SARS-CoV-2 ORF6 Disrupts Bidirectional Nucleocytoplasmic Transport through Interactions with Rae1 and Nup98

SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19), is an RNA virus with a large genome that encodes multiple accessory proteins. While these accessory proteins are not required for growth in vitro, they can contribute to the pathogenicity of the virus. ABSTRACT RNA viruses that replicate in the cytoplasm often disrupt nucleocytoplasmic transport to preferentially translate their own transcripts and prevent host antiviral responses. The Sarbecovirus accessory protein ORF6 has previously been shown to be a major inhibitor of interferon production in both severe acute respiratory syndrome coronavirus (SARS-CoV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we show SARS-CoV-2-infected cells display an elevated level of nuclear mRNA accumulation compared to mock-infected cells. We demonstrate that ORF6 is responsible for this nuclear imprisonment of host mRNA, and using a cotransfected reporter assay, we show this nuclear retention of mRNA blocks expression of newly transcribed mRNAs. ORF6’s nuclear entrapment of host mRNA is associated with its ability to copurify with the mRNA export factors, Rae1 and Nup98. These protein-protein interactions map to the C terminus of ORF6 and can be abolished by a single amino acid mutation in Met58. Overexpression of Rae1 restores reporter expression in the presence of SARS-CoV-2 ORF6. SARS-CoV ORF6 also interacts with Rae1 and Nup98. However, SARS-CoV-2 ORF6 more strongly copurifies with Rae1 and Nup98 and results in significantly reduced expression of reporter proteins compared to SARS-CoV ORF6, a potential mechanism for the delayed symptom onset and presymptomatic transmission uniquely associated with the SARS-CoV-2 pandemic. We also show that both SARS-CoV and SARS-CoV-2 ORF6 block nuclear import of a broad range of host proteins. Together, these data support a model in which ORF6 clogs the nuclear pore through its interactions with Rae1 and Nup98 to prevent both nuclear import and export, rendering host cells incapable of responding to SARS-CoV-2 infection. IMPORTANCE SARS-CoV-2, the causative agent of coronavirus disease 2019 (COVID-19), is an RNA virus with a large genome that encodes multiple accessory proteins. While these accessory proteins are not required for growth in vitro, they can contribute to the pathogenicity of the virus. We demonstrate that SARS-CoV-2-infected cells accumulate poly(A) mRNA in the nucleus, which is attributed to the accessory protein ORF6. Nuclear entrapment of mRNA and reduced expression of newly transcribed reporter proteins are associated with ORF6’s interactions with the mRNA export proteins Rae1 and Nup98. SARS-CoV ORF6 also shows the same interactions with Rae1 and Nup98. However, SARS-CoV-2 ORF6 more strongly represses reporter expression and copurifies with Rae1 and Nup98 compared to SARS-CoV ORF6. Both SARS-CoV ORF6 and SARS-CoV-2 ORF6 block nuclear import of a wide range of host factors through interactions with Rae1 and Nup98. Together, our results suggest ORF6’s disruption of nucleocytoplasmic transport prevents infected cells from responding to the invading virus.

[1]  Priya S. Shah,et al.  Zika Virus Infection Prevents Host mRNA Nuclear Export by Disrupting UPF1 Function , 2020, bioRxiv.

[2]  I. Ulitsky,et al.  SARS-CoV-2 utilizes a multipronged strategy to suppress host protein synthesis , 2020, bioRxiv.

[3]  S. Chanda,et al.  SARS-CoV-2 Orf6 hijacks Nup98 to block STAT nuclear import and antagonize interferon signaling , 2020, Proceedings of the National Academy of Sciences.

[4]  A. Addetia,et al.  Sensitive Recovery of Complete SARS-CoV-2 Genomes from Clinical Samples by Use of Swift Biosciences’ SARS-CoV-2 Multiplex Amplicon Sequencing Panel , 2020, Journal of Clinical Microbiology.

[5]  S. Nakagawa,et al.  Sarbecovirus ORF6 Proteins Hamper the Induction of Interferon Signaling by Blocking mRNA Nuclear Export , 2020, Social Science Research Network.

[6]  Vineet D. Menachery,et al.  Evasion of Type I Interferon by SARS-CoV-2 , 2020, Cell Reports.

[7]  Trevor Bedford,et al.  Cryptic transmission of SARS-CoV-2 in Washington state , 2020, Science.

[8]  P. Shi,et al.  In vivo antiviral host transcriptional response to SARS-CoV-2 by viral load, sex, and age , 2020, PLoS biology.

[9]  L. Ren,et al.  Activation and evasion of type I interferon responses by SARS-CoV-2 , 2020, Nature Communications.

[10]  Gavin J. D. Smith,et al.  Discovery and Genomic Characterization of a 382-Nucleotide Deletion in ORF7b and ORF8 during the Early Evolution of SARS-CoV-2 , 2020, mBio.

[11]  Edward C. Holmes,et al.  A dynamic nomenclature proposal for SARS-CoV-2 lineages to assist genomic epidemiology , 2020, Nature Microbiology.

[12]  Francesco Castelli,et al.  Comparing SARS-CoV-2 with SARS-CoV and influenza pandemics , 2020, The Lancet Infectious Diseases.

[13]  Malik Peiris,et al.  A Rare Deletion in SARS-CoV-2 ORF6 Dramatically Alters the Predicted Three-Dimensional Structure of the Resultant Protein , 2020, bioRxiv.

[14]  A. Addetia,et al.  Identification of multiple large deletions in ORF7a resulting in in-frame gene fusions in clinical SARS-CoV-2 isolates , 2020, Journal of Clinical Virology.

[15]  Trevor Bedford,et al.  Genomic surveillance reveals multiple introductions of SARS-CoV-2 into Northern California , 2020, Science.

[16]  Isaac I. Bogoch,et al.  Coast-to-Coast Spread of SARS-CoV-2 during the Early Epidemic in the United States , 2020, Cell.

[17]  M. Scotch,et al.  An 81-Nucleotide Deletion in SARS-CoV-2 ORF7a Identified from Sentinel Surveillance in Arizona (January to March 2020) , 2020, Journal of Virology.

[18]  Benjamin J. Polacco,et al.  A SARS-CoV-2 Protein Interaction Map Reveals Targets for Drug-Repurposing , 2020, Nature.

[19]  W. Wei,et al.  Presymptomatic Transmission of SARS-CoV-2 — Singapore, January 23–March 16, 2020 , 2020, MMWR. Morbidity and mortality weekly report.

[20]  J. Duchin,et al.  Detection of SARS-CoV-2 Among Residents and Staff Members of an Independent and Assisted Living Community for Older Adults — Seattle, Washington, 2020 , 2020, MMWR. Morbidity and mortality weekly report.

[21]  Trevor Bedford,et al.  Cryptic transmission of SARS-CoV-2 in Washington state , 2020, Science.

[22]  Yan Bai,et al.  Presumed Asymptomatic Carrier Transmission of COVID-19. , 2020, JAMA.

[23]  G. Gao,et al.  A Novel Coronavirus from Patients with Pneumonia in China, 2019 , 2020, The New England journal of medicine.

[24]  K. To,et al.  SARS-CoV-2 nsp13, nsp14, nsp15 and orf6 function as potent interferon antagonists , 2020, Emerging microbes & infections.

[25]  M. Mathews,et al.  Translational Control in Virus-Infected Cells. , 2018, Cold Spring Harbor perspectives in biology.

[26]  David K. Smith,et al.  ggtree: an r package for visualization and annotation of phylogenetic trees with their covariates and other associated data , 2017 .

[27]  Yong Hoon Kim,et al.  A Herpesvirus Protein Selectively Inhibits Cellular mRNA Nuclear Export. , 2016, Cell host & microbe.

[28]  G. Blobel,et al.  Vesiculoviral matrix (M) protein occupies nucleic acid binding site at nucleoporin pair (Rae1•Nup98) , 2014, Proceedings of the National Academy of Sciences.

[29]  Björn Usadel,et al.  Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..

[30]  Alexandros Stamatakis,et al.  RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies , 2014, Bioinform..

[31]  K. Katoh,et al.  MAFFT Multiple Sequence Alignment Software Version 7: Improvements in Performance and Usability , 2013, Molecular biology and evolution.

[32]  J. Chou,et al.  Complexes of Vesicular Stomatitis Virus Matrix Protein with Host Rae1 and Nup98 Involved in Inhibition of Host Transcription , 2012, PLoS pathogens.

[33]  Shane S. Sturrock,et al.  Geneious Basic: An integrated and extendable desktop software platform for the organization and analysis of sequence data , 2012, Bioinform..

[34]  A. Mirazimi,et al.  A putative diacidic motif in the SARS-CoV ORF6 protein influences its subcellular localization and suppression of expression of co-transfected expression constructs , 2011, BMC Research Notes.

[35]  D. Walsh,et al.  Viral subversion of the host protein synthesis machinery , 2011, Nature Reviews Microbiology.

[36]  G. Blobel,et al.  Structural and functional analysis of the interaction between the nucleoporin Nup98 and the mRNA export factor Rae1 , 2010, Proceedings of the National Academy of Sciences.

[37]  C. Gomez-Sanchez,et al.  Nuclear Import of the Glucocorticoid Receptor-hsp90 Complex through the Nuclear Pore Complex Is Mediated by Its Interaction with Nup62 and Importin β , 2009, Molecular and Cellular Biology.

[38]  Jincun Zhao,et al.  Severe Acute Respiratory Syndrome Coronavirus Protein 6 Is Required for Optimal Replication , 2008, Journal of Virology.

[39]  S. Perlman,et al.  Severe Acute Respiratory Syndrome Coronavirus Protein 6 Accelerates Murine Hepatitis Virus Infections by More than One Mechanism , 2008, Journal of Virology.

[40]  Krishna Shankara Narayanan,et al.  SARS coronavirus accessory proteins , 2007, Virus Research.

[41]  Ralph S. Baric,et al.  Severe Acute Respiratory Syndrome Coronavirus ORF6 Antagonizes STAT1 Function by Sequestering Nuclear Import Factors on the Rough Endoplasmic Reticulum/Golgi Membrane , 2007, Journal of Virology.

[42]  P. Palese,et al.  Severe Acute Respiratory Syndrome Coronavirus Open Reading Frame (ORF) 3b, ORF 6, and Nucleocapsid Proteins Function as Interferon Antagonists , 2006, Journal of Virology.

[43]  Arul Earnest,et al.  Asymptomatic SARS Coronavirus Infection among Healthcare Workers, Singapore , 2005, Emerging infectious diseases.

[44]  C. Arana,et al.  VSV disrupts the Rae1/mrnp41 mRNA nuclear export pathway. , 2005, Molecular cell.

[45]  O. Tsang,et al.  Asymptomatic Severe Acute Respiratory Syndrome–associated Coronavirus Infection , 2003, Emerging infectious diseases.

[46]  H. Atkins,et al.  VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. , 2003, Cancer cell.

[47]  D. Lyles,et al.  Ability of the Matrix Protein of Vesicular Stomatitis Virus To Suppress Beta Interferon Gene Expression Is Genetically Correlated with the Inhibition of Host RNA and Protein Synthesis , 2003, Journal of Virology.

[48]  M. Katze,et al.  Translational Control of Viral Gene Expression in Eukaryotes , 2000, Microbiology and Molecular Biology Reviews.

[49]  M. Fornerod,et al.  RAE1 Is a Shuttling mRNA Export Factor That Binds to a GLEBS-like NUP98 Motif at the Nuclear Pore Complex through Multiple Domains , 1999, The Journal of cell biology.