Porcine reproductive and respiratory syndrome virus degrades DDX10 via SQSTM1/p62-dependent selective autophagy to antagonize its antiviral activity

Porcine reproductive and respiratory syndrome virus (PRRSV) is a typical immunosuppressive virus that has been devastating the global swine industry for over three decades. DEAD-box helicases (DDXs) are a family of ATP-dependent RNA helicases that are predominantly implicated in modulating cellular RNA metabolism. Meanwhile, a growing number of studies have suggested that some DDXs are associated with innate immunity and virus infection, so they are considered potential antiviral targets. Herein, we screened 40 DDXs and found that ectopic expression of DDX10 exhibited a significant anti-PRRSV effect, while DDX10 knockdown promoted PRRSV proliferation. Further analysis revealed that DDX10 positively regulates type I interferon production, which may contribute to its anti-PRRSV effect. Interestingly, PRRSV infection promoted DDX10 translocation from the nucleus to the cytoplasm for macroautophagic/autophagic degradation to block the antiviral effect of DDX10. By screening PRRSV-encoded proteins, we found that the viral envelope (E) protein interacted with DDX10. In line with the autophagic degradation of DDX10 during PRRSV infection, E protein could induce autophagy and reduce DDX10 expression in wild-type cells, but not in ATG5 or ATG7 knockout (KO) cells. When further screening the cargo receptors for autophagic degradation, we found that SQSTM1/p62 (sequestosome 1) interacted with both DDX10 and E protein, and E protein-mediated DDX10 degradation was almost entirely blocked in SQSTM1 KO cells, demonstrating that E protein degrades DDX10 by promoting SQSTM1-mediated selective autophagy. Our study reveals a novel mechanism by which PRRSV escapes host antiviral innate immunity through selective autophagy, providing a new target for developing anti-PRRSV drugs.

[1]  Hongbin He,et al.  The ORF7a protein of SARS-CoV-2 initiates autophagy and limits autophagosome-lysosome fusion via degradation of SNAP29 to promote virus replication , 2022, Autophagy.

[2]  Yulan Jin,et al.  TRAF6 autophagic degradation by avibirnavirus VP3 inhibits antiviral innate immunity via blocking NFKB/NF-κB activation , 2022, Autophagy.

[3]  S. Xiao,et al.  DEAD-Box RNA Helicase 21 (DDX21) Positively Regulates the Replication of Porcine Reproductive and Respiratory Syndrome Virus via Multiple Mechanisms , 2022, Viruses.

[4]  Chao Sui,et al.  SARS-CoV-2 NSP13 Inhibits Type I IFN Production by Degradation of TBK1 via p62-Dependent Selective Autophagy. , 2022, Journal of immunology.

[5]  Hualan Chen,et al.  The PB1 protein of influenza A virus inhibits the innate immune response by targeting MAVS for NBR1-mediated selective autophagic degradation , 2021, PLoS pathogens.

[6]  R. Zhao,et al.  DEAH-Box RNA Helicases in Pre-mRNA Splicing. , 2020, Trends in biochemical sciences.

[7]  Yun Zhang,et al.  The DDX23 Negatively Regulates Translation and Replication of Foot-and-Mouth Disease Virus and Is Degraded by 3C Proteinase , 2020, Viruses.

[8]  Jie Zan,et al.  RNA helicase DDX5 suppresses IFN-I antiviral innate immune response by interacting with PP2A-Cβ to deactivate IRF3. , 2020, Experimental cell research.

[9]  H. Nauwynck,et al.  Porcine reproductive and respiratory syndrome virus Nsp4 cleaves ZAP to antagonize its antiviral activity. , 2020, Veterinary microbiology.

[10]  Shengliang Cao,et al.  The tail domain of PRRSV NSP2 plays a key role in aggrephagy by interacting with 14-3-3ε , 2020, Veterinary Research.

[11]  J. Cui,et al.  HFE inhibits type I IFNs signaling by targeting the SQSTM1-mediated MAVS autophagic degradation , 2020, Autophagy.

[12]  Yingfang Liu,et al.  A novel selective autophagy receptor, CCDC50, delivers K63 polyubiquitination-activated RIG-I/MDA5 for degradation during viral infection , 2020, Cell Research.

[13]  J. Cui,et al.  Selective autophagy controls the stability of transcription factor IRF3 to balance type I interferon production and immune suppression , 2020, Autophagy.

[14]  J. Zimmerman,et al.  Effects of PRRSV Infection on the Porcine Thymus. , 2019, Trends in microbiology.

[15]  Jun Han,et al.  Reprogramming the unfolded protein response for replication by porcine reproductive and respiratory syndrome virus , 2019, PLoS pathogens.

[16]  Yanhong Zhang,et al.  RNF34 functions in immunity and selective mitophagy by targeting MAVS for autophagic degradation , 2019, The EMBO journal.

[17]  F. Gao,et al.  Nucleocapsid protein of porcine reproductive and respiratory syndrome virus antagonizes the antiviral activity of TRIM25 by interfering with TRIM25-mediated RIG-I ubiquitination , 2019, Veterinary Microbiology.

[18]  Huibin Yu,et al.  DDX19 Inhibits Type I Interferon Production by Disrupting TBK1-IKKε-IRF3 Interactions and Promoting TBK1 and IKKε Degradation. , 2019, Cell reports.

[19]  S. Xiao,et al.  Porcine Reproductive and Respiratory Syndrome Virus Nonstructural Protein 4 Cleaves Porcine DCP1a To Attenuate Its Antiviral Activity , 2018, The Journal of Immunology.

[20]  L. Hwang,et al.  Stimulation of the Internal Ribosome Entry Site (IRES)-Dependent Translation of Enterovirus 71 by DDX3X RNA Helicase and Viral 2A and 3C Proteases , 2018, Front. Microbiol..

[21]  J. Cui,et al.  LRRC25 inhibits type I IFN signaling by targeting ISG15‐associated RIG‐I for autophagic degradation , 2018, The EMBO journal.

[22]  Trushar R. Patel,et al.  DEAD-box helicases: the Yin and Yang roles in viral infections , 2018, Biotechnology & genetic engineering reviews.

[23]  J. Ou,et al.  Hepatitis C Virus-Induced Autophagy and Host Innate Immune Response , 2017, Viruses.

[24]  Lei Zhou,et al.  Cellular DEAD-box RNA helicase 18 (DDX18) Promotes the PRRSV Replication via Interaction with Virus nsp2 and nsp10. , 2017, Virus research.

[25]  Jun Han,et al.  Pathogenesis and control of the Chinese highly pathogenic porcine reproductive and respiratory syndrome virus. , 2017, Veterinary microbiology.

[26]  Lei Xu,et al.  Induction of Apoptosis by the Nonstructural Protein 4 and 10 of Porcine Reproductive and Respiratory Syndrome Virus , 2016, PloS one.

[27]  Shujun Zhang,et al.  Interplay of autophagy and apoptosis during PRRSV infection of Marc145 cell. , 2016, Infection, genetics and evolution : journal of molecular epidemiology and evolutionary genetics in infectious diseases.

[28]  Shujun Zhang,et al.  Autophagy postpones apoptotic cell death in PRRSV infection through Bad-Beclin1 interaction , 2016, Virulence.

[29]  Xuehui Cai,et al.  Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus Infection Induced Apoptosis and Autophagy in Thymi of Infected Piglets , 2015, PloS one.

[30]  M. Okabe,et al.  DDX60 Is Involved in RIG-I-Dependent and Independent Antiviral Responses, and Its Function Is Attenuated by Virus-Induced EGFR Activation. , 2015, Cell reports.

[31]  Jianfeng Dai,et al.  DEAD-box RNA helicase DDX3X inhibits DENV replication via regulating type one interferon pathway. , 2015, Biochemical and biophysical research communications.

[32]  Xiaolong Wang,et al.  The DEAD-box RNA helicase 5 positively regulates the replication of porcine reproductive and respiratory syndrome virus by interacting with viral Nsp9 in vitro , 2014, Virus Research.

[33]  D. Yoo,et al.  Engineering the PRRS virus genome: Updates and perspectives , 2014, Veterinary Microbiology.

[34]  M. Veit,et al.  Membrane proteins of arterivirus particles: Structure, topology, processing and function , 2014, Virus Research.

[35]  A. Ernst,et al.  Cargo recognition and trafficking in selective autophagy , 2014, Nature Cell Biology.

[36]  S. Xiao,et al.  Molecular cloning, functional characterization and antiviral activity of porcine DDX3X. , 2014, Biochemical and biophysical research communications.

[37]  L. Enjuanes,et al.  A novel porcine reproductive and respiratory syndrome virus vector system that stably expresses enhanced green fluorescent protein as a separate transcription unit , 2013, Veterinary Research.

[38]  M. Kikkert,et al.  Arterivirus molecular biology and pathogenesis. , 2013, The Journal of general virology.

[39]  Hongbo Hu,et al.  Involvement of unfolded protein response, p53 and Akt in modulation of porcine reproductive and respiratory syndrome virus-mediated JNK activation. , 2013, Virology.

[40]  Cheng Song,et al.  Degradation of CREB-binding protein and modulation of type I interferon induction by the zinc finger motif of the porcine reproductive and respiratory syndrome virus nsp1α subunit. , 2013, Virus research.

[41]  G. Cheng,et al.  DDX41 recognizes bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response , 2012, Nature Immunology.

[42]  Hongbo Hu,et al.  Autophagy sustains the replication of porcine reproductive and respiratory virus in host cells , 2012, Virology.

[43]  E. Snijder,et al.  Identification of porcine reproductive and respiratory syndrome virus ORF1a-encoded non-structural proteins in virus-infected cells. , 2012, The Journal of general virology.

[44]  S. Xiao,et al.  Induction of autophagy enhances porcine reproductive and respiratory syndrome virus replication , 2011, Virus Research.

[45]  Yong‐jun Liu,et al.  The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells , 2011, Nature Immunology.

[46]  Yong-tang Zheng,et al.  Zinc-finger antiviral protein inhibits HIV-1 infection by selectively targeting multiply spliced viral mRNAs for degradation , 2011, Proceedings of the National Academy of Sciences.

[47]  M. Gale,et al.  Immune signaling by RIG-I-like receptors. , 2011, Immunity.

[48]  M. Murtaugh,et al.  Novel structural protein in porcine reproductive and respiratory syndrome virus encoded by an alternative ORF5 present in all arteriviruses , 2011, The Journal of general virology.

[49]  E. Jankowsky,et al.  SF1 and SF2 helicases: family matters. , 2010, Current opinion in structural biology.

[50]  Cheng Song,et al.  Modulation of type I interferon induction by porcine reproductive and respiratory syndrome virus and degradation of CREB-binding protein by non-structural protein 1 in MARC-145 and HeLa cells , 2010, Virology.

[51]  A. Bauch,et al.  The DEAD-box helicase DDX3X is a critical component of the TANK-binding kinase 1-dependent innate immune response , 2008, The EMBO journal.

[52]  G. Gao,et al.  p72 DEAD box RNA helicase is required for optimal function of the zinc-finger antiviral protein , 2008, Proceedings of the National Academy of Sciences.

[53]  Shannon L. Taylor,et al.  Ovarian Tumor Domain-Containing Viral Proteases Evade Ubiquitin- and ISG15-Dependent Innate Immune Responses , 2007, Cell Host & Microbe.

[54]  R. Mitchell,et al.  Recombinant swine beta interferon protects swine alveolar macrophages and MARC-145 cells from infection with Porcine reproductive and respiratory syndrome virus. , 2007, The Journal of general virology.

[55]  Shizuo Akira,et al.  Innate immune recognition of viral infection , 2006, Nature Immunology.

[56]  Terje Johansen,et al.  p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death , 2005, The Journal of cell biology.

[57]  Ralf Bartenschlager,et al.  Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus , 2005, Nature.

[58]  P. Rottier,et al.  Significance of the oligosaccharides of the porcine reproductive and respiratory syndrome virus glycoproteins GP2a and GP5 for infectious virus production. , 2004, The Journal of general virology.

[59]  F. Collins,et al.  A human gene (DDX10) encoding a putative DEAD-box RNA helicase at 11q22-q23. , 1996, Genomics.