SARS-CoV-2 infection in nonhuman primates alters the composition and functional activity of the gut microbiota

ABSTRACT The current pandemic of coronavirus disease (COVID) 2019 constitutes a global public health issue. Regarding the emerging importance of the gut-lung axis in viral respiratory infections, analysis of the gut microbiota’s composition and functional activity during a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection might be instrumental in understanding and controling COVID 19. We used a nonhuman primate model (the macaque), that recapitulates mild COVID-19 symptoms, to analyze the effects of a SARS-CoV-2 infection on dynamic changes of the gut microbiota. 16S rRNA gene profiling and analysis of β diversity indicated significant changes in the composition of the gut microbiota with a peak at 10–13 days post-infection (dpi). Analysis of bacterial abundance correlation networks confirmed disruption of the bacterial community at 10–13 dpi. Some alterations in microbiota persisted after the resolution of the infection until day 26. Some changes in the relative bacterial taxon abundance associated with infectious parameters. Interestingly, the relative abundance of Acinetobacter (Proteobacteria) and some genera of the Ruminococcaceae family (Firmicutes) was positively correlated with the presence of SARS-CoV-2 in the upper respiratory tract. Targeted quantitative metabolomics indicated a drop in short-chain fatty acids (SCFAs) and changes in several bile acids and tryptophan metabolites in infected animals. The relative abundance of several taxa known to be SCFA producers (mostly from the Ruminococcaceae family) was negatively correlated with systemic inflammatory markers while the opposite correlation was seen with several members of the genus Streptococcus. Collectively, SARS-CoV-2 infection in a nonhuman primate is associated with changes in the gut microbiota’s composition and functional activity.

[1]  K. Chow,et al.  Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19 , 2021, Gut.

[2]  Steven M. Holland,et al.  Autoantibodies against type I IFNs in patients with life-threatening COVID-19 , 2020, Science.

[3]  Jacques Fellay,et al.  Inborn errors of type I IFN immunity in patients with life-threatening COVID-19 , 2020, Science.

[4]  Lisa E. Gralinski,et al.  Animal models for COVID-19 , 2020, Nature.

[5]  P. Scherer,et al.  Obesity and diabetes as comorbidities for COVID-19: Underlying mechanisms and the role of viral–bacterial interactions , 2020, eLife.

[6]  X. de Lamballerie,et al.  Hydroxychloroquine use against SARS-CoV-2 infection in non-human primates , 2020, Nature.

[7]  Zigui Chen,et al.  Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19 , 2020, Gut.

[8]  Angelo Carfì,et al.  Persistent Symptoms in Patients After Acute COVID-19. , 2020, JAMA.

[9]  Chuan Qin,et al.  Primary exposure to SARS-CoV-2 protects against reinfection in rhesus macaques , 2020, Science.

[10]  H. Sokol,et al.  Potential Causes and Consequences of Gastrointestinal Disorders during a SARS-CoV-2 Infection , 2020, Cell Reports.

[11]  S. Ng,et al.  Alterations in Fecal Fungal Microbiome of Patients With COVID-19 During Time of Hospitalization until Discharge , 2020, Gastroenterology.

[12]  Y. Shah,et al.  Oxygen battle in the gut: Hypoxia and hypoxia-inducible factors in metabolic and inflammatory responses in the intestine , 2020, The Journal of Biological Chemistry.

[13]  Lanjuan Li,et al.  Alterations of the Gut Microbiota in Patients with COVID-19 or H1N1 Influenza , 2020, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[14]  P. Sorger,et al.  SARS-CoV-2 infection protects against rechallenge in rhesus macaques , 2020, Science.

[15]  J. Vaupel,et al.  National age and coresidence patterns shape COVID-19 vulnerability , 2020, Proceedings of the National Academy of Sciences.

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

[17]  S. Ng,et al.  Manifestations and prognosis of gastrointestinal and liver involvement in patients with COVID-19: a systematic review and meta-analysis , 2020, The Lancet Gastroenterology & Hepatology.

[18]  E. Fischer,et al.  Respiratory disease in rhesus macaques inoculated with SARS-CoV-2 , 2020, Nature.

[19]  Chen Liu,et al.  Influenza infection elicits an expansion of gut population of endogenous Bifidobacterium animalis which protects mice against infection , 2020, Genome Biology.

[20]  Fengzhe Xu,et al.  Gut microbiota may underlie the predisposition of healthy individuals to COVID-19 , 2020, medRxiv.

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

[22]  T. Kuiken,et al.  Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model , 2020, Science.

[23]  Y. Yazdanpanah,et al.  Characterization and Treatment of SARS-CoV-2 in Nasal and Bronchial Human Airway Epithelia , 2020, bioRxiv.

[24]  Xavier Duval,et al.  Clinical and virological data of the first cases of COVID-19 in Europe: a case series , 2020, The Lancet Infectious Diseases.

[25]  P. Adab,et al.  Covid-19: risk factors for severe disease and death , 2020, BMJ.

[26]  Ke Ma,et al.  Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study , 2020, BMJ.

[27]  Rui Ji,et al.  Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysis , 2020, International Journal of Infectious Diseases.

[28]  Hong Jiang,et al.  Immunodepletion with Hypoxemia: A Potential High Risk Subtype of Coronavirus Disease 2019 , 2020, medRxiv.

[29]  Qiurong Ruan,et al.  Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China , 2020, Intensive Care Medicine.

[30]  G. Milligan,et al.  Gut Dysbiosis during Influenza Contributes to Pulmonary Pneumococcal Superinfection through Altered Short-Chain Fatty Acid Production. , 2020, Cell reports.

[31]  J. Xiang,et al.  Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study , 2020, The Lancet.

[32]  K. Yuen,et al.  Clinical Characteristics of Coronavirus Disease 2019 in China , 2020, The New England journal of medicine.

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

[34]  H. Sokol,et al.  Gut microbiota-derived metabolites as key actors in inflammatory bowel disease , 2020, Nature Reviews Gastroenterology & Hepatology.

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

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

[37]  S. Devkota,et al.  The gut microbiome and metabolic syndrome. , 2019, The Journal of clinical investigation.

[38]  William A. Walters,et al.  Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2 , 2019, Nature Biotechnology.

[39]  H. Sokol,et al.  Validation of a global quantitative analysis methodology of tryptophan metabolites in mice using LC-MS. , 2019, Talanta.

[40]  B. Helmink,et al.  The microbiome, cancer, and cancer therapy , 2019, Nature Medicine.

[41]  Jun Yu,et al.  Diversity of macaque microbiota compared to the human counterparts , 2018, Scientific Reports.

[42]  T. R. Licht,et al.  Microbial tryptophan catabolites in health and disease , 2018, Nature Communications.

[43]  Lei Yu,et al.  Metagenomic comparison of the rectal microbiota between rhesus macaques (Macaca mulatta) and cynomolgus macaques (Macaca fascicularis) , 2018, Zoological research.

[44]  Harry Sokol,et al.  Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. , 2018, Cell host & microbe.

[45]  Weston R. Whitaker,et al.  Gut Microbiota-Produced Tryptamine Activates an Epithelial G-Protein-Coupled Receptor to Increase Colonic Secretion. , 2018, Cell host & microbe.

[46]  M. Moffatt,et al.  Respiratory Disease following Viral Lung Infection Alters the Murine Gut Microbiota , 2018, Front. Immunol..

[47]  A. Molinaro,et al.  Role of Bile Acids in Metabolic Control , 2018, Trends in Endocrinology & Metabolism.

[48]  M. Schmolke,et al.  Influenza A virus infection impacts systemic microbiota dynamics and causes quantitative enteric dysbiosis , 2018, Microbiome.

[49]  G. Weinstock,et al.  Impact of Age, Caloric Restriction, and Influenza Infection on Mouse Gut Microbiome: An Exploratory Study of the Role of Age-Related Microbiome Changes on Influenza Responses , 2017, Front. Immunol..

[50]  D. Artis,et al.  Regulation of inflammation by microbiota interactions with the host , 2017, Nature Immunology.

[51]  C. Mackay,et al.  Metabolite-Sensing G Protein-Coupled Receptors-Facilitators of Diet-Related Immune Regulation. , 2017, Annual review of immunology.

[52]  R. Xavier,et al.  CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands , 2016, Nature Medicine.

[53]  G. Cheng,et al.  Influenza Virus Affects Intestinal Microbiota and Secondary Salmonella Infection in the Gut through Type I Interferons , 2016, PLoS pathogens.

[54]  S. Ehrlich,et al.  Influence of H7N9 virus infection and associated treatment on human gut microbiota , 2015, Scientific Reports.

[55]  R. Sun,et al.  Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell–dependent inflammation , 2014, The Journal of experimental medicine.

[56]  S. Zeisel,et al.  A targeted metabolomic protocol for short-chain fatty acids and branched-chain amino acids , 2013, Metabolomics.

[57]  Susan Holmes,et al.  phyloseq: An R Package for Reproducible Interactive Analysis and Graphics of Microbiome Census Data , 2013, PloS one.

[58]  Pelin Yilmaz,et al.  The SILVA ribosomal RNA gene database project: improved data processing and web-based tools , 2012, Nucleic Acids Res..

[59]  Curtis Huttenhower,et al.  Microbial Co-occurrence Relationships in the Human Microbiome , 2012, PLoS Comput. Biol..

[60]  G. Trugnan,et al.  Bile acid profiling in human biological samples: comparison of extraction procedures and application to normal and cholestatic patients. , 2012, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[61]  C. Huttenhower,et al.  Metagenomic biomarker discovery and explanation , 2011, Genome Biology.

[62]  F. Bushman,et al.  The Macaque Gut Microbiome in Health, Lentiviral Infection, and Chronic Enterocolitis , 2008, PLoS pathogens.

[63]  M. Gershon,et al.  Physiological responses of guinea-pig myenteric neurons secondary to the release of endogenous serotonin by tryptamine , 1985, Neuroscience.