnRNP C to activate the downstream CRK-mTOR pathway. Treatment of MERS-CoV- (IC50: 0.618 µM) or SARS-CoV-2-infected (IC50: 1.233 µM) Calu-3 cells with the mTOR inhibitor OSI-027 resulted in significantly reduced viral loads. Collectively, our study identified hnRNP C as a key regulator of MERS-CoV-perturbed circRNAs and their cognate mRNAs, and the potential of targeting hnRNP C-related signalling pathways as an anticoronaviral strategy.
Persistent Identifierhttp://hdl.handle.net/10722/315385
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorZhang, Xi-
dc.contributor.authorChu, Hin-
dc.contributor.authorChik, Kenn Ka Heng-
dc.contributor.authorWen, Lei-
dc.contributor.authorShuai, Huiping-
dc.contributor.authorYang, Dong-
dc.contributor.authorWang, Yixin-
dc.contributor.authorHou, Yuxin-
dc.contributor.authorYuen, Terrence Tsz Tai-
dc.contributor.authorCai, Jian Piao-
dc.contributor.authorYuan, Shuofeng-
dc.contributor.authorYin, Feifei-
dc.contributor.authorYuen, Kwok Yung-
dc.contributor.authorChan, Jasper Fuk Woo-
dc.date.accessioned2022-08-05T10:18:42Z-
dc.date.available2022-08-05T10:18:42Z-
dc.date.issued2022-
dc.identifier.citationEmerging Microbes and Infections, 2022, v. 11, n. 1, p. 519-531-
dc.identifier.urihttp://hdl.handle.net/10722/315385-
dc.description.abstractHost circular RNAs (circRNAs) play critical roles in the pathogenesis of viral infections. However, how viruses modulate the biogenesis of host proviral circRNAs to facilitate their replication remains unclear. We have recently shown that Middle East respiratory syndrome coronavirus (MERS-CoV) infection increases co-expression of circRNAs and their cognate messenger RNAs (mRNAs), possibly by hijacking specific host RNA binding proteins (RBPs). In this study, we systemically analysed the interactions between the representative circRNA–mRNA pairs upregulated upon MERS-CoV infection and host RBPs. Our analysis identified heterogeneous nuclear ribonucleoprotein C (hnRNP C) as a key host factor that governed the expression of numerous MERS-CoV-perturbed circRNAs, including hsa_circ_0002846, hsa_circ_0002061, and hsa_circ_0004445. RNA immunoprecipitation assay showed that hnRNP C could bind physically to these circRNAs. Specific knockdown of hnRNP C by small interfering RNA significantly (P < 0.05 to P < 0.0001) suppressed MERS-CoV replication in human lung adenocarcinoma (Calu-3) and human small airway epithelial (HSAEC) cells. Both MERS-CoV and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection increased the total and phosphorylated forms of hnRNP C to activate the downstream CRK-mTOR pathway. Treatment of MERS-CoV- (IC50: 0.618 µM) or SARS-CoV-2-infected (IC50: 1.233 µM) Calu-3 cells with the mTOR inhibitor OSI-027 resulted in significantly reduced viral loads. Collectively, our study identified hnRNP C as a key regulator of MERS-CoV-perturbed circRNAs and their cognate mRNAs, and the potential of targeting hnRNP C-related signalling pathways as an anticoronaviral strategy.-
dc.languageeng-
dc.relation.ispartofEmerging Microbes and Infections-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectcircRNA-
dc.subjectCoronavirus-
dc.subjecthnRNP C-
dc.subjectmRNA-
dc.subjectRNA binding protein-
dc.titlehnRNP C modulates MERS-CoV and SARS-CoV-2 replication by governing the expression of a subset of circRNAs and cognitive mRNAs-
dc.typeArticle-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1080/22221751.2022.2032372-
dc.identifier.pmid35060842-
dc.identifier.pmcidPMC8843244-
dc.identifier.scopuseid_2-s2.0-85124439491-
dc.identifier.volume11-
dc.identifier.issue1-
dc.identifier.spage519-
dc.identifier.epage531-
dc.identifier.eissn2222-1751-
dc.identifier.isiWOS:000753692800001-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats