Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19

Tao Zuo, Qin Liu, Fen Zhang, Grace Chung-Yan Lui, Eugene Yk Tso, Yun Kit Yeoh, Zigui Chen, Siaw Shi Boon, Francis Kl Chan, Paul Ks Chan, Siew C Ng, Tao Zuo, Qin Liu, Fen Zhang, Grace Chung-Yan Lui, Eugene Yk Tso, Yun Kit Yeoh, Zigui Chen, Siaw Shi Boon, Francis Kl Chan, Paul Ks Chan, Siew C Ng

Abstract

Objective: Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA was detected in faeces of patients with COVID-19, the activity and infectivity of the virus in the GI tract during disease course is largely unknown. We investigated temporal transcriptional activity of SARS-CoV-2 and its association with longitudinal faecal microbiome alterations in patients with COVID-19.

Design: We performed RNA shotgun metagenomics sequencing on serial faecal viral extractions from 15 hospitalised patients with COVID-19. Sequencing coverage of the SARS-CoV-2 genome was quantified. We assessed faecal microbiome composition and microbiome functionality in association with signatures of faecal SARS-CoV-2 infectivity.

Results: Seven (46.7%) of 15 patients with COVID-19 had stool positivity for SARS-CoV-2 by viral RNA metagenomic sequencing. Even in the absence of GI manifestations, all seven patients showed strikingly higher coverage (p=0.0261) and density (p=0.0094) of the 3' vs 5' end of SARS-CoV-2 genome in their faecal viral metagenome profile. Faecal viral metagenome of three patients continued to display active viral infection signature (higher 3' vs 5' end coverage) up to 6 days after clearance of SARS-CoV-2 from respiratory samples. Faecal samples with signature of high SARS-CoV-2 infectivity had higher abundances of bacterial species Collinsella aerofaciens, Collinsella tanakaei, Streptococcus infantis, Morganella morganii, and higher functional capacity for nucleotide de novo biosynthesis, amino acid biosynthesis and glycolysis, whereas faecal samples with signature of low-to-none SARS-CoV-2 infectivity had higher abundances of short-chain fatty acid producing bacteria, Parabacteroides merdae, Bacteroides stercoris, Alistipes onderdonkii and Lachnospiraceae bacterium 1_1_57FAA.

Conclusion: This pilot study provides evidence for active and prolonged 'quiescent' GI infection even in the absence of GI manifestations and after recovery from respiratory infection of SARS-CoV-2. Gut microbiota of patients with active SARS-CoV-2 GI infection was characterised by enrichment of opportunistic pathogens, loss of salutary bacteria and increased functional capacity for nucleotide and amino acid biosynthesis and carbohydrate metabolism.

Keywords: diagnostic virology; gut inflammation; infectious disease.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Timeline of patient symptom onset, hospitalisation, throat (nasopharyngeal) swab clearance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and discharge, through the course of disease for 15 patients hospitalised with COVID-19.
Figure 2
Figure 2
Hypothetical scenarios for the fecal viral RNA metagenomic profile of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in association with its infectivity in gut. (A) Schematic presentation of the full-length genome, transcribed subgenomic RNAs (sgRNA) and the virion structure of SARS-CoV-2 virus. (B) Three scenarios hypothesised for the presence and infectivity of SARS-CoV-2 in the gut of patients with COVID-19, and the detection of SARS-CoV-2 virus by faecal viral RNA metegenomics sequencing. If SARS-CoV-2 virus infects the host cells in the gut, its genomic and sgRNAs should be highly expressed and released into the gut lumen on cytolysis, where the 3’ end of SARS-CoV-2 genome should be highly covered by faecal viral RNA metagenomics sequencing.
Figure 3
Figure 3
Depiction of the viral infectivity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in serial faeces of patients with COVID-19. Infectivity of SARS-CoV-2 virus in gut was investigated by faecal viral RNA metagenomics coverage profile of SARS-CoV-2 genome. (A) The subset of patients who manifested higher 3’ vs 5’ end coverage of the SARS-CoV-2 genome (signature of active SARS-CoV-2 infectivity) before throat swab turned negative for SARS-CoV-2. (B) The subset of patients who manifested higher 3’ vs 5’ end coverage of the SARS-CoV-2 genome (signature of SARS-CoV-2 infectivity) but gradually lost this signature over time of hospitalisation before throat swab turned negative for SARS-CoV-2. (C) The subset of patients who manifested higher 3’ vs 5’ end coverage of the SARS-CoV-2 genome (signature of SARS-CoV-2 infectivity) after throat swab turned negative for SARS-CoV-2. ‘Day 0’ is defined as the date when throat swab turned negative for SARS-CoV-2, as measured by RT-PCR. (D) The coverage and density of the 5’ and 3’ ends of the SARS-CoV-2 genome in COVID-19 faecal viral RNA metagenome. The baseline (the date of first stool collection after hospitalisation) faecal viral RNA metagenomes of the seven patients who were detected faecal positive for SARS-CoV-2 were plotted and subject to comparison. Coverage was defined as the number of shotgun reads mapped to a given genomic region of SARS-CoV-2 genome. Density was defined as the frequency of sequenced sites in a given genomic region of SARS-CoV-2 genome.
Figure 4
Figure 4
Differential bacterial species and functional capacities between faeces with high severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity and faeces with low-to-none SARS-CoV-2 infectivity. Differential bacterial species (A) and functionality (B) were identified via LefSE analysis across all time-point stools of 15 patients with COVID-19. Only species and functional modules with LDA effect size >2 and FDR-corrected p value

Figure 5

Longitudal changes in the faecal…

Figure 5

Longitudal changes in the faecal microbiome of patients with COVID-19 in association with…

Figure 5
Longitudal changes in the faecal microbiome of patients with COVID-19 in association with faecal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity. Patients 3 and 7 had serial stools displaying positive to negative faecal SARS-CoV-2 infectivity during follow-up, while patients 11, 12 and 15 had serial stools constantly displaying a signature high viral infectivity during follow-up. Only the most abundant 20 species were plotted and shown in relative abundance.
Figure 5
Figure 5
Longitudal changes in the faecal microbiome of patients with COVID-19 in association with faecal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity. Patients 3 and 7 had serial stools displaying positive to negative faecal SARS-CoV-2 infectivity during follow-up, while patients 11, 12 and 15 had serial stools constantly displaying a signature high viral infectivity during follow-up. Only the most abundant 20 species were plotted and shown in relative abundance.

References

    1. Wölfel R, Corman VM, Guggemos W, et al. . Virological assessment of hospitalized patients with COVID-2019. Nature 2020;581:465–9. 10.1038/s41586-020-2196-x
    1. Huang C, Wang Y, Li X, et al. . Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. The Lancet 2020;395:497–506. 10.1016/S0140-6736(20)30183-5
    1. Chen N, Zhou M, Dong X, et al. . Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020;395:507–13. 10.1016/S0140-6736(20)30211-7
    1. Liang W, Feng Z, Rao S, et al. . Diarrhoea may be underestimated: a missing link in 2019 novel coronavirus. Gut 2020;69:1141–3. 10.1136/gutjnl-2020-320832
    1. Cheung KS, Hung IFN, Chan PPY, 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. Gastroenterology 2020. 10.1053/j.gastro.2020.03.065
    1. Effenberger M, Grabherr F, Mayr L, et al. . Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut 2020;69:1543–4. 10.1136/gutjnl-2020-321388
    1. Xu Y, Li X, Zhu B, et al. . Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding. Nat Med 2020;26:502–5. 10.1038/s41591-020-0817-4
    1. Wang W, Xu Y, Gao R, et al. . Detection of SARS-CoV-2 in different types of clinical specimens. JAMA 2020. 10.1001/jama.2020.3786
    1. Zuo T, Zhang F, Lui GCY, et al. . Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 2020. 10.1053/j.gastro.2020.05.048
    1. Wang J, Zhao S, Liu M, et al. . ACE2 expression by colonic epithelial cells is associated with viral infection immunity and energy metabolism. medRxiv 2020.
    1. Xiao F, Tang M, Zheng X, et al. . Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020;158:1831–3. 10.1053/j.gastro.2020.02.055
    1. Lamers MM, Beumer J, van der Vaart J, et al. . SARS-CoV-2 productively infects human gut enterocytes. Science 2020;369:50–4. 10.1126/science.abc1669
    1. Zhou J, Li C, Liu X, et al. . Infection of bat and human intestinal organoids by SARS-CoV-2. Nat Med 2020;26:1077–83. 10.1038/s41591-020-0912-6
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 2014;30:2114–20. 10.1093/bioinformatics/btu170
    1. Bushnell B. BBMap: a fast, accurate, splice-aware aligner. Berkeley, CA (United States): Lawrence Berkeley National Lab.(LBNL), 2014.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012;9:357–9. 10.1038/nmeth.1923
    1. Segata N, Waldron L, Ballarini A, et al. . Metagenomic microbial community profiling using unique clade-specific marker genes. Nat Methods 2012;9:811–4. 10.1038/nmeth.2066
    1. Franzosa EA, McIver LJ, Rahnavard G, et al. . Species-Level functional profiling of metagenomes and metatranscriptomes. Nat Methods 2018;15:962–8. 10.1038/s41592-018-0176-y
    1. Wu A, Peng Y, Huang B, et al. . Genome composition and divergence of the novel coronavirus (2019-nCoV) originating in China. Cell Host Microbe 2020;27:325–8. 10.1016/j.chom.2020.02.001
    1. Kim D, Lee J-Y, Yang J-S, et al. . The architecture of SARS-CoV-2 transcriptome. Cell 2020;181:914–21. 10.1016/j.cell.2020.04.011
    1. Shinagawa N, Taniguchi M, Hirata K, et al. . [Bacteria isolated from surgical infections and its susceptibilities to antimicrobial agents--special references to bacteria isolated between April 2010 and March 2011]. Jpn J Antibiot 2014;67:293–334.
    1. Liu H, Zhu J, Hu Q, et al. . Morganella morganii, a non-negligent opportunistic pathogen. Int J Infect Dis 2016;50:10–17. 10.1016/j.ijid.2016.07.006
    1. Bek-Thomsen M, Tettelin H, Hance I, et al. . Population diversity and dynamics of Streptococcus mitis, Streptococcus oralis, and Streptococcus infantis in the upper respiratory tracts of adults, determined by a nonculture strategy. Infect Immun 2008;76:1889–96. 10.1128/IAI.01511-07
    1. Hwang N, Eom T, Gupta SK, et al. . Genes and gut bacteria involved in luminal butyrate reduction caused by diet and loperamide. Genes 2017;8:350. 10.3390/genes8120350
    1. Kläring K, Just S, Lagkouvardos I, et al. . Murimonas intestini gen. nov., sp. nov., an acetate-producing bacterium of the family Lachnospiraceae isolated from the mouse gut. Int J Syst Evol Microbiol 2015;65:870–8. 10.1099/ijs.0.000030
    1. Remely M, Aumueller E, Merold C, et al. . Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene 2014;537:85–92. 10.1016/j.gene.2013.11.081
    1. Ratajczak W, Rył A, Mizerski A, et al. . Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim Pol 2019;66:1–12. 10.18388/abp.2018_2648
    1. Gao J, Xu K, Liu H, et al. . Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front Cell Infect Microbiol 2018;8:13. 10.3389/fcimb.2018.00013
    1. Verdu EF, Hayes CL, O’Mahony SM. Importance of the microbiota in early life and influence on future health. The Gut-Brain Axis: Elsevier, 2016: 159–84.
    1. Geva-Zatorsky N, Sefik E, Kua L, et al. . Mining the human gut microbiota for immunomodulatory organisms. Cell 2017;168:928–43. 10.1016/j.cell.2017.01.022
    1. Jimmerson LC, Bushman LR, Ray ML, et al. . A LC-MS/MS method for quantifying adenosine, guanosine and inosine nucleotides in human cells. Pharm Res 2017;34:73–83. 10.1007/s11095-016-2040-z
    1. Antonioli L, Blandizzi C, Pacher P, et al. . Immunity, inflammation and cancer: a leading role for adenosine. Nat Rev Cancer 2013;13:842–57. 10.1038/nrc3613
    1. Shibata T, Ohto U, Nomura S, et al. . Guanosine and its modified derivatives are endogenous ligands for TLR7. Int Immunol 2016;28:211–22. 10.1093/intimm/dxv062
    1. Kitamoto S, Alteri CJ, Rodrigues M, et al. . Dietary L-serine confers a competitive fitness advantage to Enterobacteriaceae in the inflamed gut. Nat Microbiol 2020;5:116–25. 10.1038/s41564-019-0591-6
    1. Yeo C, Kaushal S, Yeo D. Enteric involvement of coronaviruses: is faecal-oral transmission of SARS-CoV-2 possible? Lancet Gastroenterol Hepatol 2020;5:335–7. 10.1016/S2468-1253(20)30048-0
    1. Gu J, Han B, Wang J. COVID-19: gastrointestinal manifestations and potential Fecal-Oral transmission. Gastroenterology 2020;158:1518–9. 10.1053/j.gastro.2020.02.054
    1. Zang R, Gomez Castro MF, McCune BT, et al. . Tmprss2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci Immunol 2020;5. 10.1126/sciimmunol.abc3582. [Epub ahead of print: 13 05 2020].

Source: PubMed

Подписаться