GET THE APP

Journal of Clinical Trials

Journal of Clinical Trials
Open Access

ISSN: 2167-0870

+44 1478 350008

Mini Review - (2020)Volume 10, Issue 4

Intestinal Dysbiosis and Probiotics in COVID-19

Yasunari Kageyama1,2, Tetsu Akiyama3 and Tsutomu Nakamura1,3*
 
*Correspondence: Tsutomu Nakamura, Laboratory of Molecular and Genetic Information, Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan, Email:

Author info »

Abstract

The COVID-19 pandemic has been an emerging threat to global public health. Several lines of evidence suggest that the causative virus SARS-CoV-2 infects the human intestinal epithelial cells as well as airway epithelial cells, suggesting that the enteric infection of SARS-CoV-2 has destructive effects on the intestinal microbiota and subsequently airway physiology and immunity through the gut-lung axis. Despite the important roles of the gut-lung axis in the antiviral immunity, only limited information is currently available concerning COVID-19-specific changes in the gut microbiome. This review summarizes recent knowledge of intestinal dysbiosis associated with COVID-19 patients and its potential contribution to the respiratory symptoms through the gut-lung axis. We also discuss the possibility of prophylactic and therapeutic use of probiotics in COVID-19, including our ongoing trial using Lactobacillus plantarum, which is known to have a wide variety of immunomodulatory activity against respiratory viral infections.

Keywords

COVID-19; Dysbiosis; Gut-lung axis; Microbiome; Microbiota; Probiotics

Introduction

The recent pandemic of COVID-19 has been an emerging threat to global public health. COVID-19 patients present with a spectrum of disease severity, ranging from asymptomatic or mild flu-like to critically ill cases with severe pneumonia potentially accompanied by acute respiratory distress syndrome (ARDS) and multiple organ failure [1,2]. The causative coronavirus, SARSCoV- 2, primarily infects a subpopulation of airway epithelial cells co-expressing angiotensin-converting enzyme 2 (ACE2), a cell surface viral receptor, and transmembrane serine protease 2 (TMPRSS2), a transmembrane protease that facilitates viral entry into host cells [3-5].

Notably, several lines of evidence suggest that the human intestinal tract is an additional target of SARS-CoV-2 infection. Many clinical studies have revealed that a considerable percentage of COVID-19 patients suffer from concurrent gastrointestinal manifestations, most commonly diarrhea and abdominal pain [6-8]. SARS-CoV-2 viral RNA has been detected in stool specimens and anal/rectal swabs of patients even after the clearance of the virus in the nasopharynx [9-11], and furthermore, infectious viruses have been isolated from feces of a COVID-19 patient with diarrhea [12]. ACE2 and TMPRSS2 are co-expressed in the lower gastrointestinal tract, particularly enterocytes and progenitor cells of the ileum and colon [4,5,13-15]. In fact, SARS-CoV-2 can infect human intestinal organoids and replicate actively [12,15-17]. These findings raise the possibility that the enteric infection of SARS-CoV-2 has negative effects on intestinal commensal microbesand subsequently airway physiology and immunity through the gutlung axis.

In this review, we summarize recent knowledge of intestinal dysbiosis in COVID-19 patients and discuss its potential contribution to the respiratory symptoms through the gut-lung axis and the possibility of anti-COVID-19 therapy using probiotic intestinal bacteria.

Intestinal Dysbiosis Associated with Covid-19

Accumulating evidence has highlighted a cross-talk between the intestinal microbiota and the respiratory tract, termed the gutlung axis, in which gut microbe-derived components and metabolites modulate airway physiology and immunity [18-21]. Dysbiosis of the gut microbiota leads to aberrant immune tone in the respiratory epithelium and mucosa, which can trigger impaired immune responses to respiratory viral infections. Despite the important roles of the gut-lung axis in the antiviral immunity, only limited information is currently available concerning COVID-19-specific changes in the gut microbiome.

Intestinal Microbiota and Host’s Inflammatory Tone

Shen et al. previously conducted serum proteomic profiling of COVID-19 patients and established a blood proteomic risk score (PRS) for the prediction of the patients’ clinical outcomes [22]. In a recent preprint paper, the same research group has discovered a core set of gut bacterial taxa that are closely associated with the blood PRS in uninfected individuals [23]. The core taxa include genus Bacteroides, genus Streptococcus, genus Lactobacillus, family Ruminococcaceae, family Lachnospiraceae and order Clostridiales. Furthermore, the authors suggest a link between compositions of specific gut bacteria and susceptibility to severe COVID-19. Interestingly, a subset of the core taxa is correlated with the serum levels of proinflammatory cytokines, including TNF-α, IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13. In general, Bacteroides, Streptococcus, and Clostridiales are negatively correlated with the serum levels of tested cytokines, whereas Ruminococcus, Blautia, and Lactobacillus are positively correlated. Fecal metabolome and pathway analyses have identified 45 metabolites associated with the core taxa and revealed that the metabolites are involved in the amino acid-related metabolic pathways. The core bacterial taxa and the relevant metabolites may mediate between the gut microbiota and host inflammatory tone.

In the above-mentioned cytokine test, Ruminococcus gnavus has a strong positive correlation with most of the inflammatory cytokines examined. R.gnavus is commonly present in the gut microbiome of more than 90% of individuals [24]. Several microbiome studies have linked R. gnavus to inflammatory bowel disease, two conditions [Crohn's disease (CD) and ulcerative colitis (UC)] characterized by chronic mucosal inflammation of the gastrointestinal tract [25-31], suggesting the possible role of R. gnavus in inducing aberrant inflammatory responses in the intestinal mucosa. Notably, R. gnavus produces a proinflammatoryglucorhamnan polysaccharide that potently induces the Toll-like receptor 4–dependent secretion of TNF-α by dendritic cells [32]. In addition, R. gnavus can degrade mucin to utilize as a carbon source [29,33-35], which leads to the breakdown of the mucus layer and its barrier function. These metabolic properties may increase the exposure of the intestinal immune system to the glucorhamnan polysaccharide to enhance the host’s inflammatory tone. R. gnavus and/or other mucolytic bacteria may contribute to systemic hyperinflammation characteristic for COVID-19 patients.

Covid-19-Specific Alterations in Intestinal Microbiome

What are the gut microbiota changes that are specific for COVID-19 patients and how do the changes contribute to the COVID-19 symptoms? In a recent paper in Gastroenterology, Zuo and colleagues have clearly showed the COVID-19-specific alterations in gut bacterial community [36]. They performed shotgun metagenomic sequencing to analyze fecal samples from 15 hospitalized mild to critical patients and 15 uninfected individuals and found patient-specific dysbiosis of the gut microbiome. The observed dysbiosis is characterized by an increase in opportunistic pathogens and a decrease in beneficial symbionts. Interestingly, COVID-19 severity is positively correlated with the relative abundance of genus Coprobacillus, Clostridium ramosum, and Clostridium hathewayi and inversely with Faecalibacterium prausnitzii. F. prausnitzii is known to have an immunomodulatory property by producing an antiinflammatory protein and butyrate, a short-chain fatty acid that are essential for regulated intestinal immunity and epithelial barrier function [37]. Microbiome studies have associated the decreased relative abundance of F. prausnitzii with chronic inflammatory diseases such as CD [38,39], asthma [40,41], and obesity [42].

Zuo et al. have also presented interesting data that SARS-CoV-2 virus load in patients’ fecal samples is inversely correlated with Bacteroides dorei, B. thetaiotaomicron, B. massiliensis, and B. ovatus. In this context, it is worthy of note that these Bacteroides species can downregulate intestinal Ace2 expression when monocolonized in the colon of germ-free mice [43]. In particular, B. thetaiotaomicron, a dominant commensal anaerobe of the human intestine, is known to have an anti-inflammatory property and improve mucosal barrier function in rodents with chemically induced colitis, experimental models of CD [44-46]. Collectively, a decrease in B. thetaiotaomicron may lead to elevated Ace2 expression and declined anti-inflammatory activity, which may enhance SARS-CoV-2 infectivity and intestinal/systemic inflammatory tone.

There is another piece of evidence that gut microbiota can modulate colonic Ace2 expression and systemic inflammation in its host. Yang et al. have demonstrated that reconstitution of the gut microbiota in germ-free rats significantly decreased the colonic Ace2expression [47]. Serum metabolome analysis has revealed the higher levels of tryptophan metabolites, kynurenic acid and hydroxykynurenine, suggesting that these metabolites are implicated in the Ace2 regulation. The conventionalization also resulted in elevated systemic inflammation as evidenced by the increased serum level of lipocalin-2, also known as neutrophil gelatinase-associated lipocalin that is involved in innate immunity, and increased proportion of CD11b+ granulocytes. This study suggests that gut microbiota can modulate intestinal Ace2 expression and cause the immune dysregulation characteristic of COVID-19.

The two animal studies by Geva-Zatorsky et al. [43] and Yang et al. [47] raise further intriguing questions, and further studies are required to identify the bacterial species that release the metabolites with regulatory activity for Ace2 expression and elucidate the mechanism of the Ace2downregulation by the bacterial metabolites. More importantly, we should clarify whether the intestinal ACE2 expression is regulated by the similar microbiota-mediated mechanisms in human and whether the gut microbiota can also downregulate airway Ace2 expression.

Identification of patient-specific changes in the gut microbiota may enable us to make a microbiome-based diagnosis of COVID-19. Gu et al. have identified differences in the gut microbial signature between 30 moderate to severe COVID-19 patients and 30 healthy controls using 16S ribosomal RNA gene amplicon sequencing [48]. The differences include reduced bacterial diversity, higher relative abundance of opportunistic pathogens such as genera Streptococcus, Rothia, Veillonella and Actinomyces, and lower relative abundance of beneficial commensals in patients. In addition, a combination of five genera, Fusicatenibacter, Romboutsia, Intestinibacter, Actinomyces, and Erysipelato clostridium, was identified as a biomarker that can distinguish patients from healthy individuals. This finding suggests the potential diagnostic and therapeutic value of the specific gut microbiota for COVID-19 even though further validation studies are required.

Prophylactic and Therapeutic use of Probiotics in Covid-19

Components and metabolites derived from gut commensal bacteria play a key role in gut-lung axis-mediated regulation of the airway physiology and immunity [18-21]. Hence, there is the possibility that normalization of intestinal dysbiosis using probiotics is effective for the control of COVID-19. Previous animal and clinical studies have shown that oral intake of probiotic strains has protective and therapeutic efficacy in respiratory tract infections caused by RNA viruses such as influenza virus and rhinovirus [49-53]. However, there have been no reports of clinical trials focusing on the efficacy of probiotics in COVID-19 to date, and the rationale for using probiotics against COVID-19 remains unclear. Life-threatening symptoms and complications of COVID-19 are caused by hyperinflammation due to complex immune dysregulation involving neutrophilia, lymphocytopenia, declined T-cell immunity, and excessive production of inflammatory mediators [54-56]. It is therefore particularly notable that specific probiotic strains have the superior strain-specific immunomodulatory ability against respiratory viral infections.

There is a large body of evidence that Lactobacillus plantarum has a wide variety of immunomodulatory activity particularly upon the infection of seasonal and highly pathogenic influenza viruses. Oral administration of L. plantarum in mice alleviates inflammation upon viral infection [57], enhances NK cell activity [58], activates Th1-mediated immune responses and Th1 cytokine production (IL-12 and IFN-γ) [57-59], and notably, promotes IgA-mediated mucosal immunity in the small intestine and lung [59,60]. Similarly, randomized controlled trials have demonstrated that oral intake of L. plantarum reduces the risk of acquiring upper/lower respiratory tract infections and alleviates the respiratory symptoms [61-63]. These effects are shown to be associated with the immunomodulatory ability of L. plantarum, including activation of CD8+ T cells, enhancement of the phagocytic activity of granulocytes, reduction of plasma proinflammatory cytokines IFN-γ and TNF-α, and elevation of anti-inflammatory cytokines IL-4 and IL-10 [62,64]. These findings suggest that L. plantarum strengthens many aspects of the host defense mechanism against viral infection. On the basis of these findings, we have employed L. plantarum as anti- COVID-19 probiotics in our recent clinical trial for the immunological efficacy of probiotic lactic acid bacteria against COVID-19. The ongoing trial focusing on uninfected individuals is registered on University Hospital Medical Information Network-Clinical Trials Registry (UMIN-CTR, https: //www.umin.ac.jp/ctr/index.htm) under the trial number UMIN000040479.

Future Perspectives

The composition of the intestinal microbiota is highly variable among individuals, which may lead to high interpersonal disparity in the susceptibility to SARS-CoV-2 infection and the severity of COVID-19-associated symptoms. To test this possibility, we need to conduct comparative studies of gut microbiota profiles among COVID-19 patients with different severity and identify the compositional changes in specific intestinal bacteria that determine the disparity. Similarly, it would be interesting to investigate the differences in the composition of the gut microbiome among COVID-19 patients with or without extra-respiratory manifestations such as thrombosis, myocardial injury, renal damage, or neurological symptoms [65-67].

To develop probiotics for the management of COVID-19, more effort is required to isolate and/or identify more probiotic strains with the ability to reduce the viral load and suppress hyperinflammation via different mechanisms of action. Bacterial species outside of the genera Lactobacillus and Bifidobacterium should be explored. Randomized controlled trials with large cohorts aimed at evaluating the prophylactic and therapeutic use of probiotics are also essential. As described above, the respiratory tract is inhabited by commensal bacterial communities, and the respiratory microbiota is known to be closely associated with susceptibility to and severity of respiratory infections. In fact, metatranscriptomic studies have revealed that COVID-19 patients have more disrupted airway microbiome. Interestingly, bacteria within gut-specific genus Bacteroides are commonly and abundantly found in bronchoalveolar lavage fluid from ARDS patients. Indirect manipulation of the respiratory microbiome with oral probiotics may be a promising adjunctive therapy to alleviate lifethreatening hyperinflammation in COVID-19.

References

  1. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382: 1708-1720.
  2. Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA, Muley T, et al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J. 2020;39: 105114.
  3. Sungnak W, Huang N, Becavin C, Berg M, Queen R. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26: 681-687.
  4. Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020;181: 1016-1035.
  5. Gu J, Han B, Wang J. COVID-19: Gastrointestinal manifestations and potential fecal-oral transmission. Gastroenterol. 2020;158: 1518-1519.
  6. Wong SH, Lui RN, Sung JJ. Covid-19 and the digestive system. J GastroenterolHepatol. 2020;35: 744-748.
  7. Yang L, Tu L. Implications of gastrointestinal manifestations of COVID-19. Lancet GastroenterolHepatol. 2020;5: 629-630.
  8. Chen Y, Chen L, Deng Q, Zhang G, Wu K. The presence of SARS-CoV-2 RNA in the feces of COVID-19 patients. J Med Virol. 2020;92: 833-840.
  9. Kipkorir V, Cheruiyot I, Ngure B, Misiani M, Munguti J. Prolonged SARS-Cov-2 RNA detection in anal/rectal swabs and stool specimens in COVID-19 patients after negative conversion in nasopharyngeal RT-PCR test. J Med Virol. 2020.
  10. Zhou J, Li C, Liu X, Chiu MC, Zhao X. Infection of bat and human intestinal organoids by SARS-CoV-2. Nat Med. 2020.
  11. Lee JJ, Kopetz S, Vilar E, Shen JP, Chen K. Relative abundance of SARS-CoV-2 entry genes in the enterocytes of the lower gastrointestinal tract. Genes (Basel). 2020;11: 645.
  12. Zhang H, Kang Z, Gong H, Xu D, Wang J. Digestive system is a potential route of COVID-19: An analysis of single-cell coexpression pattern of key proteins in viral entry process. Gut. 2020;69: 1010
  13. Zang R, Gomez Castro MF, McCune BT, Zeng Q, Rothlauf PW.TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. SciImmunol. 2020;5: 3582.
  14. Lamers MM, Beumer J, van derVaart J, Knoops K, Puschhof J. SARS-CoV-2 productively infects human gut enterocytes. 2020.
  15. Xiao F, Tang M, Zheng X, Liu Y, Li X. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterol. 2020;158: 1831-1833.
  16. Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15: 55-63.
  17. Dumas A, Bernard L, Poquet Y, Lugo-Villarino G, Neyrolles O. The role of the lung microbiota and the gut-lung axis in respiratory infectious diseases. Cell Microbiol. 2018;20: 12966.
  18. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014;20: 159-166.
  19. Zhang D, Li S, Wang N, Tan HY, Zhang Z. The cross-talk between gut microbiota and lungs in common lung diseases. Front Microbiol. 2020;11: 301.
  20. Shen B, Yi X, Sun Y, Bi X, Du J. Proteomic and metabolomic characterization of COVID-19 patient sera. Cell. 2020.
  21. Gou W, Fu Y, Yue L, Chen GD, Cai X. Gut microbiota may underlie the predisposition of healthy individuals to COVID-19. 2020.
  22. Hall AB, Yassour M, Sauk J, Garner A, Jiang X. A novel ruminococcusgnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017;9: 103.
  23. Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K.Dysbiosis of the faecalmicrobiota in patients with Crohn's disease and their unaffected relatives. Gut. 2011;60: 631-637.
  24. Nishikawa J, Kudo T, Sakata S, Benno Y, Sugiyama T. Diversity of mucosa-associated microbiota in active and inactive ulcerative colitis. Scand J Gastroenterol.2009;44: 180-186.
  25. Nishino K, Nishida A, Inoue R, Kawada Y, Ohno M. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. J Gastroenterol. 2018;53: 95-106.
  26. Png CW, Linden SK, Gilshenan KS, Zoetendal EG, McSweeney CS. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol. 2010;105: 2420-2428.
  27. Prindiville T, Cantrell M, Wilson KH. Ribosomal DNA sequence analysis of mucosa-associated bacteria in Crohn's disease. Inflamm Bowel Dis. 2004;10: 824-833.
  28. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterol. 2010;139: 1844-1854.
  29. Henke MT, Kenny DJ, Cassilly CD, Vlamakis H, Xavier RJ.Ruminococcusgnavus, a member of the human gut microbiome associated with Crohn's disease, produces an inflammatory polysaccharide. ProcNatlAcad Sci. 2019;116: 12672-12677.
  30. Bell A, Brunt J, Crost E, Vaux L, Nepravishta R. Elucidation of a sialic acid metabolism pathway in mucus-foraging Ruminococcusgnavus unravels mechanisms of bacterial adaptation to the gut. Nat Microbiol. 2019;4: 2393-2404.
  31. Crost EH, Tailford LE, Le Gall G, Fons M, Henrissat B. Utilisation of mucinglycans by the human gut symbiontRuminococcusgnavus is strain-dependent. PLoS One. 2013;8: 76341.
  32. Tailford LE, Owen CD, Walshaw J, Crost EH, Hardy-Goddard J. Discovery of intramolecular trans-sialidases in human gut microbiota suggests novel mechanisms of mucosal adaptation. Nat Commun. 2015;6: 7624.
  33. Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterol. 2020.
  34. Quevrain E, Maubert MA, Michon C, Chain F, Marquant R. Identification of an anti-inflammatory protein from Faecalibacteriumprausnitzii, a commensal bacterium deficient in Crohn's disease. Gut. 2016;65: 415-425.
  35. Prosberg M, Bendtsen F, Vind I, Petersen AM, Gluud LL. The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta-analysis. Scand J Gastroenterol. 2016;51: 1407-1415.
  36. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG.Faecalibacteriumprausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. ProcNatlAcad Sci. 2008;105: 16731-16736.
  37. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S. Early infancy microbial and metabolic alterations affect risk of childhood asthma. SciTransl Med. 2015;7: 307.
  38. Wang Q, Li F, Liang B, Liang Y, Chen S. A metagenome-wide association study of gut microbiota in asthma in UK adults. BMC Microbiol. 2015;18: 114.
  39. Thingholm LB, Ruhlemann MC, Koch M, Fuqua B, Laucke G. Obese individuals with and without type 2 diabetes show different gut microbial functional capacity and composition. Cell Host Microbe. 2019;26: 252-264.
  40. Geva-Zatorsky N, Sefik E, Kua L, Pasman L, Tan TG. Mining the human gut microbiota for immunomodulatory organisms. Cell. 2017;168: 928-943.
  41. Delday M, Mulder I, Logan ET, Grant G. Bacteroidesthetaiotaomicron ameliorates colon inflammation in preclinical models of crohn's disease. Inflamm Bowel Dis. 2019;25: 85-96.
  42. Kelly D, Campbell JI, King TP, Grant G, Jansson EA. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nat Immunol. 2004;5: 104-112.
  43. Wrzosek L, Miquel S, Noordine ML, Bouet S, Joncquel Chevalier-Curt M, et al. Bacteroidesthetaiotaomicron and faecalibacteriumprausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013;11: 61.
  44. Yang T, Chakraborty S, Saha P, Mell B, Cheng X. Gnotobiotic rats reveal that gut microbiota regulates colonic mRNA of Ace2, the receptor for SARS-CoV-2 infectivity. Hypertension. 2020;76: 1-3.
  45. Gu S, Chen Y, Wu Z, Chen Y, Gao H. Alterations of the gut microbiota in patients with COVID-19 or H1N1 influenza. Clin Infect Dis. 2020;709.
  46. Lehtoranta L, Pitkaranta A, Korpela R. Probiotics in respiratory virus infections. Eur J ClinMicrobiol Infect Dis. 2014;33: 1289-1302.
  47. Lei WT, Shih PC, Liu SJ, Lin CY, Yeh TL. Effect of probiotics and prebiotics on immune response to influenza vaccination in adults: A systematic review and meta-analysis of randomized controlled trials. Nutrients. 2017;9: 1175.
  48. Luoto R, Ruuskanen O, Waris M, Kalliomaki M, Salminen S. Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: A randomized, placebo-controlled trial. J Allergy ClinImmunol. 2014;133: 405-413.
  49. Turner RB, Woodfolk JA, Borish L, Steinke JW, Patrie JT. Effect of probiotic on innate inflammatory response and viral shedding in experimental rhinovirus infection arandomised controlled trial. Benef Microbes. 2017;8: 207-215.
  50. Yeh TL, Shih PC, Liu SJ, Lin CH, Liu JM. The influence of prebiotic or probiotic supplementation on antibody titers after influenza vaccination: A systematic review and meta-analysis of randomized controlled trials. Drug Des DevelTher. 2018;12: 217-230.
  51. Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181: 1036-1045.
  52. Cao X (2020) COVID-19: Immunopathology and its implications for therapy. Nat Rev Immunol. 20: 269-270.
  53. Giamarellos-Bourboulis EJ, Netea MG, Rovina N, Akinosoglou K, Antoniadou A. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. 2020;27: 992-1000.
  54. Park MK, Ngo V, Kwon YM, Lee YT, Yoo S. Lactobacillus plantarum DK119 as a probiotic confers protection against influenza virus by modulating innate immunity. PLoS One. 2013;8: 75368.
  55. Takeda S, Takeshita M, Kikuchi Y, Dashnyam B, Kawahara S. Efficacy of oral administration of heat-killed probiotics from mongolian dairy products against influenza infection in mice: alleviation of influenza infection by its immunomodulatory activity through intestinal immunity. IntImmunopharmacol. 2011;11: 1976-1983.
  56. Kawashima T, Hayashi K, Kosaka A, Kawashima M, Igarashi T. Lactobacillus plantarum strain YU from fermented foods activates Th1 and protective immune responses. IntImmunopharmacol. 2011;11: 2017-2024.
  57. Kikuchi Y, Kunitoh-Asari A, Hayakawa K, Imai S, Kasuya K. Oral administration of Lactobacillus plantarum strain AYA enhances IgA secretion and provides survival protection against influenza virus infection in mice. PLoS One. 2014;9: 86416.
  58. Berggren A, LazouAhren I, Larsson N, Onning G. Randomised, double-blind and placebo-controlled study using new probiotic lactobacilli for strengthening the body immune defence against viral infections. Eur J Nutr. 2011;50: 203-210.
  59. Chong HX, Yusoff NAA, Hor YY, Lew LC, Jaafar MH. Lactobacillus plantarum DR7 improved upper respiratory tract infections via enhancing immune and inflammatory parameters: A randomized, double-blind, placebo-controlled study. J Dairy Sci. 2019;102: 4783-4797.
  60. Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548: 407-412.
  61. Rask C, Adlerberth I, Berggren A, Ahren IL, Wold AE. Differential effect on cell-mediated immunity in human volunteers after intake of different lactobacilli. ClinExpImmunol. 2013;172: 321-332.
  62. Man WH, de SteenhuijsenPiters WA, Bogaert D. The microbiota of the respiratory tract: gatekeeper to respiratory health. Nat Rev Microbiol. 2017;15: 259-270.
  63. Unger SA, Bogaert D. The respiratory microbiome and respiratory infections. J Infect. 2017;74(1): 84-88.
  64. Shen Z, Xiao Y, Kang L, Ma W, Shi L. Genomic diversity of SARS-CoV-2 in Coronavirus Disease 2019 patients. Clin Infect Dis. 2020;203.
  65. Zhang H, Ai JW, Yang W, Zhou X, He F. Metatranscriptomic characterization of COVID-19 identified a host transcriptional classifier associated with immune signaling. Clin Infect Dis. 2020;663.
  66. Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1: 16113.

Author Info

Yasunari Kageyama1,2, Tetsu Akiyama3 and Tsutomu Nakamura1,3*
 
1Takanawa Clinic, Minato-ku, Tokyo, Japan
2Tokai University Hospital, Isehara-shi, Kanagawa, Japan
3Laboratory of Molecular and Genetic Information, Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
 

Citation: Kageyama Y, Akiyama T, Nakamura T (2020) Intestinal Dysbiosis and Probiotics in COVID-19. J Clin Trials 10:421. doi: 10.35248/2167-0870.20.10.421

Received: 04-Jul-2020 Accepted: 17-Jul-2020 Published: 24-Jul-2020 , DOI: 10.35248/2167-0870.20.10.421

Copyright: © 2020 Kageyama Y, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top