Inactivating Three Interferon Antagonists Attenuates Pathogenesis of an Enteric Coronavirus

Emerging coronaviruses, including SARS-CoV-2 and porcine CoVs, can infect enterocytes, cause diarrhea, and be shed in the feces. New approaches are needed to understand enteric pathogenesis and to develop vaccines and therapeutics to prevent the spread of these viruses. Here, we exploited a reverse genetic system for an enteric CoV, porcine epidemic diarrhea virus (PEDV), and outline an approach of genetically inactivating highly conserved viral factors known to limit the host innate immune response to infection. Our report reveals that generating PEDV with inactive versions of three viral interferon antagonists, nonstructural proteins 1, 15, and 16, results in a highly attenuated virus that does not cause diarrhea in animals and elicits a neutralizing antibody response in virus-infected animals. This strategy may be useful for generating live attenuated vaccine candidates that prevent disease and fecal spread of enteric CoVs, including SARS-CoV-2. ABSTRACT Coronaviruses (CoVs) have repeatedly emerged from wildlife hosts and infected humans and livestock animals to cause epidemics with significant morbidity and mortality. CoVs infect various organs, including respiratory and enteric systems, as exemplified by newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The constellation of viral factors that contribute to developing enteric disease remains elusive. Here, we investigated CoV interferon antagonists for their contribution to enteric pathogenesis. Using an infectious clone of an enteric CoV, porcine epidemic diarrhea virus (icPEDV), we generated viruses with inactive versions of interferon antagonist nonstructural protein 1 (nsp1), nsp15, and nsp16 individually or combined into one virus designated icPEDV-mut4. Interferon-responsive PK1 cells were infected with these viruses and produced higher levels of interferon responses than were seen with wild-type icPEDV infection. icPEDV-mut4 elicited robust interferon responses and was severely impaired for replication in PK1 cells. To evaluate viral pathogenesis, piglets were infected with either icPEDV or icPEDV-mut4. While the icPEDV-infected piglets exhibited clinical disease, the icPEDV-mut4-infected piglets showed no clinical symptoms and exhibited normal intestinal pathology at day 2 postinfection. icPEDV-mut4 replicated in the intestinal tract, as revealed by detection of viral RNA in fecal swabs, with sequence analysis documenting genetic stability of the input strain. Importantly, icPEDV-mut4 infection elicited IgG and neutralizing antibody responses to PEDV. These results identify nsp1, nsp15, and nsp16 as virulence factors that contribute to the development of PEDV-induced diarrhea in swine. Inactivation of these CoV interferon antagonists is a rational approach for generating candidate vaccines to prevent disease and spread of enteric CoVs, including SARS-CoV-2. IMPORTANCE Emerging coronaviruses, including SARS-CoV-2 and porcine CoVs, can infect enterocytes, cause diarrhea, and be shed in the feces. New approaches are needed to understand enteric pathogenesis and to develop vaccines and therapeutics to prevent the spread of these viruses. Here, we exploited a reverse genetic system for an enteric CoV, porcine epidemic diarrhea virus (PEDV), and outline an approach of genetically inactivating highly conserved viral factors known to limit the host innate immune response to infection. Our report reveals that generating PEDV with inactive versions of three viral interferon antagonists, nonstructural proteins 1, 15, and 16, results in a highly attenuated virus that does not cause diarrhea in animals and elicits a neutralizing antibody response in virus-infected animals. This strategy may be useful for generating live attenuated vaccine candidates that prevent disease and fecal spread of enteric CoVs, including SARS-CoV-2.

[1]  O. Tsang,et al.  Infection of bat and human intestinal organoids by SARS-CoV-2 , 2020, Nature Medicine.

[2]  Hans Clevers,et al.  SARS-CoV-2 productively infects human gut enterocytes , 2020, Science.

[3]  John O. Konz,et al.  Engineering the Live-Attenuated Polio Vaccine to Prevent Reversion to Virulence , 2020, Cell host & microbe.

[4]  A. Tam,et al.  Gastrointestinal Manifestations of SARS-CoV-2 Infection and Virus Load in Fecal Samples From a Hong Kong Cohort: Systematic Review and Meta-analysis , 2020, Gastroenterology.

[5]  P. Vollmar,et al.  Virological assessment of hospitalized patients with COVID-2019 , 2020, Nature.

[6]  E. Holmes,et al.  Author Correction: A new coronavirus associated with human respiratory disease in China , 2020, Nature.

[7]  S. Baker,et al.  Coronavirus endoribonuclease targets viral polyuridine sequences to evade activating host sensors , 2020, Proceedings of the National Academy of Sciences.

[8]  S. Baker,et al.  Coronavirus Endoribonuclease and Deubiquitinating Interferon Antagonists Differentially Modulate the Host Response during Replication in Macrophages , 2020, Journal of Virology.

[9]  Huiying Liang,et al.  Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding , 2020, Nature Medicine.

[10]  H. Shan,et al.  Evidence for Gastrointestinal Infection of SARS-CoV-2 , 2020, Gastroenterology.

[11]  Yan Zhao,et al.  Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. , 2020, JAMA.

[12]  E. Holmes,et al.  A new coronavirus associated with human respiratory disease in China , 2020, Nature.

[13]  Kai Zhao,et al.  A pneumonia outbreak associated with a new coronavirus of probable bat origin , 2020, Nature.

[14]  S. Lindstrom,et al.  First Case of 2019 Novel Coronavirus in the United States , 2020, The New England journal of medicine.

[15]  Y. Hu,et al.  Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China , 2020, The Lancet.

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

[17]  David K. Meyerholz,et al.  IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. , 2019, The Journal of clinical investigation.

[18]  A. Vlasova,et al.  Engineering a Live Attenuated Porcine Epidemic Diarrhea Virus Vaccine Candidate via Inactivation of the Viral 2'-O-Methyltransferase and the Endocytosis Signal of the Spike Protein , 2019, Journal of Virology.

[19]  A. Ploss,et al.  Decoding type I and III interferon signalling during viral infection , 2019, Nature Microbiology.

[20]  K. Lager,et al.  Coronavirus Endoribonuclease Activity in Porcine Epidemic Diarrhea Virus Suppresses Type I and Type III Interferon Responses , 2019, Journal of Virology.

[21]  R. Baric,et al.  Evaluation of a recombination-resistant coronavirus as a broadly applicable, rapidly implementable vaccine platform , 2018, Communications Biology.

[22]  Lisa E. Gralinski,et al.  Combination Attenuation Offers Strategy for Live Attenuated Coronavirus Vaccines , 2018, Journal of Virology.

[23]  R. Sun,et al.  Genome-wide identification of interferon-sensitive mutations enables influenza vaccine design , 2018, Science.

[24]  T. Fujita,et al.  Type III Interferon Restriction by Porcine Epidemic Diarrhea Virus and the Role of Viral Protein nsp1 in IRF1 Signaling , 2017, Journal of Virology.

[25]  Lisa E. Gralinski,et al.  Middle East Respiratory Syndrome Coronavirus Nonstructural Protein 16 Is Necessary for Interferon Resistance and Viral Pathogenesis , 2017, mSphere.

[26]  Lisa E. Gralinski,et al.  MERS-CoV Accessory ORFs Play Key Role for Infection and Pathogenesis , 2017, mBio.

[27]  D. Yoo,et al.  Inhibition of NF-κB activity by the porcine epidemic diarrhea virus nonstructural protein 1 for innate immune evasion , 2017, Virology.

[28]  C. Kao,et al.  Coronavirus nonstructural protein 15 mediates evasion of dsRNA sensors and limits apoptosis in macrophages , 2017, Proceedings of the National Academy of Sciences.

[29]  David K. Meyerholz,et al.  Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice , 2016, Cell Host & Microbe.

[30]  K. Lager,et al.  Evaluation of two real-time polymerase chain reaction assays for Porcine epidemic diarrhea virus (PEDV) to assess PEDV transmission in growing pigs , 2016, Journal of veterinary diagnostic investigation : official publication of the American Association of Veterinary Laboratory Diagnosticians, Inc.

[31]  Joseph T. Thomas,et al.  Effect of porcine epidemic diarrhea virus infectious doses on infection outcomes in naïve conventional neonatal and weaned pigs , 2018 .

[32]  A. García-Sastre,et al.  Interference of viral effector proteins with chromatin, transcription, and the epigenome. , 2015, Current opinion in microbiology.

[33]  Jane Christopher-Hennings,et al.  Development of an indirect ELISA, blocking ELISA, fluorescent microsphere immunoassay and fluorescent focus neutralization assay for serologic evaluation of exposure to North American strains of Porcine Epidemic Diarrhea Virus , 2015, BMC Veterinary Research.

[34]  L. Saif,et al.  Comparative pathogenesis of US porcine epidemic diarrhea virus (PEDV) strain PC21A in conventional 9-day-old nursing piglets vs. 26-day-old weaned pigs , 2015, Veterinary Microbiology.

[35]  Krishna Shankara Narayanan,et al.  Coronavirus nonstructural protein 1: Common and distinct functions in the regulation of host and viral gene expression , 2014, Virus Research.

[36]  V. Thiel,et al.  To sense or not to sense viral RNA — essentials of coronavirus innate immune evasion , 2014, Current Opinion in Microbiology.

[37]  C. Maluquer de Motes,et al.  Vaccinia virus immune evasion: mechanisms, virulence and immunogenicity. , 2013, The Journal of general virology.

[38]  Lisa E. Gralinski,et al.  A Double-Inactivated Severe Acute Respiratory Syndrome Coronavirus Vaccine Provides Incomplete Protection in Mice and Induces Increased Eosinophilic Proinflammatory Pulmonary Response upon Challenge , 2011, Journal of Virology.

[39]  Burkhard Ludewig,et al.  Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5 , 2011, Nature Immunology.

[40]  Abraham J Koster,et al.  SARS-Coronavirus Replication Is Supported by a Reticulovesicular Network of Modified Endoplasmic Reticulum , 2008, PLoS biology.

[41]  R. L. Roper,et al.  Unique SARS-CoV protein nsp1: bioinformatics, biochemistry and potential effects on virulence , 2007, Trends in Microbiology.

[42]  A. Joachimiak,et al.  New Antiviral Target Revealed by the Hexameric Structure of Mouse Hepatitis Virus Nonstructural Protein nsp15 , 2006, Journal of Virology.

[43]  Didier Nurizzo,et al.  Crystal structure and mechanistic determinants of SARS coronavirus nonstructural protein 15 define an endoribonuclease family , 2006, Proceedings of the National Academy of Sciences.

[44]  K. Bhardwaj,et al.  The Severe Acute Respiratory Syndrome Coronavirus Nsp15 Protein Is an Endoribonuclease That Prefers Manganese as a Cofactor , 2004, Journal of Virology.

[45]  J. Ziebuhr,et al.  Major genetic marker of nidoviruses encodes a replicative endoribonuclease. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[46]  Y. Guan,et al.  Unique and Conserved Features of Genome and Proteome of SARS-coronavirus, an Early Split-off From the Coronavirus Group 2 Lineage , 2003, Journal of Molecular Biology.

[47]  K. Bienz,et al.  RNA Replication of Mouse Hepatitis Virus Takes Place at Double-Membrane Vesicles , 2002, Journal of Virology.

[48]  L. Reed,et al.  A SIMPLE METHOD OF ESTIMATING FIFTY PER CENT ENDPOINTS , 1938 .

[49]  Geoffrey L. Smith,et al.  How Does Vaccinia Virus Interfere With Interferon? , 2018, Advances in virus research.

[50]  J. Ziebuhr The Coronavirus Replicase , 2005, Current topics in microbiology and immunology.