Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19

Yun Kit Yeoh, Tao Zuo, Grace Chung-Yan Lui, Fen Zhang, Qin Liu, Amy Yl Li, Arthur Ck Chung, Chun Pan Cheung, Eugene Yk Tso, Kitty Sc Fung, Veronica Chan, Lowell Ling, Gavin Joynt, David Shu-Cheong Hui, Kai Ming Chow, Susanna So Shan Ng, Timothy Chun-Man Li, Rita Wy Ng, Terry Cf Yip, Grace Lai-Hung Wong, Francis Kl Chan, Chun Kwok Wong, Paul Ks Chan, Siew C Ng, Yun Kit Yeoh, Tao Zuo, Grace Chung-Yan Lui, Fen Zhang, Qin Liu, Amy Yl Li, Arthur Ck Chung, Chun Pan Cheung, Eugene Yk Tso, Kitty Sc Fung, Veronica Chan, Lowell Ling, Gavin Joynt, David Shu-Cheong Hui, Kai Ming Chow, Susanna So Shan Ng, Timothy Chun-Man Li, Rita Wy Ng, Terry Cf Yip, Grace Lai-Hung Wong, Francis Kl Chan, Chun Kwok Wong, Paul Ks Chan, Siew C Ng

Abstract

Objective: Although COVID-19 is primarily a respiratory illness, there is mounting evidence suggesting that the GI tract is involved in this disease. We investigated whether the gut microbiome is linked to disease severity in patients with COVID-19, and whether perturbations in microbiome composition, if any, resolve with clearance of the SARS-CoV-2 virus.

Methods: In this two-hospital cohort study, we obtained blood, stool and patient records from 100 patients with laboratory-confirmed SARS-CoV-2 infection. Serial stool samples were collected from 27 of the 100 patients up to 30 days after clearance of SARS-CoV-2. Gut microbiome compositions were characterised by shotgun sequencing total DNA extracted from stools. Concentrations of inflammatory cytokines and blood markers were measured from plasma.

Results: Gut microbiome composition was significantly altered in patients with COVID-19 compared with non-COVID-19 individuals irrespective of whether patients had received medication (p<0.01). Several gut commensals with known immunomodulatory potential such as Faecalibacterium prausnitzii, Eubacterium rectale and bifidobacteria were underrepresented in patients and remained low in samples collected up to 30 days after disease resolution. Moreover, this perturbed composition exhibited stratification with disease severity concordant with elevated concentrations of inflammatory cytokines and blood markers such as C reactive protein, lactate dehydrogenase, aspartate aminotransferase and gamma-glutamyl transferase.

Conclusion: Associations between gut microbiota composition, levels of cytokines and inflammatory markers in patients with COVID-19 suggest that the gut microbiome is involved in the magnitude of COVID-19 severity possibly via modulating host immune responses. Furthermore, the gut microbiota dysbiosis after disease resolution could contribute to persistent symptoms, highlighting a need to understand how gut microorganisms are involved in inflammation and COVID-19.

Keywords: colonic bacteria; colonic microflora; inflammation.

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
Compositional differences in gut microbiota between patients with COVID-19 and non-COVID-19 subjects. (A) Average relative abundances of microbial phyla detected in stools from in-hospital patients with COVID-19, patients discharged after negative RT-qPCR for viral RNA in nasopharyngeal swabs, and non-COVID-19 individuals. (B) Principal component analysis of gut microbiota composition of patients with COVID-19 with and without antibiotics compared with non-COVID-19 subjects. Filled circles represent the first stool samples (if serial samples are available) of in-hospital patients whereas crosses represent non-COVID-19 subjects. Group centroids are indicated by the group labels.
Figure 2
Figure 2
Associations between gut microbiota composition of in-hospital patients with COVID-19 and plasma concentrations of inflammatory cytokines and blood inflammation markers. (A) Principal component analysis (PCA) of gut microbiota composition and association with plasma concentrations of cytokines/chemokines. (B) PCA of gut microbiota composition and association with blood inflammation markers. Statistical correlations were determined using Procrustes tests. Only cytokines and inflammation markers significantly correlated with gut microbiota composition are shown. Red arrows represent gradients of the corresponding cytokines/inflammation marker concentrations and point to the direction of greatest increase in these measures. Colour of the circles represents disease severity groups, and ellipses represent SD of the group centroid. Group centroids are indicated by the group labels. AST, aspartate aminotransferase; CRP, C-reactive protein; ESR, erythrocyte sedimentation rate; GGT, gamma-glutamyl transferase; LDH, lactate dehydrogenase; NT-proBNP, N-terminal-pro-brain natriuretic peptide; TNF, tumour necrosis factor.
Figure 3
Figure 3
Correlations between COVID-19 enriched/depleted gut microbial taxa and plasma concentrations of (A) CXCL10, (B) IL-10, (C) TNF-α, (D) CCL2, (E) CXCL8, (F) IL-1β and (G) IL-6. Only statistically significant correlations are shown. Linear regression lines are shown in each scatter plot in blue, and shaded regions represent 95% CIs. CCL, C-C motif ligand; CXCL, C-X-C motif ligand; TNF, tumour necrosis factor.
Figure 4
Figure 4
Principal component analysis of gut microbiota composition in recovered patients with COVID-19 who had or had not received antibiotics compared with non-COVID-19 subjects. Patients were considered recovered following negative quantitative reverse transcription PCR (RT-qPCR) tests for SARS-CoV-2 RNA in nasopharyngeal swabs. Filled circles represent all stools collected after discharge from hospital whereas crosses represent non-COVID-19 subjects.

References

    1. Tay MZ, Poh CM, Rénia L, et al. . The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol 2020;20:363–74. 10.1038/s41577-020-0311-8
    1. Vabret N, Britton GJ, Gruber C, et al. . Immunology of COVID-19: current state of the science. Immunity 2020;52:910–41. 10.1016/j.immuni.2020.05.002
    1. Zhou F, Yu T, Du R, et al. . Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020;395:1054–62. 10.1016/S0140-6736(20)30566-3
    1. Cheung EW, Zachariah P, Gorelik M, et al. . Multisystem inflammatory syndrome related to COVID-19 in previously healthy children and adolescents in New York City. JAMA 2020;324:294. 10.1001/jama.2020.10374
    1. Galeotti C, Bayry J. Autoimmune and inflammatory diseases following COVID-19. Nat Rev Rheumatol 2020;16:413–4. 10.1038/s41584-020-0448-7
    1. Verdoni L, Mazza A, Gervasoni A, et al. . An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet 2020;395:1771–8. 10.1016/S0140-6736(20)31103-X
    1. Lamers MM, Beumer J, van der Vaart J, et al. . SARS-CoV-2 productively infects human gut enterocytes. Science 2020;369:eabc1669. 10.1126/science.abc1669
    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. 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. Zuo T, Zhang F, Lui GCY, et al. . Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 2020;159:944–55. 10.1053/j.gastro.2020.05.048
    1. Gu S, Chen Y, Wu Z, et al. . Alterations of the gut microbiota in patients with COVID-19 or H1N1 influenza. Clin Infect Dis 2020:ciaa709. 10.1093/cid/ciaa709
    1. Schirmer M, Smeekens SP, Vlamakis H, et al. . Linking the human gut microbiome to inflammatory cytokine production capacity. Cell 2016;167:1125–36. 10.1016/j.cell.2016.10.020
    1. Yip TC-F, Lui GC-Y, Wong VW-S. Liver injury is independently associated with adverse clinical outcomes in patients with COVID-19. Gut 2021;70:733–42. 10.1136/gutjnl-2020-321726
    1. Yeoh YK, Chen Z, Wong MCS, et al. . Southern Chinese populations harbour non-nucleatum Fusobacteria possessing homologues of the colorectal cancer-associated fadA virulence factor. Gut 2020;69:1998–2007. 10.1136/gutjnl-2019-319635
    1. Zhang J, Hoedt EC, Liu Q, et al. . Elucidation of Proteus mirabilis as a Key Bacterium in Crohn’s Disease Inflammation. Gastroenterology 2020;S0016-5085:35217–3.
    1. Wu J, Liu J, Zhao X, et al. . Clinical characteristics of imported cases of coronavirus disease 2019 (COVID-19) in Jiangsu Province: a multicenter descriptive study. Clin Infect Dis 2020;71:706–12. 10.1093/cid/ciaa199
    1. Chen Z, Hui PC, Hui M, et al. . Impact of preservation method and 16S rRNA hypervariable region on gut microbiota profiling. mSystems 2019;4:e00271–18. 10.1128/mSystems.00271-18
    1. Truong DT, Franzosa EA, Tickle TL, et al. . MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods 2015;12:902–3. 10.1038/nmeth.3589
    1. R Core Team . R: a language and environment for statistical computing, 2018. Available:
    1. Oksanen J, Blanchet FG, Kindt R, et al. . Package ‘vegan.’ Community ecology package, version, 2013: 2.
    1. Shen B, Yi X, Sun Y, et al. . Proteomic and metabolomic characterization of COVID-19 patient sera. Cell 2020;182:59–72. 10.1016/j.cell.2020.05.032
    1. Gao L, Jiang D, Wen X-S, et al. . Prognostic value of NT-proBNP in patients with severe COVID-19. Respir Res 2020;21:83. 10.1186/s12931-020-01352-w
    1. Henry BM, Aggarwal G, Wong J, et al. . Lactate dehydrogenase levels predict coronavirus disease 2019 (COVID-19) severity and mortality: a pooled analysis. Am J Emerg Med 2020;38:1722–6. 10.1016/j.ajem.2020.05.073
    1. Sokol H, Pigneur B, Watterlot L, et al. . Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 2008;105:16731–6. 10.1073/pnas.0804812105
    1. van den Munckhof ICL, Kurilshikov A, Ter Horst R, et al. . Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: a systematic review of human studies. Obes Rev 2018;19:1719–34. 10.1111/obr.12750
    1. Parada Venegas D, De la Fuente MK, Landskron G, et al. . Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol 2019;10:277. 10.3389/fimmu.2019.00277
    1. Carfì A, Bernabei R, Landi F, et al. . Persistent symptoms in patients after acute COVID-19. JAMA 2020;324:603. 10.1001/jama.2020.12603
    1. Laing AG, Lorenc A, Barrio DMD, et al. . A consensus Covid-19 immune signature combines immuno-protection with discrete sepsis-like traits associated with poor prognosis. medRxiv 2020:2020.06.08.20125112.
    1. Long Q-X, Tang X-J, Shi Q-L, et al. . Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat Med 2020;26:1200–4. 10.1038/s41591-020-0965-6
    1. Ong EZ, Chan YFZ, Leong WY, et al. . A dynamic immune response shapes COVID-19 progression. Cell Host Microbe 2020;27:879–82. 10.1016/j.chom.2020.03.021
    1. Alameddine J, Godefroy E, Papargyris L, et al. . Faecalibacterium prausnitzii Skews Human DC to Prime IL10-Producing T Cells Through TLR2/6/JNK Signaling and IL-10, IL-27, CD39, and IDO-1 Induction. Front Immunol 2019;10:143. 10.3389/fimmu.2019.00143
    1. Cattaneo A, Cattane N, Galluzzi S, et al. . Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging 2017;49:60–8. 10.1016/j.neurobiolaging.2016.08.019
    1. Riedel C-U, Foata F, Philippe D, et al. . Anti-Inflammatory effects of bifidobacteria by inhibition of LPS-induced NF-kappaB activation. World J Gastroenterol 2006;12:3729–35. 10.3748/wjg.v12.i23.3729
    1. Matsuoka K, Kanai T. The gut microbiota and inflammatory bowel disease. Semin Immunopathol 2015;37:47–55. 10.1007/s00281-014-0454-4
    1. Hall AB, Yassour M, Sauk J, et al. . A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med 2017;9:103. 10.1186/s13073-017-0490-5
    1. Davis-Richardson AG, Ardissone AN, Dias R, et al. . Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes. Front Microbiol 2014;5:678. 10.3389/fmicb.2014.00678
    1. Ahmed M, Advani S, Moreira A, et al. . Multisystem inflammatory syndrome in children: a systematic review. EClinicalMedicine 2020;26:100527. 10.1016/j.eclinm.2020.100527
    1. Townsend L, Dyer AH, Jones K, et al. . Persistent fatigue following SARS-CoV-2 infection is common and independent of severity of initial infection. PLoS One 2020;15:2020.07.29.20164293. 10.1371/journal.pone.0240784
    1. Goërtz YMJ, Van Herck M, Delbressine JM, et al. . Persistent symptoms 3 months after a SARS-CoV-2 infection: the post-COVID-19 syndrome? ERJ Open Res 2020;6:00542-2020. 10.1183/23120541.00542-2020
    1. Langford BJ, So M, Raybardhan S, et al. . Bacterial co-infection and secondary infection in patients with COVID-19: a living rapid review and meta-analysis. Clin Microbiol Infect 2020. 10.1016/j.cmi.2020.07.016. [Epub ahead of print: 22 Jul 2020].
    1. Vaughn VM, Gandhi T, Petty LA, et al. . Empiric antibacterial therapy and community-onset bacterial co-infection in patients hospitalized with COVID-19: a Multi-Hospital cohort study. Clin Infect Dis 2020:ciaa1239. 10.1093/cid/ciaa1239
    1. Goncalves Mendes Neto A, Lo KB, Wattoo A, et al. . Bacterial infections and patterns of antibiotic use in patients with COVID-19. J Med Virol 2020. 10.1002/jmv.26441. [Epub ahead of print: 18 Aug 2020].
    1. Thevaranjan N, Puchta A, Schulz C, et al. . Age-Associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe 2017;21:455–66. 10.1016/j.chom.2017.03.002

Source: PubMed

3
订阅