SARS-CoV-2 main protease Nsp5 cleaves and inactivates human tRNA methyltransferase TRMT1.

IF 5.3 2区 生物学 Q2 CELL BIOLOGY Journal of Molecular Cell Biology Pub Date : 2023-08-03 DOI:10.1093/jmcb/mjad024
Jia-Li Lu, Xiao-Long Zhou
{"title":"SARS-CoV-2 main protease Nsp5 cleaves and inactivates human tRNA methyltransferase TRMT1.","authors":"Jia-Li Lu, Xiao-Long Zhou","doi":"10.1093/jmcb/mjad024","DOIUrl":null,"url":null,"abstract":"is crucial for tRNA structure and function. TRMT1 is localized in the nucleus, cytoplasm, and mitochondria. Its mitochondrial localization is mediated by an N-terminal mitochondrial targeting sequence ( MTS ) of ∼35 residues ( Dewe et al., 2017 ) . However, it is not known whether TRMT1 is a bone-fide substrate of Nsp5. Moreover, the consequence of the potential cleavage remains unclear. Here, we expressed and purified a recombinant SARS-CoV-2 Nsp5 with a C-terminal His 6 tag and an MTS-deleted TRMT1 with an N-terminal FLAG and a C-terminal His 6 tag ( TRMT1 35 ) using Escherichia coli ( Figure 1 A; Supplementary Figure S1 A and B ) . Western blot analysis using anti-FLAG antibodies showed that the amount of TRMT1 35 decreased after incubation with Nsp5 in a dose-dependent manner, accompanied by the appearance of a digested product of ∼58 kDa ( Figure 1 B ) , suggesting that the cleavage site is in the latter part of TRMT1. However, the C-terminal cleavage product with a lower molecular weight was not observed using anti-His 6 antibodies. Coomassie blue staining using the same protein samples and digestion conditions also showed a clear cleaved fragment after incubation with Nsp5 ( Supplementary Figure S1 C ) . To confirm the digestion of TRMT1 by SARS-CoV-2 Nsp5 in vivo , we co-expressed TRMT1 35 ( with N-terminal HA and C-terminal FLAG tags ) and 3 × Myc-tagged Nsp5 ( Figure 1 A ) in HEK293T cells. Indeed, western blot analysis using anti-HA antibodies showed the generation of a digested product of ∼58 kDa, while the cleaved fragment with the lower molecular mass was likewise invisible ( Figure 1 C ) . Considering the high sequence identity ( ∼96% ) between SARS-CoV-2 Nsp5 and SARS-CoV Nsp5, we further purified SARS-CoV Nsp5 ( Supplementary Figure S1 D ) . Consistently, SARS-CoV Nsp5 was also able to digest TRMT1 35 in vitro with a high efficiency ( Figure 1 D ) , indicating that TRMT1 cleavage is a conserved mechanism among different CoVs. To better understand whether TRMT1 digestion is mediated by the protease activity of Nsp5, the C145 residue of SARS-CoV-2 Nsp5 was replaced by alanine, resulting in a C145A mutant that was defective in cleavage ( Figure 1 E ) . The C145A mutant is a well-known variant that was constructed to show the contribution of the C145–H41 catalytic dyad to substrate cleavage ( Jin et al., 2020 ) . The C145A mutant exhibits the same three-dimensional structure as the wild-type enzyme ( Hsu et al., 2005 ) . These data clearly demonstrated that Nsp5 is able to cleave TRMT1 in vitro and in vivo via its protease activity. Subsequently, we intended to identify the cleavage site in TRMT1. Nsp5 digests all protein substrates at conserved glutamine residues. NetCorona 1.0, a website predicting Nsp5 cleavage sites ( Kiemer et al., 2004 ) , showed that Q530 was the only confident candidate with a high score of 0.942, while other glutamine residues ( such as Q584 ) were excluded ( Supplementary Figure S2 A ) . This prediction was roughly consistent with the molecular mass of the digested product in both in vitro ( Figure 1 B ) and in vivo ( Figure 1 C ) assays. To experimentally validate the predicted cleavage site, we mutated Q530 to alanine in the TRMT1 35 eukaryotic expression construct. In parallel, a Q584A mutant was also constructed for comparison. Immunoblotting with anti-HA antibodies showed that the cleavage of TRMT1 35 Q530A was abolished ( Figure 1 F ) . According Dear Editor, Coronavirus disease 2019 ( COVID-19 ) , caused by severe acute respiratory syndrome coronavirus 2 ( SARS-CoV-2 ) , has become a serious emergency to global public health since its outbreak in 2019. SARS-CoV-2 is an enveloped, positive-sense, single-strand RNA virus, belonging to the Betacoronavirus genus of the Coronaviridae family ( Wu et al., 2020 ) including SARS-CoV, SARS-CoV-2, MERS-CoV, etc. After viral entry, the RNA genome first encodes 16 nonstructural proteins, including papain-like protease and Nsp5 ( also called the main protease ) , which are essential for viral replication and transcription ( Bai et al., 2022 ) . Nsp5 is a 33-kDa cysteine protease whose most frequently found recognition sequence is LQˆ ( S/A/G ) . Many protein–protein interactions between the virus and the host have been identified ( Gordon et al., 2020a ) . Only a small number of interactions between host proteins and the viral protein Nsp5 have been experimentally validated ( Wenzel et al., 2021 ) . Few reports have linked the role of host protein cleavage by Nsp5 beyond immune evasion. An interaction between catalytically inactive Nsp5 mutant ( C145A ) and tRNA methyltransferase 1 ( TRMT1 ) has been described previously ( Gordon et al., 2020a , b ) . TRMT1 ( 659 amino acids ) catalyzes the formation of N 2 -methylguanosine ( m 2 G ) or N 2 , N 2 -dimethylguanosine ( m 2,2 G ) at position 26 of most human tRNAs, which","PeriodicalId":16433,"journal":{"name":"Journal of Molecular Cell Biology","volume":null,"pages":null},"PeriodicalIF":5.3000,"publicationDate":"2023-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/30/85/mjad024.PMC10399916.pdf","citationCount":"2","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Molecular Cell Biology","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1093/jmcb/mjad024","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 2

Abstract

is crucial for tRNA structure and function. TRMT1 is localized in the nucleus, cytoplasm, and mitochondria. Its mitochondrial localization is mediated by an N-terminal mitochondrial targeting sequence ( MTS ) of ∼35 residues ( Dewe et al., 2017 ) . However, it is not known whether TRMT1 is a bone-fide substrate of Nsp5. Moreover, the consequence of the potential cleavage remains unclear. Here, we expressed and purified a recombinant SARS-CoV-2 Nsp5 with a C-terminal His 6 tag and an MTS-deleted TRMT1 with an N-terminal FLAG and a C-terminal His 6 tag ( TRMT1 35 ) using Escherichia coli ( Figure 1 A; Supplementary Figure S1 A and B ) . Western blot analysis using anti-FLAG antibodies showed that the amount of TRMT1 35 decreased after incubation with Nsp5 in a dose-dependent manner, accompanied by the appearance of a digested product of ∼58 kDa ( Figure 1 B ) , suggesting that the cleavage site is in the latter part of TRMT1. However, the C-terminal cleavage product with a lower molecular weight was not observed using anti-His 6 antibodies. Coomassie blue staining using the same protein samples and digestion conditions also showed a clear cleaved fragment after incubation with Nsp5 ( Supplementary Figure S1 C ) . To confirm the digestion of TRMT1 by SARS-CoV-2 Nsp5 in vivo , we co-expressed TRMT1 35 ( with N-terminal HA and C-terminal FLAG tags ) and 3 × Myc-tagged Nsp5 ( Figure 1 A ) in HEK293T cells. Indeed, western blot analysis using anti-HA antibodies showed the generation of a digested product of ∼58 kDa, while the cleaved fragment with the lower molecular mass was likewise invisible ( Figure 1 C ) . Considering the high sequence identity ( ∼96% ) between SARS-CoV-2 Nsp5 and SARS-CoV Nsp5, we further purified SARS-CoV Nsp5 ( Supplementary Figure S1 D ) . Consistently, SARS-CoV Nsp5 was also able to digest TRMT1 35 in vitro with a high efficiency ( Figure 1 D ) , indicating that TRMT1 cleavage is a conserved mechanism among different CoVs. To better understand whether TRMT1 digestion is mediated by the protease activity of Nsp5, the C145 residue of SARS-CoV-2 Nsp5 was replaced by alanine, resulting in a C145A mutant that was defective in cleavage ( Figure 1 E ) . The C145A mutant is a well-known variant that was constructed to show the contribution of the C145–H41 catalytic dyad to substrate cleavage ( Jin et al., 2020 ) . The C145A mutant exhibits the same three-dimensional structure as the wild-type enzyme ( Hsu et al., 2005 ) . These data clearly demonstrated that Nsp5 is able to cleave TRMT1 in vitro and in vivo via its protease activity. Subsequently, we intended to identify the cleavage site in TRMT1. Nsp5 digests all protein substrates at conserved glutamine residues. NetCorona 1.0, a website predicting Nsp5 cleavage sites ( Kiemer et al., 2004 ) , showed that Q530 was the only confident candidate with a high score of 0.942, while other glutamine residues ( such as Q584 ) were excluded ( Supplementary Figure S2 A ) . This prediction was roughly consistent with the molecular mass of the digested product in both in vitro ( Figure 1 B ) and in vivo ( Figure 1 C ) assays. To experimentally validate the predicted cleavage site, we mutated Q530 to alanine in the TRMT1 35 eukaryotic expression construct. In parallel, a Q584A mutant was also constructed for comparison. Immunoblotting with anti-HA antibodies showed that the cleavage of TRMT1 35 Q530A was abolished ( Figure 1 F ) . According Dear Editor, Coronavirus disease 2019 ( COVID-19 ) , caused by severe acute respiratory syndrome coronavirus 2 ( SARS-CoV-2 ) , has become a serious emergency to global public health since its outbreak in 2019. SARS-CoV-2 is an enveloped, positive-sense, single-strand RNA virus, belonging to the Betacoronavirus genus of the Coronaviridae family ( Wu et al., 2020 ) including SARS-CoV, SARS-CoV-2, MERS-CoV, etc. After viral entry, the RNA genome first encodes 16 nonstructural proteins, including papain-like protease and Nsp5 ( also called the main protease ) , which are essential for viral replication and transcription ( Bai et al., 2022 ) . Nsp5 is a 33-kDa cysteine protease whose most frequently found recognition sequence is LQˆ ( S/A/G ) . Many protein–protein interactions between the virus and the host have been identified ( Gordon et al., 2020a ) . Only a small number of interactions between host proteins and the viral protein Nsp5 have been experimentally validated ( Wenzel et al., 2021 ) . Few reports have linked the role of host protein cleavage by Nsp5 beyond immune evasion. An interaction between catalytically inactive Nsp5 mutant ( C145A ) and tRNA methyltransferase 1 ( TRMT1 ) has been described previously ( Gordon et al., 2020a , b ) . TRMT1 ( 659 amino acids ) catalyzes the formation of N 2 -methylguanosine ( m 2 G ) or N 2 , N 2 -dimethylguanosine ( m 2,2 G ) at position 26 of most human tRNAs, which

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
严重急性呼吸系统综合征冠状病毒2型主要蛋白酶Nsp5切割并失活人tRNA甲基转移酶TRMT1。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
9.60
自引率
1.80%
发文量
1383
期刊介绍: The Journal of Molecular Cell Biology ( JMCB ) is a full open access, peer-reviewed online journal interested in inter-disciplinary studies at the cross-sections between molecular and cell biology as well as other disciplines of life sciences. The broad scope of JMCB reflects the merging of these life science disciplines such as stem cell research, signaling, genetics, epigenetics, genomics, development, immunology, cancer biology, molecular pathogenesis, neuroscience, and systems biology. The journal will publish primary research papers with findings of unusual significance and broad scientific interest. Review articles, letters and commentary on timely issues are also welcome. JMCB features an outstanding Editorial Board, which will serve as scientific advisors to the journal and provide strategic guidance for the development of the journal. By selecting only the best papers for publication, JMCB will provide a first rate publishing forum for scientists all over the world.
期刊最新文献
Probing centromere-kinetochore core complex CENP-L/M assembly using cenpemlin. Correction to: Mitochondrial aldehyde dehydrogenase rescues against diabetic cardiomyopathy through GSK3β-mediated preservation of mitochondrial integrity and Parkin-mediated mitophagy. PHLDA2 is critical for p53-mediated ferroptosis and tumor suppression. Regulation of m6Am RNA modification and its implications in human diseases. Comments on 'Adeno-to-squamous transition drives resistance to KRAS inhibition in LKB1 mutant lung cancer'.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1