Gut Microbiota May Not Be Fully Restored in Recovered COVID-19 Patients After 3-Month Recovery

Yu Tian, Kai-Yi Sun, Tian-Qing Meng, Zhen Ye, Shi-Meng Guo, Zhi-Ming Li, Cheng-Liang Xiong, Ying Yin, Hong-Gang Li, Li-Quan Zhou, Yu Tian, Kai-Yi Sun, Tian-Qing Meng, Zhen Ye, Shi-Meng Guo, Zhi-Ming Li, Cheng-Liang Xiong, Ying Yin, Hong-Gang Li, Li-Quan Zhou

Abstract

Coronavirus disease 2019 (COVID-19) has infected over 124 million people worldwide. In addition to the development of therapeutics and vaccines, the evaluation of the sequelae in recovered patients is also important. Recent studies have indicated that COVID-19 has the ability to infect intestinal tissues and to trigger alterations of the gut microbiota. However, whether these changes in gut microbiota persist into the recovery stage remains largely unknown. Here, we recruited seven healthy Chinese men and seven recovered COVID-19 male patients with an average of 3-months after discharge and analyzed their fecal samples by 16S rRNA sequencing analysis to identify the differences in gut microbiota. Our results suggested that the gut microbiota differed in male recovered patients compared with healthy controls, in which a significant difference in Chao index, Simpson index, and β-diversity was observed. And the relative abundance of several bacterial species differed clearly between two groups, characterized by enrichment of opportunistic pathogens and insufficiency of some anti-inflammatory bacteria in producing short chain fatty acids. The above findings provide preliminary clues supporting that the imbalanced gut microbiota may not be fully restored in recovered patients, highlighting the importance of continuous monitoring of gut health in people who have recovered from COVID-19.

Keywords: 16S sequence; SARS-CoV-2; gut microbiota; recovered COVID-19 patient; short chain fatty acids.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Tian, Sun, Meng, Ye, Guo, Li, Xiong, Yin, Li and Zhou.

Figures

Figure 1
Figure 1
The relative abundance of gut microbiota at the Phylum (A) and Genus (B) levels in the healthy control group and the recovered COVID-19 patient group. The category “Other” covers all other phyla or genera with a low taxa abundance. RPs, recovered patients; HCs, healthy controls.
Figure 2
Figure 2
Differences in the diversity and richness of fecal microbiota between recovered COVID-19 patients and healthy controls. The Shannon (A), Chao (B), and Simpson index (C) were calculated at a 3% distance. (D) A Venn diagram indicates the overlapping and unique OTUs among the groups. (E) Non-metric multidimensional analysis (NMDS) was conducted at the OTU level to show the different distribution of recovered patients and controls.
Figure 3
Figure 3
The taxonomic differences of recovered COVID-19 patients and controls in the level of Class (A), Order (B), Family (C), and Genus (D).
Figure 4
Figure 4
Identifying bacterial differences by the LEfSe. (A) Taxonomic cladogram shows the taxa that were considered statistically significant between two groups from the Phylum to the Genus level. (B) the LDA score of differentially enriched taxa in healthy controls and COVID-19 group (LDA threshold value >3.5).
Figure 5
Figure 5
The levels of SCFAs in the fecal samples of recovered patients and healthy controls. Examination of fecal samples of the two groups on levels of (A) Acetic acid (AA), (B) Propionic acid (PA), (C) Isobutyric acid (IBA), (D) Butyric acid (BA), (E) Isovaleric acid (IVA), (F) Valeric acid (VA), and (G) Hexanoic acid (HA). (H) Correlation between relative abundance of bacteria genera and levels of seven SCFAs. *p < 0.05; **p < 0.01.

References

    1. Dong E, Du H, Gardner L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Infect Dis. (2020) 20:533–4. 10.1016/s1473-309930120-1
    1. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. . SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. (2020) 181:271–80.e8. 10.1016/j.cell.2020.02.052
    1. Xiao F, Tang M, Zheng X, Liu Y, Li X, Shan H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology. (2020) 158:1831–3 e3. 10.1053/j.gastro.2020.02.055
    1. Lin L, Jiang X, Zhang Z, Huang S, Zhang Z, Fang Z, et al. . Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut. (2020) 69:997–1001. 10.1136/gutjnl-2020-321013
    1. Xie C, Jiang L, Huang G, Pu H, Gong B, Lin H, et al. . Comparison of different samples for 2019 novel coronavirus detection by nucleic acid amplification tests. Int J Infect Dis. (2020) 93:264–7. 10.1016/j.ijid.2020.02.050
    1. Zhou J, Li C, Liu X, Chiu MC, Zhao X, Wang D, 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. Gu S, Chen Y, Wu Z, Chen Y, Gao H, Lv L, et al. . Alterations of the gut microbiota in patients with COVID-19 or H1N1 influenza. Clin Infect Dis. (2020) 71:2669–78. 10.1093/cid/ciaa709
    1. Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, et al. . Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. (2020) 159:944–55.e8. 10.1053/j.gastro.2020.05.048
    1. Zuo T, Liu Q, Zhang F, Lui GC, Tso EY, Yeoh YK, et al. . Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut. (2020) 70:276–84. 10.1136/gutjnl-2020-322294
    1. Gou W, Fu Y, Yue L, Chen G-D, Cai X, Shuai M, et al. . Gut microbiota may underlie the predisposition of healthy individuals to COVID-19. medRxiv. (2020). 10.1101/2020.04.22.20076091
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. (2016) 16:341–52. 10.1038/nri.2016.42
    1. Budden KF, Gellatly SL, Wood DLA, Cooper MA, Morrison M, Hugenholtz P, et al. . Emerging pathogenic links between microbiota and the gut–lung axis. Nat Rev Microbiol. (2016) 15:55–63. 10.1038/nrmicro.2016.142
    1. Wang J, Li F, Wei H, Lian ZX, Sun R, Tian Z. Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell-dependent inflammation. J Exp Med. (2014) 211:2397–410. 10.1084/jem.20140625
    1. National Health Commission of the People's Republic of China . Guidelines for COVID-19 Diagnosis and Treatment (Trial version 7). (2020). Available online at:
    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. Magoc T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics. (2011) 27:2957–63. 10.1093/bioinformatics/btr507
    1. Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. (2016) 165:1332–45. 10.1016/j.cell.2016.05.041
    1. Ferreira-Halder CV, Faria AVS, 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. Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ Microbiol. (2010) 12:304–14. 10.1111/j.1462-2920.2009.02066.x
    1. Sencio V, Barthelemy A, Tavares LP, Machado MG, Soulard D, Cuinat C, et al. . Gut dysbiosis during influenza contributes to pulmonary pneumococcal superinfection through altered short-chain fatty acid production. Cell Rep. (2020) 30:2934–47.e6. 10.1016/j.celrep.2020.02.013
    1. Segal LN, Clemente JC, Tsay JC, Koralov SB, Keller BC, Wu BG, et al. . Enrichment of the lung microbiome with oral taxa is associated with lung inflammation of a Th17 phenotype. Nat Microbiol. (2016) 1:16031. 10.1038/nmicrobiol.2016.31
    1. Maraki S, Papadakis IS. Rothia mucilaginosa pneumonia: a literature review. Infect Dis. (2015) 47:125–9. 10.3109/00365548.2014.980843
    1. Trottein F, Sokol H. Potential causes and consequences of gastrointestinal disorders during a SARS-CoV-2 infection. Cell Rep. (2020) 32:107915. 10.1016/j.celrep.2020.107915
    1. Hashimoto T, Perlot T, Rehman A, Trichereau J, Ishiguro H, Paolino M, et al. . ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature. (2012) 487:477–81. 10.1038/nature11228
    1. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. . Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. (2009) 461:1282–6. 10.1038/nature08530
    1. Luhrs H, Gerke T, Muller JG, Melcher R, Schauber J, Boxberge F, et al. . Butyrate inhibits NF-kappaB activation in lamina propria macrophages of patients with ulcerative colitis. Scand J Gastroenterol. (2002) 37:458–66. 10.1080/003655202317316105
    1. Sokol H, Contreras V, Maisonnasse P, Desmons A, Delache B, Sencio V, et al. . SARS-CoV-2 infection in nonhuman primates alters the composition and functional activity of the gut microbiota. Gut Microbes. (2021) 13:1–19. 10.1080/19490976.2021.1893113
    1. Miquel S, Leclerc M, Martin R, Chain F, Lenoir M, Raguideau S, et al. . Identification of metabolic signatures linked to anti-inflammatory effects of Faecalibacterium prausnitzii. mBio. (2015) 6:e00300–15. 10.1128/mBio.00300-15
    1. Hiippala K, Jouhten H, Ronkainen A, Hartikainen A, Kainulainen V, Jalanka J, et al. . The potential of gut commensals in reinforcing intestinal barrier function and alleviating inflammation. Nutrients. (2018) 10:988. 10.3390/nu10080988
    1. Walter J, Armet AM, Finlay BB, Shanahan F. Establishing or exaggerating causality for the gut microbiome: lessons from human microbiota-associated rodents. Cell. (2020) 180:221–32. 10.1016/j.cell.2019.12.025

Source: PubMed

3
Sottoscrivi