Health
Gut microbiota in COVID-19: key microbial changes, potential mechanisms and clinical applications
Tregoning, J. S., Flight, K. E., Higham, S. L., Wang, Z. & Pierce, B. F. Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat. Rev. Immunol. 21, 626–636 (2021).
Files, J. K. et al. Duration of post-COVID-19 symptoms is associated with sustained SARS-CoV-2-specific immune responses. JCI Insight 6, e151544 (2021).
Lamers, M. M. et al. SARS-CoV-2 productively infects human gut enterocytes. Science 369, 50–54 (2020).
Zang, R. et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci. Immunol. 5, eabc3582 (2020).
Zuo, T. et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 70, 276–284 (2020).
Xiao, F. et al. Infectious SARS-CoV-2 in feces of patient with severe COVID-19. Emerg. Infect. Dis. 26, 1920 (2020).
Natarajan, A. et al. Gastrointestinal symptoms and fecal shedding of SARS-CoV-2 RNA suggest prolonged gastrointestinal infection. Med 3, 371–387 (2022).
Zollner, A. et al. Post-acute COVID-19 is characterized by gut viral antigen persistence in inflammatory bowel diseases. Gastroenterology 163, 495–506 (2022).
Cheung, K. S. 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 159, 81–95 (2020).
Mao, R. et al. Manifestations and prognosis of gastrointestinal and liver involvement in patients with COVID-19: a systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 5, 667–678 (2020).
Song, Y. et al. SARS-CoV-2 induced diarrhoea as onset symptom in patient with COVID-19. Gut 69, 1143–1144 (2020).
Wang, D. et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA 323, 1061–1069 (2020).
Gilbert, J. A. et al. Current understanding of the human microbiome. Nat. Med. 24, 392–400 (2018).
Rothschild, D. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215 (2018).
Kurilshikov, A. et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat. Genet. 53, 156–165 (2021).
Wolter, M. et al. Leveraging diet to engineer the gut microbiome. Nat. Rev. Gastroenterol. Hepatol. 18, 885–902 (2021).
Ruff, W. E., Greiling, T. M. & Kriegel, M. A. Host–microbiota interactions in immune-mediated diseases. Nat. Rev. Microbiol. 18, 521–538 (2020).
Groves, H. T., Higham, S. L., Moffatt, M. F., Cox, M. J. & Tregoning, J. S. Respiratory viral infection alters the gut microbiota by inducing inappetence. mBio 11, e03236-19 (2020).
Zhang, D. et al. The cross-talk between gut microbiota and lungs in common lung diseases. Front. Microbiol. 11, 301 (2020).
Sun, Z. et al. Gut microbiome alterations and gut barrier dysfunction are associated with host immune homeostasis in COVID-19 patients. BMC Med. 20, 24 (2022).
Vatanen, T. et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 165, 842–853 (2016).
Anand, S. & Mande, S. S. Diet, microbiota and gut-lung connection. Front. Microbiol. 9, 2147 (2018).
Parrot, T. et al. MAIT cell activation and dynamics associated with COVID-19 disease severity. Sci. Immunol. 5, eabe1670 (2020).
Legoux, F., Salou, M. & Lantz, O. MAIT cell development and functions: the microbial connection. Immunity 53, 710–723 (2020).
Hashimoto, T. et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 487, 477–481 (2012).
Viana, S. D., Nunes, S. & Reis, F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities–role of gut microbiota dysbiosis. Ageing Res. Rev. 62, 101123 (2020).
Gaibani, P. et al. The gut microbiota of critically ill patients with COVID-19. Front. Cell. Infect. Microbiol. 11, 670424 (2021).
Gu, S. et al. Alterations of the gut microbiota in patients with coronavirus disease 2019 or H1N1 influenza. Clin. Infect. Dis. 71, 2669–2678 (2020).
Ren, Z. et al. Alterations in the human oral and gut microbiomes and lipidomics in COVID-19. Gut 70, 1253–1265 (2021).
Xu, R. et al. Temporal association between human upper respiratory and gut bacterial microbiomes during the course of COVID-19 in adults. Commun. Biol. 4, 240 (2021).
Mizutani, T. et al. Correlation analysis between gut microbiota alterations and the cytokine response in patients with coronavirus disease during hospitalization. Microbiol. Spectr. 10, e0168921 (2022).
Rafiqul Islam, S. et al. Dysbiosis of oral and gut microbiomes in SARS-CoV-2 infected patients in Bangladesh: elucidating the role of opportunistic gut microbes. Front. Med. 9, 163 (2022).
Reinold, J. et al. A pro-inflammatory gut microbiome characterizes SARS-CoV-2 infected patients and a reduction in the connectivity of an anti-inflammatory bacterial network associates with severe COVID-19. Front. Cell. Infect. Microbiol. 11, 1154 (2021).
Tang, L. et al. Clinical significance of the correlation between changes in the major intestinal bacteria species and COVID-19 severity. Engineering 6, 1178–1184 (2020).
Tao, W. et al. Analysis of the intestinal microbiota in COVID-19 patients and its correlation with the inflammatory factor IL-18. Med. Microecol. 5, 100023 (2020).
Wu, Y. et al. Altered oral and gut microbiota and its association with SARS-CoV-2 viral load in COVID-19 patients during hospitalization. NPJ Biofilms Microbiomes 7, 61 (2021).
Yeoh, Y. K. et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 70, 698–706 (2021).
Li, S. et al. Microbiome profiling using shotgun metagenomic sequencing identified unique microorganisms in COVID-19 patients with altered gut microbiota. Front. Microbiol. 12, 712081 (2021).
Zuo, T. et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 159, 944–955.e8 (2020).
Bolte, E. E., Moorshead, D. & Aagaard, K. M. Maternal and early life exposures and their potential to influence development of the microbiome. Genome Med. 14, 4 (2022).
de Steenhuijsen Piters, W. A., Binkowska, J. & Bogaert, D. Early life microbiota and respiratory tract infections. Cell Host Microbe 28, 223–232 (2020).
Renz, H. & Skevaki, C. Early life microbial exposures and allergy risks: opportunities for prevention. Nat. Rev. Immunol. 21, 177–191 (2021).
Zhang, X.-S. et al. Maternal cecal microbiota transfer rescues early-life antibiotic-induced enhancement of type 1 diabetes in mice. Cell Host Microbe 29, 1249–1265.e9 (2021).
Sarkar, A., Yoo, J. Y., Valeria Ozorio Dutra, S., Morgan, K. H. & Groer, M. The association between early-life gut microbiota and long-term health and diseases. J. Clin. Med. 10, 459 (2021).
Xu, R. et al. Progressive deterioration of the upper respiratory tract and the gut microbiomes in children during the early infection stages of COVID-19. J. Genet. Genomics 48, 803–814 (2021).
Nashed, L. et al. Gut microbiota changes are detected in asymptomatic very young children with SARS-CoV-2 infection. Gut https://doi.org/10.1136/gutjnl-2021-326599 (2022).
Jiang, L. et al. COVID-19 and multisystem inflammatory syndrome in children and adolescents. Lancet Infect. Dis. 20, e276–e288 (2020).
Suskun, C. et al. Intestinal microbiota composition of children with infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and multisystem inflammatory syndrome (MIS-C). Eur. J. Pediatr. 181, 3175–3191 (2022).
Nash, A. K. et al. The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome 5, 153 (2017).
Sender, R., Fuchs, S. & Milo, R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 14, e1002533 (2016).
Lv, L. et al. Gut mycobiota alterations in patients with COVID-19 and H1N1 infections and their associations with clinical features. Commun. Biol. 4, 480 (2021).
Zuo, T. et al. Alterations in fecal fungal microbiome of patients with COVID-19 during time of hospitalization until discharge. Gastroenterology 159, 1302–1310.e5 (2020).
Roudbary, M. et al. Overview on the prevalence of fungal infections, immune response, and microbiome role in COVID-19 patients. J. Fungi 7, 720 (2021).
Arastehfar, A. et al. COVID-19-associated candidiasis (CAC): an underestimated complication in the absence of immunological predispositions? J. Fungi 6, 211 (2020).
Zuo, T. et al. Temporal landscape of human gut RNA and DNA virome in SARS-CoV-2 infection and severity. Microbiome 9, 91 (2021).
Cao, J. et al. Integrated gut virome and bacteriome dynamics in COVID-19 patients. Gut Microbes 13, 1887722 (2021).
Matheson, N. J. & Lehner, P. J. How does SARS-CoV-2 cause COVID-19? Science 369, 510–511 (2020).
Katz-Agranov, N. & Zandman-Goddard, G. Autoimmunity and COVID-19–the microbiotal connection. Autoimmun. Rev. 20, 102865 (2021).
Vignesh, R. et al. Could perturbation of gut microbiota possibly exacerbate the severity of COVID-19 via cytokine storm? Front. Immunol. 11, 607734 (2021).
Prasad, R. et al. Plasma microbiome in COVID-19 subjects: an indicator of gut barrier defects and dysbiosis. Preprint at bioRxiv https://doi.org/10.1101/2021.04.06.438634 (2021).
Effenberger, M. et al. Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut 69, 1543–1544 (2020).
Giron, L. B. et al. Plasma markers of disrupted gut permeability in severe COVID-19 patients. Front. Immunol. 12, 686240 (2021).
Chhibber-Goel, J., Gopinathan, S. & Sharma, A. Interplay between severities of COVID-19 and the gut microbiome: implications of bacterial co-infections? Gut Pathog. 13, 14 (2021).
Langford, B. J. et al. Bacterial co-infection and secondary infection in patients with COVID-19: a living rapid review and meta-analysis. Clin. Microbiol. Infect. 26, 1622–1629 (2020).
Saade, A. et al. Infectious events in patients with severe COVID-19: results of a cohort of patients with high prevalence of underlying immune defect. Ann. Intensive Care 11, 83 (2021).
Huang, C. et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395, 497–506 (2020).
Garcia-Vidal, C. et al. Incidence of co-infections and superinfections in hospitalized patients with COVID-19: a retrospective cohort study. Clin. Microbiol. Infect. 27, 83–88 (2021).
Zhou, F. et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 395, 1054–1062 (2020).
Moreira-Rosário, A. et al. Gut microbiota diversity and C-reactive protein are predictors of disease severity in COVID-19 patients. Front. Microbiol. 12, 1820 (2021).
Liu, Y. et al. Distinct metagenomic signatures in the SARS-CoV-2 infection. Front. Cell. Infect. Microbiol. 11, 1019 (2021).
Zhou, Y. et al. Gut microbiota dysbiosis correlates with abnormal immune response in moderate COVID-19 patients with fever. J. Inflamm. Res. 14, 2619 (2021).
Brodin, P. Immune determinants of COVID-19 disease presentation and severity. Nat. Med. 27, 28–33 (2021).
Nowarski, R. et al. Epithelial IL-18 equilibrium controls barrier function in colitis. Cell 163, 1444–1456 (2015).
Huang, N. et al. SARS-CoV-2 infection of the oral cavity and saliva. Nat. Med. 27, 892–903 (2021).
Soffritti, I. et al. Oral microbiome dysbiosis is associated with symptoms severity and local immune/inflammatory response in COVID-19 patients: a cross-sectional study. Front. Microbiol. 12, 687513 (2021).
de Castilhos, J. et al. Severe dysbiosis and specific Haemophilus and Neisseria signatures as hallmarks of the oropharyngeal microbiome in critically ill COVID-19 patients. Clin. Infect. Dis. 75, e1063–e1071 (2021).
Miller, E. H. et al. Oral microbiome alterations and SARS-CoV-2 saliva viral load in patients with COVID-19. Microbiol. Spectr. 9, e0005521 (2021).
Ma, S. et al. Metagenomic analysis reveals oropharyngeal microbiota alterations in patients with COVID-19. Signal. Transduct. Target. Ther. 6, 191 (2021).
Newsome, R. C. et al. The gut microbiome of COVID-19 recovered patients returns to uninfected status in a minority-dominated United States cohort. Gut Microbes 13, 1926840 (2021).
Nasserie, T., Hittle, M. & Goodman, S. N. Assessment of the frequency and variety of persistent symptoms among patients with COVID-19: a systematic review. JAMA Netw. Open 4, e2111417 (2021).
Huang, C. et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet 397, 220–232 (2021).
Nalbandian, A. et al. Post-acute COVID-19 syndrome. Nat. Med. 27, 601–615 (2021).
Liu, Q. et al. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 71, 544–552 (2022).
Evans, R. A. et al. Physical, cognitive, and mental health impacts of COVID-19 after hospitalisation (PHOSP-COVID): a UK multicentre, prospective cohort study. Lancet Respir. Med. 9, 1275–1287 (2021).
Choutka, J., Jansari, V., Hornig, M. & Iwasaki, A. Unexplained post-acute infection syndromes. Nat. Med. 28, 911–923 (2022).
Lee, Y. Y., Annamalai, C. & Rao, S. S. Post-infectious irritable bowel syndrome. Curr. Gastroenterol. Rep. 19, 56 (2017).
Holtmann, G. J., Ford, A. C. & Talley, N. J. Pathophysiology of irritable bowel syndrome. Lancet Gastroenterol. Hepatol. 1, 133–146 (2016).
Austhof, E. et al. Persisting gastrointestinal symptoms and post-infectious irritable bowel syndrome following SARS-CoV-2 infection: results from the Arizona CoVHORT. Epidemiol. Infect. 150, e136 (2022).
Ghoshal, U. C. et al. Post‐infection functional gastrointestinal disorders following coronavirus disease‐19: a case–control study. J. Gastroenterol. Hepatol. 37, 489–498 (2022).
Al-Aly, Z., Bowe, B. & Xie, Y. Long COVID after breakthrough SARS-CoV-2 infection. Nat. Med. 28, 1461–1467 (2022).
Hu, J. et al. Probiotics, prebiotics and dietary approaches during COVID-19 pandemic. Trends Food Sci. Technol. 108, 187–196 (2021).
Chen, Y. et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut 71, 222–225 (2022).
Vestad, B. et al. Respiratory dysfunction three months after severe COVID‐19 is associated with gut microbiota alterations. J. Intern. Med. 291, 801–812 (2022).
Su, Q., Lau, R. I., Liu, Q., Chan, F. K. L. & Ng, S. C. Post-acute COVID-19 syndrome and gut dysbiosis linger beyond 1 year after SARS-CoV-2 clearance. Gut https://doi.org/10.1136/gutjnl-2022-328319 (2022).
Cui, G. Y. et al. Characterization of oral and gut microbiome and plasma metabolomics in COVID-19 patients after 1-year follow-up. Mil. Med. Res. 9, 32 (2022).
Cervia, C. et al. Immunoglobulin signature predicts risk of post-acute COVID-19 syndrome. Nat. Commun. 13, 446 (2022).
Marx, V. Scientists set out to connect the dots on long COVID. Nat. Methods 18, 449–453 (2021).
Zhou, Y., Zhang, J., Zhang, D., Ma, W.-L. & Wang, X. Linking the gut microbiota to persistent symptoms in survivors of COVID-19 after discharge. J. Microbiol. 59, 941–948 (2021).
Jalanka-Tuovinen, J. et al. Faecal microbiota composition and host–microbe cross-talk following gastroenteritis and in postinfectious irritable bowel syndrome. Gut 63, 1737–1745 (2014).
Zhang, H. et al. Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection. Int. J. Infect. Dis. 96, 19–24 (2020).
Koester, S. T., Li, N., Lachance, D. M., Morella, N. M. & Dey, N. Variability in digestive and respiratory tract Ace2 expression is associated with the microbiome. PLoS ONE 16, e0248730 (2021).
Hirayama, M. et al. Intestinal Collinsella may mitigate infection and exacerbation of COVID-19 by producing ursodeoxycholate. PLoS ONE 16, e0260451 (2021).
Geva-Zatorsky, N. et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 168, 928–943.e11 (2017).
Vijay, A. & Valdes, A. M. Role of the gut microbiome in chronic diseases: a narrative review. Eur. J. Clin. Nutr. 76, 489–501 (2021).
O’Toole, P. W. & Jeffery, I. B. Gut microbiota and aging. Science 350, 1214–1215 (2015).
Li, Y. et al. Systematic profiling of ACE2 expression in diverse physiological and pathological conditions for COVID‐19/SARS‐CoV‐2. J. Cell. Mol. Med. 24, 9478–9482 (2020).
Gracia-Ramos, A. E. Is the ACE2 overexpression a risk factor for COVID-19 infection? Arch. Med. Res. 51, 345–346 (2020).
Martino, C. et al. Bacterial modification of the host glycosaminoglycan heparan sulfate modulates SARS-CoV-2 infectivity. Preprint at bioRxiv https://doi.org/10.1101/2020.08.17.238444 (2020).
Merad, M. & Martin, J. C. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat. Rev. Immunol. 20, 355–362 (2020).
Kircheis, R. et al. NF-κB pathway as a potential target for treatment of critical stage COVID-19 patients. Front. Immunol. 11, 3446 (2020).
Guo, Y. et al. SARS-CoV-2 induced intestinal responses with a biomimetic human gut-on-chip. Sci. Bull. 66, 783–793 (2021).
Cerf-Bensussan, N. & Gaboriau-Routhiau, V. The immune system and the gut microbiota: friends or foes? Nat. Rev. Immunol. 10, 735–744 (2010).
Yan, R. et al. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 367, 1444–1448 (2020).
Lecarpentier, Y. & Vallée, A. The key role of the level of ACE2 gene expression in SARS-CoV-2 infection. Aging 13, 14552 (2021).
Brogna, C. et al. Could SARS-CoV-2 have bacteriophage behavior or induce the activity of other bacteriophages? Vaccines 10, 708 (2022).
Seibert, B. et al. Mild and severe SARS-CoV-2 infection induces respiratory and intestinal microbiome changes in the K18-hACE2 transgenic mouse model. Microbiol. Spectr. 9, e0053621 (2021).
Sokol, H. et al. SARS-CoV-2 infection in nonhuman primates alters the composition and functional activity of the gut microbiota. Gut Microbes 13, 1893113 (2021).
Sencio, V. et al. Alteration of the gut microbiota following SARS-CoV-2 infection correlates with disease severity in hamsters. Gut Microbes 14, 2018900 (2022).
Pascoal, L. B. et al. Microbiota-derived short-chain fatty acids do not interfere with SARS-CoV-2 infection of human colonic samples. Gut Microbes 13, 1874740 (2021).
Zhou, T. et al. SARS‐CoV‐2 triggered oxidative stress and abnormal energy metabolism in gut microbiota. MedComm 3, 41–56 (2022).
He, F. et al. Fecal multi-omics analysis reveals diverse molecular alterations of gut ecosystem in COVID-19 patients. Anal. Chim. Acta 1180, 338881 (2021).
Lv, L. et al. The faecal metabolome in COVID-19 patients is altered and associated with clinical features and gut microbes. Anal. Chim. Acta 1152, 338267 (2021).
Al Bataineh, M. T. et al. Gut microbiota interplay with COVID-19 reveals links to host lipid metabolism among Middle Eastern populations. Front. Microbiol. 12, 761067 (2021).
Zhang, F. et al. Prolonged impairment of short-chain fatty acid and L-isoleucine biosynthesis in gut microbiome in patients with COVID-19. Gastroenterology 162, 548–561.e4 (2022).
Geirnaert, A. et al. Butyrate-producing bacteria supplemented in vitro to Crohn’s disease patient microbiota increased butyrate production and enhanced intestinal epithelial barrier integrity. Sci. Rep. 7, 11450 (2017).
Yao, Y. et al. The role of short-chain fatty acids in immunity, inflammation and metabolism. Crit. Rev. Food Sci. Nutr. 62, 1–12 (2020).
Lionetto, L. et al. Increased kynurenine-to-tryptophan ratio in the serum of patients infected with SARS-CoV2: an observational cohort study. Biochim. Biophys. Acta Mol. Basis Dis. 1867, 166042 (2021).
Barberis, E. et al. Large-scale plasma analysis revealed new mechanisms and molecules associated with the host response to SARS-CoV-2. Int. J. Mol. Sci. 21, 8623 (2020).
Robertson, J. et al. Serum neopterin levels in relation to mild and severe COVID-19. BMC Infect. Dis. 20, 942 (2020).
Eroğlu, İ., Eroğlu, B. Ç. & Güven, G. S. Altered tryptophan absorption and metabolism could underlie long-term symptoms in survivors of coronavirus disease 2019 (COVID-19). Nutrition 90, 111308 (2021).
Dagenais-Lussier, X. et al. Latest developments in tryptophan metabolism: understanding its role in B cell immunity. Cytokine Growth Factor Rev. 59, 111–117 (2021).
Cervenka, I., Agudelo, L. Z. & Ruas, J. L. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science 357, eaaf9794 (2017).
Agus, A., Planchais, J. & Sokol, H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23, 716–724 (2018).
Gao, J. et al. Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front. Cell. Infect. Microbiol. 8, 13 (2018).
Sonner, J. K. et al. Dietary tryptophan links encephalogenicity of autoreactive T cells with gut microbial ecology. Nat. Commun. 10, 4877 (2019).
Shen, B. et al. Proteomic and metabolomic characterization of COVID-19 patient sera. Cell 182, 59–72.e15 (2020).
Rohrhofer, J., Zwirzitz, B., Selberherr, E. & Untersmayr, E. The impact of dietary sphingolipids on intestinal microbiota and gastrointestinal immune homeostasis. Front. Immunol. 12, 635704 (2021).
Brown, E. M. et al. Bacteroides-derived sphingolipids are critical for maintaining intestinal homeostasis and syMbiosis. Cell Host Microbe 25, 668–680.e7 (2019).
Kue, C. S. et al. C6-ceramide in combination with transforming growth factor-β enhances Treg cell differentiation and stable FoxP3 expression in vitro and in vivo. Immunobiology 218, 952–959 (2013).
Gericke, B., Amiri, M. & Naim, H. Y. The multiple roles of sucrase-isomaltase in the intestinal physiology. Mol. Cell. Pediatr. 3, 2 (2016).
Deb, C. et al. Sucrase-isomaltase gene variants in patients with abnormal sucrase activity and functional gastrointestinal disorders. J. Pediatr. Gastroenterol. Nutr. 72, 29–35 (2021).
Kalantar-Zadeh, K., Berean, K. J., Burgell, R. E., Muir, J. G. & Gibson, P. R. Intestinal gases: influence on gut disorders and the role of dietary manipulations. Nat. Rev. Gastroenterol. Hepatol. 16, 733–747 (2019).
Nasreen, S. et al. Effectiveness of COVID-19 vaccines against symptomatic SARS-CoV-2 infection and severe outcomes with variants of concern in Ontario. Nat. Microbiol. 7, 379–385 (2022).
Andrews, N. et al. Effectiveness of COVID-19 booster vaccines against covid-19 related symptoms, hospitalisation and death in England. Nat. Med. 28, 831–837 (2022).
Lynn, D. J., Benson, S. C., Lynn, M. A. & Pulendran, B. Modulation of immune responses to vaccination by the microbiota: implications and potential mechanisms. Nat. Rev. Immunol. 22, 33–46 (2021).
Pulendran, B., S Arunachalam, P. & O’Hagan, D. T. Emerging concepts in the science of vaccine adjuvants. Nat. Rev. Drug Discov. 20, 454–475 (2021).
Oh, J. Z. et al. TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity 41, 478–492 (2014).
Kim, D. et al. Nod2-mediated recognition of the microbiota is critical for mucosal adjuvant activity of cholera toxin. Nat. Med. 22, 524–530 (2016).
Kim, M., Qie, Y., Park, J. & Kim, C. H. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe 20, 202–214 (2016).
Ng, S. C. et al. Gut microbiota composition is associated with SARS-CoV-2 vaccine immunogenicity and adverse events. Gut 71, 1106–1116 (2022).
Alexander, J. L. et al. The gut microbiota and metabolome is associated with diminished COVID-19 vaccine-induced antibody responses in immunosuppressed inflammatory bowel disease patients. Lancet Gastroenterol. Hepatol. 7, 342–352 (2022).
Uehara, O. et al. Alterations in the oral microbiome of individuals with a healthy oral environment following COVID-19 vaccination. BMC Oral Health 22, 50 (2022).
Levine-Tiefenbrun, M. et al. Waning of SARS-CoV-2 booster viral-load reduction effectiveness. Nat. Commun. 13, 1237 (2022).
Ward, H. et al. Population antibody responses following COVID-19 vaccination in 212,102 individuals. Nat. Commun. 13, 907 (2022).
Pérez-Alós, L. et al. Modeling of waning immunity after SARS-CoV-2 vaccination and influencing factors. Nat. Commun. 13, 1614 (2022).
Araos, R. et al. Effectiveness of CoronaVac in children 3 to 5 years during the omicron SARS-CoV-2 outbreak. Nat. Med. 28, 1377–1380 (2022).
Finlay, B. B. et al. The hygiene hypothesis, the COVID pandemic, and consequences for the human microbiome. Proc. Natl Acad. Sci. USA 118, e2010217118 (2021).
Aguilera, P. et al. A two-time point analysis of gut microbiota in the general population of buenos aires and its variation due to preventive and compulsory social isolation during the COVID-19 pandemic. Front. Microbiol. 13, 803121 (2022).
Peng, Y. et al. Gut microbiome and resistome changes during the first wave of the COVID-19 pandemic in comparison with pre-pandemic travel-related changes. J. Travel Med. 28, taab067 (2021).
Romano-Keeler, J., Zhang, J. & Sun, J. COVID-19 and the neonatal microbiome: will the pandemic cost infants their microbes? Gut Microbes 13, 1912562 (2021).
Xia, J. S., Oliphant, K. & Claud, E. The pandemic effects on the microbiome of infants in the neonatal intensive care unit (NICU). FASEB J. https://doi.org/10.1096/fasebj.2022.36.S1.0R562 (2022).
Rundle, A. G., Park, Y., Herbstman, J. B., Kinsey, E. W. & Wang, Y. C. COVID-19 related school closings and risk of weight gain among children. Obesity 28, 1008 (2020).
Liu, J. J., Bao, Y., Huang, X., Shi, J. & Lu, L. Mental health considerations for children quarantined because of COVID-19. Lancet Child. Adolesc. Health 4, 347–349 (2020).
Wang, J. et al. Progression of myopia in school-aged children after COVID-19 home confinement. JAMA Ophthal. 139, 293–300 (2021).
Bauer, K. W. et al. A safety net unraveling: Feeding young children during COVID-19. Am. J. Public Health 111, 116–120 (2021).
Langford, B. J. et al. Antibiotic prescribing in patients with COVID-19: rapid review and meta-analysis. Clin. Microbiol. Infect. 27, 520–531 (2021).
Hagan, T. et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell 178, 1313–1328.e13 (2019).
Hegazy, M. et al. Beyond probiotic legend: ESSAP gut microbiota health score to delineate SARS-COV-2 infection severity. Br. J. Nutr. 127, 1180–1189 (2021).
Merino, J. et al. Diet quality and risk and severity of COVID-19: a prospective cohort study. Gut 70, 2096–2104 (2021).
Hou, Y.-C., Su, W.-L. & Chao, Y.-C. COVID-19 illness severity in the elderly in relation to vegetarian and non-vegetarian diets: a single-center experience. Front. Nutr. 9, 837458 (2022).
Zhu, L. et al. Association of blood glucose control and outcomes in patients with COVID-19 and pre-existing type 2 diabetes. Cell Metab. 31, 1068–1077.e3 (2020).
Bousquet, J. et al. Is diet partly responsible for differences in COVID‐19 death rates between and within countries? Clin. Transl. Allergy 10, 16 (2020).
Wang, Y., Wu, G., Zhao, L. & Wang, W. Nutritional modulation of gut microbiota alleviates severe gastrointestinal symptoms in a patient with post-acute COVID-19 syndrome. mBio 13, e03801–21 (2022).
Schupack, D. A., Mars, R. A., Voelker, D. H., Abeykoon, J. P. & Kashyap, P. C. The promise of the gut microbiome as part of individualized treatment strategies. Nat. Rev. Gastroenterol. Hepatol. 19, 7–25 (2021).
Zhang, L. et al. Probiotics use is associated with improved clinical outcomes among hospitalized patients with COVID-19. Ther. Adv. Gastroenterol. 14, 17562848211035670 (2021).
Ceccarelli, G. et al. Oral bacteriotherapy in patients with COVID-19: a retrospective cohort study. Front. Nutr. 7, 341 (2021).
Rathi, A., Jadhav, S. B. & Shah, N. A randomized controlled trial of the efficacy of systemic enzymes and probiotics in the resolution of post-COVID fatigue. Medicines 8, 47 (2021).
Gutiérrez-Castrellón, P. et al. Probiotic improves symptomatic and viral clearance in Covid19 outpatients: a randomized, quadruple-blinded, placebo-controlled trial. Gut Microbes 14, 2018899 (2022).
Wu, C. et al. The volatile and heterogeneous gut microbiota shifts of COVID‐19 patients over the course of a probiotics‐assisted therapy. Clin. Transl. Med. 11, e643 (2021).
Zhang, L. et al. Gut microbiota‐derived synbiotic formula (SIM01) as a novel adjuvant therapy for COVID‐19: An open‐label pilot study. J. Gastroenterol. Hepatol. 37, 823–831 (2022).
Heavey, M. K., Durmusoglu, D., Crook, N. & Anselmo, A. C. Discovery and delivery strategies for engineered live biotherapeutic products. Trends Biotechnol. 40, 254–369 (2021).
Piscotta, F. J. et al. Metabolites with SARS-CoV-2 inhibitory activity identified from human microbiome commensals. mSphere 6, e0071121 (2021).
Liu, F. et al. Gastrointestinal disturbance and effect of fecal microbiota transplantation in discharged COVID-19 patients. J. Med. Case Rep. 15, 60 (2021).
Biliński, J. et al. Rapid resolution of COVID-19 after faecal microbiota transplantation. Gut 71, 230–232 (2022).
Constantinides, M. G. Interactions between the microbiota and innate and innate‐like lymphocytes. J. Leukoc. Biol. 103, 409–419 (2018).
Sources 2/ https://www.nature.com/articles/s41575-022-00698-4 The mention sources can contact us to remove/changing this article |
What Are The Main Benefits Of Comparing Car Insurance Quotes Online
LOS ANGELES, CA / ACCESSWIRE / June 24, 2020, / Compare-autoinsurance.Org has launched a new blog post that presents the main benefits of comparing multiple car insurance quotes. For more info and free online quotes, please visit https://compare-autoinsurance.Org/the-advantages-of-comparing-prices-with-car-insurance-quotes-online/ The modern society has numerous technological advantages. One important advantage is the speed at which information is sent and received. With the help of the internet, the shopping habits of many persons have drastically changed. The car insurance industry hasn't remained untouched by these changes. On the internet, drivers can compare insurance prices and find out which sellers have the best offers. View photos The advantages of comparing online car insurance quotes are the following: Online quotes can be obtained from anywhere and at any time. Unlike physical insurance agencies, websites don't have a specific schedule and they are available at any time. Drivers that have busy working schedules, can compare quotes from anywhere and at any time, even at midnight. Multiple choices. Almost all insurance providers, no matter if they are well-known brands or just local insurers, have an online presence. Online quotes will allow policyholders the chance to discover multiple insurance companies and check their prices. Drivers are no longer required to get quotes from just a few known insurance companies. Also, local and regional insurers can provide lower insurance rates for the same services. Accurate insurance estimates. Online quotes can only be accurate if the customers provide accurate and real info about their car models and driving history. Lying about past driving incidents can make the price estimates to be lower, but when dealing with an insurance company lying to them is useless. Usually, insurance companies will do research about a potential customer before granting him coverage. Online quotes can be sorted easily. Although drivers are recommended to not choose a policy just based on its price, drivers can easily sort quotes by insurance price. Using brokerage websites will allow drivers to get quotes from multiple insurers, thus making the comparison faster and easier. For additional info, money-saving tips, and free car insurance quotes, visit https://compare-autoinsurance.Org/ Compare-autoinsurance.Org is an online provider of life, home, health, and auto insurance quotes. This website is unique because it does not simply stick to one kind of insurance provider, but brings the clients the best deals from many different online insurance carriers. In this way, clients have access to offers from multiple carriers all in one place: this website. On this site, customers have access to quotes for insurance plans from various agencies, such as local or nationwide agencies, brand names insurance companies, etc. "Online quotes can easily help drivers obtain better car insurance deals. All they have to do is to complete an online form with accurate and real info, then compare prices", said Russell Rabichev, Marketing Director of Internet Marketing Company. CONTACT: Company Name: Internet Marketing CompanyPerson for contact Name: Gurgu CPhone Number: (818) 359-3898Email: [email protected]: https://compare-autoinsurance.Org/ SOURCE: Compare-autoinsurance.Org View source version on accesswire.Com:https://www.Accesswire.Com/595055/What-Are-The-Main-Benefits-Of-Comparing-Car-Insurance-Quotes-Online View photos
to request, modification Contact us at Here or [email protected]