Lost microbes of COVID-19: Bifidobacterium, Faecalibacterium depletion and decreased microbiome diversity associated with SARS-CoV-2 infection severity

Sabine Hazan, Neil Stollman, Huseyin S Bozkurt, Sonya Dave, Andreas J Papoutsis, Jordan Daniels, Brad D Barrows, Eamonn Mm Quigley, Thomas J Borody, Sabine Hazan, Neil Stollman, Huseyin S Bozkurt, Sonya Dave, Andreas J Papoutsis, Jordan Daniels, Brad D Barrows, Eamonn Mm Quigley, Thomas J Borody

Abstract

Objective: The study objective was to compare gut microbiome diversity and composition in SARS-CoV-2 PCR-positive patients whose symptoms ranged from asymptomatic to severe versus PCR-negative exposed controls.

Design: Using a cross-sectional design, we performed shotgun next-generation sequencing on stool samples to evaluate gut microbiome composition and diversity in both patients with SARS-CoV-2 PCR-confirmed infections, which had presented to Ventura Clinical Trials for care from March 2020 through October 2021 and SARS-CoV-2 PCR-negative exposed controls. Patients were classified as being asymptomatic or having mild, moderate or severe symptoms based on National Institute of Health criteria. Exposed controls were individuals with prolonged or repeated close contact with patients with SARS-CoV-2 infection or their samples, for example, household members of patients or frontline healthcare workers. Microbiome diversity and composition were compared between patients and exposed controls at all taxonomic levels.

Results: Compared with controls (n=20), severely symptomatic SARS-CoV-2-infected patients (n=28) had significantly less bacterial diversity (Shannon Index, p=0.0499; Simpson Index, p=0.0581), and positive patients overall had lower relative abundances of Bifidobacterium (p<0.0001), Faecalibacterium (p=0.0077) and Roseburium (p=0.0327), while having increased Bacteroides (p=0.0075). Interestingly, there was an inverse association between disease severity and abundance of the same bacteria.

Conclusion: We hypothesise that low bacterial diversity and depletion of Bifidobacterium genera either before or after infection led to reduced proimmune function, thereby allowing SARS-CoV-2 infection to become symptomatic. This particular dysbiosis pattern may be a susceptibility marker for symptomatic severity from SARS-CoV-2 infection and may be amenable to preinfection, intrainfection or postinfection intervention.

Trial registration number: NCT04031469 (PCR-) and 04359836 (PCR+).

Keywords: BACTERIAL INFECTION; BIFIDOBACTERIA; ENTERIC BACTERIAL MICROFLORA; INTESTINAL MICROBIOLOGY.

Conflict of interest statement

Competing interests: SH declares that she has pecuniary interest in Topelia Pty Ltd in Australia, and Topelia Pty Ltd in USA where development of COVID-19 preventative/treatment options is being pursued. She has also filed patents relevant to Coronavirus treatments. She is the founder and owner of Microbiome research foundation, Progenabiome and Ventura Clinical Trials. TJB declares that he has pecuniary interest in Topelia Pty Ltd in Australia, and Topelia Therapeutics Inc. in USA developing COVID-19 preventative/treatment medications. He has also filed patents relevant to COVID-19 treatments. SD declares she has corporate affiliation to McKesson Specialty Health/Ontada and North End Advisory, LLC. SD is unaware of SARS-CoV-2 and microbiome projects and not directly involved in COVID-19 relevant projects at McKesson, but they may exist. AJP and BDB have corporate affiliations to Progenabiome. EMMQ serves as a consultant to Precisionbiotics, Novazymes, Salix, Biocodex and Axon Pharma and has received research support from 4D Pharma.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Distribution of bacterial relative abundance in various (A) phlya and (B) genera for exposed control (n=20, left) and SARS-CoV-2 positive subjects (n=50, right).
Figure 2
Figure 2
Diversity of gut microbiome composition of SARS-CoV-2 positive patients (severely symptomatic: n=28; moderately symptomatic: n=12; mildly symptomatic: n = 6; asymptomatic: n=4) versus exposed controls (n=20). (A) Shannon index (p=0.0499), (B) Simpson index (p=0.0581). Differences between severely symptomatic positive and exposed negative controls were analysed via Kruskal-Wallis test Dunn’s post-hoc, correcting for multiple comparisons, showing significant for Shannon index at p=0.0201.
Figure 3
Figure 3
Graphic of relative abundance of the 12 most common (A) families and (B) genera. The top group represents the SARS-CoV-2 positive samples (n=50), stratified by severity. The bottom group represents the exposed control samples (n=20). The coloured boxes represent the fraction of the entire rectangle composed of the given family/genera of bacteria.
Figure 4
Figure 4
Diagram of taxa comparing the gut microbiome of SARS-CoV-2 patients and exposed controls. Red or green background indicates a significant depletion or increase (due to positivity or severity), respectively, of the genus or species in SARS-CoV-2 positive subjects.
Figure 5
Figure 5
Relative abundance of Bifidobacterium in SARS-CoV-2 positive patients (n=50) versus SARS-CoV-2 negative exposed controls (n=20). Data are plotted as (A) mean with error bars for 95% CI and (B) individual points of relative abundance for varying SARS-CoV-2 infection severity. Analysed via Kruskal-Wallis test, there were significant reductions in Bifidobacterium relative abundance for severely (p<0.0001) and moderately (p=0.0002) symptomatic patients. Subjects 1–28 = severely symptomatic; subjects 29–40 = moderately symptomatic; subjects 41–46 = mildly symptomatic subjects; subjects 47–50 = asymptomatic; subjects 51–70 = exposed control. Figure A, B depicts same data.
Figure 6
Figure 6
SARS-CoV-2 positive patients’ microbiome is more similar to each other than to that of exposed controls. (A) Weighted UniFrac distance matrix of phylum level SARS-CoV-2 positive (n=50) and exposed negative control samples (n=20). Distance of microbiome differences increases with increasing blue colour intensity (see legend top right). The centre of the diagram consists of negative subjects on both axis and is yellow indicative of less distance (ie, lessdifference in microbiome). The central area of the left as well as central-top side of diagram, consists of negative subjects on one axis and positive on the other, and are darker blue, indicative of more distance (more difference in microbiome). (B) Principal component analysis of microbiota from SARS-CoV-2 positive (n=50) and exposed negative controls (n=20). Dots closer in distance are more similar in microbiome composition. Axes depict the per cent of variance explained by principal component (PC) 1 and 2. Plots are based on bacterial genera relative abundance profiles.
Figure 7
Figure 7
Potential mechanism for cytokine storm and immune hyper-response in SARS-CoV-2 positive patients. In individuals infected with SARS-CoV-2, the macrophages become activated; these in turn activate T-cells, additional macrophages, and neutrophils―all of which release cytokines, including TNF-α. Bifidobacterium, when present in sufficient numbers, can bind to TNF-α and prevent the subsequent cytokine storm. Therefore, patients with a bifidobacterial dysbiosis characterised by low levels of Bifidobacterium lack this line of defense, which may lead to a cytokine storm.

References

    1. De Vuyst L, Moens F, Selak M, et al. . Summer meeting 2013: growth and physiology of bifidobacteria. J Appl Microbiol 2014;116:477–91. 10.1111/jam.12415
    1. Parameswaran N, Patial S. Tumor necrosis factor-α signaling in macrophages. Crit Rev Eukaryot Gene Expr 2010;20:87–103. 10.1615/CritRevEukarGeneExpr.v20.i2.10
    1. Martins AKS, Martins FS, Gomes DA, et al. . Evaluation of in vitro antagonism and of in vivo immune modulation and protection against pathogenic experimental challenge of two probiotic strains of Bifidobacterium animalis var. lactis. Arch Microbiol 2010;192:995–1003. 10.1007/s00203-010-0626-0
    1. Groeger D, Schiavi E, Grant R, et al. . Intranasal Bifidobacterium longum protects against viral-induced lung inflammation and injury in a murine model of lethal influenza infection. EBioMedicine 2020;60:102981. 10.1016/j.ebiom.2020.102981
    1. Konieczna P, Akdis CA, Quigley EMM, et al. . Portrait of an immunoregulatory Bifidobacterium. Gut Microbes 2012;3:261–6. 10.4161/gmic.20358
    1. Konieczna P, Ferstl R, Ziegler M, et al. . Immunomodulation by Bifidobacterium infantis 35624 in the murine lamina propria requires retinoic acid-dependent and independent mechanisms. PLoS One 2013;8:e62617. 10.1371/journal.pone.0062617
    1. Schiavi E, Plattner S, Rodriguez-Perez N, et al. . Exopolysaccharide from Bifidobacterium longum subsp. longum 35624™ modulates murine allergic airway responses. Benef Microbes 2018;9:761–73. 10.3920/BM2017.0180
    1. Ruiz L, Delgado S, Ruas-Madiedo P, et al. . Bifidobacteria and their molecular communication with the immune system. Front Microbiol 2017;8:2345. 10.3389/fmicb.2017.02345
    1. Marras L, Caputo M, Bisicchia S, et al. . The role of bifidobacteria in predictive and preventive medicine: a focus on eczema and hypercholesterolemia. Microorganisms 2021;9. 10.3390/microorganisms9040836. [Epub ahead of print: 14 04 2021].
    1. Stavropoulou E, Bezirtzoglou E. Probiotics in medicine: a long debate. Front Immunol 2020;11:2192. 10.3389/fimmu.2020.02192
    1. Ahlawat S, Sharma KK. Immunological co-ordination between gut and lungs in SARS-CoV-2 infection. Virus Res 2020;286:198103. 10.1016/j.virusres.2020.198103
    1. Follmer C. Viral infection-induced gut dysbiosis, neuroinflammation, and α-synuclein aggregation: updates and perspectives on COVID-19 and neurodegenerative disorders. ACS Chem Neurosci 2020;11:4012–6. 10.1021/acschemneuro.0c00671
    1. Marsland BJ, Trompette A, Gollwitzer ES. The Gut-Lung axis in respiratory disease. Ann Am Thorac Soc 2015;12 Suppl 2:S150–6. 10.1513/AnnalsATS.201503-133AW
    1. Janda L, Mihalčin M, Šťastná M. Is a healthy microbiome responsible for lower mortality in COVID-19? Biologia 2021;76:819–29. 10.2478/s11756-020-00614-8
    1. Tiwari SK, Dicks LMT, Popov IV, et al. . Probiotics at war against viruses: what is missing from the picture? Front Microbiol 2020;11:11. 10.3389/fmicb.2020.01877
    1. Bozkurt HS, Quigley EM. The probiotic Bifidobacterium in the management of Coronavirus: A theoretical basis. Int J Immunopathol Pharmacol 2020;34:2058738420961304. 10.1177/2058738420961304
    1. Jin X, Lian J-S, Hu J-H, et al. . Epidemiological, clinical and virological characteristics of 74 cases of coronavirus-infected disease 2019 (COVID-19) with gastrointestinal symptoms. Gut 2020;69:1002–9. 10.1136/gutjnl-2020-320926
    1. Gu S, Chen Y, Wu Z, et al. . Alterations of the gut microbiota in patients with coronavirus disease 2019 or H1N1 influenza. Clin Infect Dis 2020;71:2669–78. 10.1093/cid/ciaa709
    1. Zuo T, Liu Q, Zhang F, et al. . Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 2021;70:276–84. 10.1136/gutjnl-2020-322294
    1. Yeoh YK, Zuo T, Lui GC-Y, et al. . Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021;70:698–706. 10.1136/gutjnl-2020-323020
    1. Xu K, Cai H, Shen Y. Management of COVID-19: the Zhejiang experience. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020;49:147–57.
    1. Liu Q, Mak JWY, Su Q, et al. . Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 2022;71:544–52. 10.1136/gutjnl-2021-325989
    1. Din AU, Hassan A, Zhu Y, et al. . Inhibitory effect of Bifidobacterium bifidum ATCC 29521 on colitis and its mechanism. J Nutr Biochem 2020;79:108353. 10.1016/j.jnutbio.2020.108353
    1. Nitzan O, Elias M, Peretz A, et al. . Role of antibiotics for treatment of inflammatory bowel disease. World J Gastroenterol 2016;22:1078–87. 10.3748/wjg.v22.i3.1078
    1. Valdés-Varela L, Hernández-Barranco AM, Ruas-Madiedo P, et al. . Effect of Bifidobacterium upon Clostridium difficile growth and toxicity when co-cultured in different prebiotic substrates. Front Microbiol 2016;7:738. 10.3389/fmicb.2016.00738
    1. Wei Y, Yang F, Wu Q, et al. . Protective Effects of Bifidobacterial Strains Against Toxigenic Clostridium difficile. Front Microbiol 2018;9:888. 10.3389/fmicb.2018.00888
    1. Philippe D, Heupel E, Blum-Sperisen S, et al. . Treatment with Bifidobacterium bifidum 17 partially protects mice from Th1-driven inflammation in a chemically induced model of colitis. Int J Food Microbiol 2011;149:45–9. 10.1016/j.ijfoodmicro.2010.12.020
    1. Overview of variants in countries. Available:
    1. Tao W, Zhang G, Wang X, et al. . Analysis of the intestinal microbiota in COVID-19 patients and its correlation with the inflammatory factor IL-18. Med Microecol 2020;5:100023. 10.1016/j.medmic.2020.100023
    1. Yagisawa MFP, Hanaki H, Ōmura S. Global trends in clinical studies of ivermectin in COVID-19. Japanese Journal of Antibiotics 2021;74:44–94.
    1. Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–99. 10.1053/j.gastro.2014.02.009
    1. Negi S, Das DK, Pahari S, et al. . Potential role of gut microbiota in induction and regulation of innate immune memory. Front Immunol 2019;10:2441. 10.3389/fimmu.2019.02441
    1. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med 2016;8:51. 10.1186/s13073-016-0307-y
    1. Ghouri YA, Richards DM, Rahimi EF, et al. . Systematic review of randomized controlled trials of probiotics, prebiotics, and synbiotics in inflammatory bowel disease. Clin Exp Gastroenterol 2014;7:473–87. 10.2147/CEG.S27530
    1. Hughes KR, Harnisch LC, Alcon-Giner C, et al. . Bifidobacterium breve reduces apoptotic epithelial cell shedding in an exopolysaccharide and MyD88-dependent manner. Open Biol 2017;7:160155. 10.1098/rsob.160155
    1. Fanning S, Hall LJ, Cronin M, et al. . Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection. Proc Natl Acad Sci U S A 2012;109:2108–13. 10.1073/pnas.1115621109
    1. Arboleya S, Watkins C, Stanton C, et al. . Gut bifidobacteria populations in human health and aging. Front Microbiol 2016;7:1204. 10.3389/fmicb.2016.01204
    1. Ferreira-Halder CV, Faria AVdeS, Andrade SS. Action and function of Faecalibacterium prausnitzii in health and disease. Best Pract Res Clin Gastroenterol 2017;31:643–8. 10.1016/j.bpg.2017.09.011
    1. Ganesan K, Chung SK, Vanamala J, et al. . Causal relationship between diet-induced gut microbiota changes and diabetes: a novel strategy to transplant Faecalibacterium prausnitzii in preventing diabetes. Int J Mol Sci 2018;19. 10.3390/ijms19123720. [Epub ahead of print: 22 Nov 2018].
    1. Greene MW, Roberts AP, Frugé AD. Negative association between Mediterranean diet adherence and COVID-19 cases and related deaths in Spain and 23 OECD countries: an ecological study. Front Nutr 2021;8:591964. 10.3389/fnut.2021.591964
    1. Tang L, Yin Z, Hu Y, et al. . Controlling cytokine storm is vital in COVID-19. Front Immunol 2020;11:570993. 10.3389/fimmu.2020.570993
    1. Zhao M. Cytokine storm and immunomodulatory therapy in COVID-19: role of chloroquine and anti-IL-6 monoclonal antibodies. Int J Antimicrob Agents 2020;55:105982. 10.1016/j.ijantimicag.2020.105982
    1. Biagi E, Nylund L, Candela M, et al. . Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One 2010;5:e10667. 10.1371/journal.pone.0010667
    1. Center for Disease Control . People with certain medical conditions. Available:

Source: PubMed

3
Suscribir