Skip to main content
Top
Published in: Critical Care 1/2020

01-12-2020 | Probiotics | Review

The gut microbiome’s role in the development, maintenance, and outcomes of sepsis

Authors: Max W. Adelman, Michael H. Woodworth, Charles Langelier, Lindsay M. Busch, Jordan A. Kempker, Colleen S. Kraft, Greg S. Martin

Published in: Critical Care | Issue 1/2020

Login to get access

Abstract

The gut microbiome regulates a number of homeostatic mechanisms in the healthy host including immune function and gut barrier protection. Loss of normal gut microbial structure and function has been associated with diseases as diverse as Clostridioides difficile infection, asthma, and epilepsy. Recent evidence has also demonstrated a link between the gut microbiome and sepsis. In this review, we focus on three key areas of the interaction between the gut microbiome and sepsis. First, prior to sepsis onset, gut microbiome alteration increases sepsis susceptibility through several mechanisms, including (a) allowing for expansion of pathogenic intestinal bacteria, (b) priming the immune system for a robust pro-inflammatory response, and (c) decreasing production of beneficial microbial products such as short-chain fatty acids. Second, once sepsis is established, gut microbiome disruption worsens and increases susceptibility to end-organ dysfunction. Third, there is limited evidence that microbiome-based therapeutics, including probiotics and selective digestive decontamination, may decrease sepsis risk and improve sepsis outcomes in select patient populations, but concerns about safety have limited uptake. Case reports of a different microbiome-based therapy, fecal microbiota transplantation, have shown correlation with gut microbial structure restoration and decreased inflammatory response, but these results require further validation. While much of the evidence linking the gut microbiome and sepsis has been established in pre-clinical studies, clinical evidence is lacking in many areas. To address this, we outline a potential research agenda for further investigating the interaction between the gut microbiome and sepsis.
Literature
1.
go back to reference Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801–10.PubMedPubMedCentralCrossRef Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801–10.PubMedPubMedCentralCrossRef
2.
go back to reference Rhee C, Dantes R, Epstein L, Murphy DJ, Seymour CW, Iwashyna TJ, Kadri SS, Angus DC, Danner RL, Fiore AE, et al. Incidence and trends of sepsis in US hospitals using clinical vs claims data, 2009-2014. JAMA. 2017;318(13):1241–9.PubMedPubMedCentralCrossRef Rhee C, Dantes R, Epstein L, Murphy DJ, Seymour CW, Iwashyna TJ, Kadri SS, Angus DC, Danner RL, Fiore AE, et al. Incidence and trends of sepsis in US hospitals using clinical vs claims data, 2009-2014. JAMA. 2017;318(13):1241–9.PubMedPubMedCentralCrossRef
3.
go back to reference Kadri SS, Rhee C, Strich JR, Morales MK, Hohmann S, Menchaca J, Suffredini AF, Danner RL, Klompas M. Estimating ten-year trends in septic shock incidence and mortality in United States academic medical centers using clinical data. Chest. 2017;151(2):278–85.PubMedCrossRef Kadri SS, Rhee C, Strich JR, Morales MK, Hohmann S, Menchaca J, Suffredini AF, Danner RL, Klompas M. Estimating ten-year trends in septic shock incidence and mortality in United States academic medical centers using clinical data. Chest. 2017;151(2):278–85.PubMedCrossRef
6.
go back to reference Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2(2):135–43.PubMedCrossRef Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2(2):135–43.PubMedCrossRef
7.
go back to reference Shaw AG, Sim K, Randell P, Cox MJ, McClure ZE, Li MS, Donaldson H, Langford PR, Cookson WO, Moffatt MF, et al. Late-onset bloodstream infection and perturbed maturation of the gastrointestinal microbiota in premature infants. PLoS One. 2015;10(7):e0132923.PubMedPubMedCentralCrossRef Shaw AG, Sim K, Randell P, Cox MJ, McClure ZE, Li MS, Donaldson H, Langford PR, Cookson WO, Moffatt MF, et al. Late-onset bloodstream infection and perturbed maturation of the gastrointestinal microbiota in premature infants. PLoS One. 2015;10(7):e0132923.PubMedPubMedCentralCrossRef
8.
go back to reference Singer JR, Blosser EG, Zindl CL, Silberger DJ, Conlan S, Laufer VA, DiToro D, Deming C, Kumar R, Morrow CD, et al. Preventing dysbiosis of the neonatal mouse intestinal microbiome protects against late-onset sepsis. Nat Med. 2019;25(11):1772-1782. Singer JR, Blosser EG, Zindl CL, Silberger DJ, Conlan S, Laufer VA, DiToro D, Deming C, Kumar R, Morrow CD, et al. Preventing dysbiosis of the neonatal mouse intestinal microbiome protects against late-onset sepsis. Nat Med. 2019;25(11):1772-1782.
9.
go back to reference Graspeuntner S, Waschina S, Kunzel S, Twisselmann N, Rausch TK, Cloppenborg-Schmidt K, Zimmermann J, Viemann D, Herting E, Gopel W, et al. Gut dysbiosis with bacilli dominance and accumulation of fermentation products precedes late-onset sepsis in preterm infants. Clin Infect Dis. 2019;69(2):268–77.PubMedCrossRef Graspeuntner S, Waschina S, Kunzel S, Twisselmann N, Rausch TK, Cloppenborg-Schmidt K, Zimmermann J, Viemann D, Herting E, Gopel W, et al. Gut dysbiosis with bacilli dominance and accumulation of fermentation products precedes late-onset sepsis in preterm infants. Clin Infect Dis. 2019;69(2):268–77.PubMedCrossRef
10.
go back to reference Carl MA, Ndao IM, Springman AC, Manning SD, Johnson JR, Johnston BD, Burnham CA, Weinstock ES, Weinstock GM, Wylie TN, et al. Sepsis from the gut: the enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. Clin Infect Dis. 2014;58(9):1211–8.PubMedPubMedCentralCrossRef Carl MA, Ndao IM, Springman AC, Manning SD, Johnson JR, Johnston BD, Burnham CA, Weinstock ES, Weinstock GM, Wylie TN, et al. Sepsis from the gut: the enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. Clin Infect Dis. 2014;58(9):1211–8.PubMedPubMedCentralCrossRef
11.
go back to reference MacFie J, O'Boyle C, Mitchell CJ, Buckley PM, Johnstone D, Sudworth P. Gut origin of sepsis: a prospective study investigating associations between bacterial translocation, gastric microflora, and septic morbidity. Gut. 1999;45(2):223–8.PubMedPubMedCentralCrossRef MacFie J, O'Boyle C, Mitchell CJ, Buckley PM, Johnstone D, Sudworth P. Gut origin of sepsis: a prospective study investigating associations between bacterial translocation, gastric microflora, and septic morbidity. Gut. 1999;45(2):223–8.PubMedPubMedCentralCrossRef
14.
go back to reference Adhi FI, Littmann ER, Taur Y, Maloy MA, Markey KA, Fontana E, Amoretti LA, Wright R, Sanchez-Escamilla M, Castillo Flores N, et al. Pre-transplant fecal microbial diversity independently predicts critical illness after hematopoietic cell transplantation. Blood. 2019;134:3264.CrossRef Adhi FI, Littmann ER, Taur Y, Maloy MA, Markey KA, Fontana E, Amoretti LA, Wright R, Sanchez-Escamilla M, Castillo Flores N, et al. Pre-transplant fecal microbial diversity independently predicts critical illness after hematopoietic cell transplantation. Blood. 2019;134:3264.CrossRef
15.
go back to reference Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, Lee YJ, Dubin KA, Socci ND, Viale A, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55(7):905–14.PubMedPubMedCentralCrossRef Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, Lee YJ, Dubin KA, Socci ND, Viale A, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55(7):905–14.PubMedPubMedCentralCrossRef
16.
go back to reference Tamburini FB, Andermann TM, Tkachenko E, Senchyna F, Banaei N, Bhatt AS. Precision identification of diverse bloodstream pathogens in the gut microbiome. Nat Med. 2018;24(12):1809–14.PubMedPubMedCentralCrossRef Tamburini FB, Andermann TM, Tkachenko E, Senchyna F, Banaei N, Bhatt AS. Precision identification of diverse bloodstream pathogens in the gut microbiome. Nat Med. 2018;24(12):1809–14.PubMedPubMedCentralCrossRef
17.
go back to reference Freedberg DE, Zhou MJ, Cohen ME, Annavajhala MK, Khan S, Moscoso DI, Brooks C, Whittier S, Chong DH, Uhlemann AC, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med. 2018;44(8):1203–11.PubMedPubMedCentralCrossRef Freedberg DE, Zhou MJ, Cohen ME, Annavajhala MK, Khan S, Moscoso DI, Brooks C, Whittier S, Chong DH, Uhlemann AC, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med. 2018;44(8):1203–11.PubMedPubMedCentralCrossRef
18.
go back to reference Prescott HC, Dickson RP, Rogers MA, Langa KM, Iwashyna TJ. Hospitalization type and subsequent severe sepsis. Am J Respir Crit Care Med. 2015;192(5):581–8.PubMedPubMedCentralCrossRef Prescott HC, Dickson RP, Rogers MA, Langa KM, Iwashyna TJ. Hospitalization type and subsequent severe sepsis. Am J Respir Crit Care Med. 2015;192(5):581–8.PubMedPubMedCentralCrossRef
19.
go back to reference Baggs J, Jernigan JA, Halpin AL, Epstein L, Hatfield KM, McDonald LC. Risk of subsequent sepsis within 90 days after a hospital stay by type of antibiotic exposure. Clin Infect Dis. 2018;66(7):1004–12.PubMedCrossRef Baggs J, Jernigan JA, Halpin AL, Epstein L, Hatfield KM, McDonald LC. Risk of subsequent sepsis within 90 days after a hospital stay by type of antibiotic exposure. Clin Infect Dis. 2018;66(7):1004–12.PubMedCrossRef
20.
go back to reference Fay KT, Klingensmith NJ, Chen CW, Zhang W, Sun Y, Morrow KN, Liang Z, Burd EM, Ford ML, Coopersmith CM. The gut microbiome alters immunophenotype and survival from sepsis. FASEB J. 2019;33(10):11258-11269. Fay KT, Klingensmith NJ, Chen CW, Zhang W, Sun Y, Morrow KN, Liang Z, Burd EM, Ford ML, Coopersmith CM. The gut microbiome alters immunophenotype and survival from sepsis. FASEB J. 2019;33(10):11258-11269.
21.
go back to reference Gong S, Yan Z, Liu Z, Niu M, Fang H, Li N, Huang C, Li L, Chen G, Luo H, et al. Intestinal microbiota mediates the susceptibility to polymicrobial sepsis-induced liver injury by Granisetron generation in mice. Hepatology. 2019;69(4):1751–67.PubMedCrossRef Gong S, Yan Z, Liu Z, Niu M, Fang H, Li N, Huang C, Li L, Chen G, Luo H, et al. Intestinal microbiota mediates the susceptibility to polymicrobial sepsis-induced liver injury by Granisetron generation in mice. Hepatology. 2019;69(4):1751–67.PubMedCrossRef
22.
go back to reference Panpetch W, Somboonna N, Bulan DE, Issara-Amphorn J, Worasilchai N, Finkelman M, Chindamporn A, Palaga T, Tumwasorn S, Leelahavanichkul A. Gastrointestinal colonization of Candida albicans increases serum (1-->3)-beta-D-glucan, without Candidemia, and worsens cecal ligation and puncture sepsis in murine model. Shock. 2018;49(1):62–70.PubMedCrossRef Panpetch W, Somboonna N, Bulan DE, Issara-Amphorn J, Worasilchai N, Finkelman M, Chindamporn A, Palaga T, Tumwasorn S, Leelahavanichkul A. Gastrointestinal colonization of Candida albicans increases serum (1-->3)-beta-D-glucan, without Candidemia, and worsens cecal ligation and puncture sepsis in murine model. Shock. 2018;49(1):62–70.PubMedCrossRef
23.
go back to reference Chen G, Huang B, Fu S, Li B, Ran X, He D, Jiang L, Li Y, Liu B, Xie L, et al. G protein-coupled receptor 109A and host microbiota modulate intestinal epithelial integrity during sepsis. Front Immunol. 2018;9:2079.PubMedPubMedCentralCrossRef Chen G, Huang B, Fu S, Li B, Ran X, He D, Jiang L, Li Y, Liu B, Xie L, et al. G protein-coupled receptor 109A and host microbiota modulate intestinal epithelial integrity during sepsis. Front Immunol. 2018;9:2079.PubMedPubMedCentralCrossRef
24.
go back to reference Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza a virus infection. Proc Natl Acad Sci U S A. 2011;108(13):5354–9.PubMedPubMedCentralCrossRef Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza a virus infection. Proc Natl Acad Sci U S A. 2011;108(13):5354–9.PubMedPubMedCentralCrossRef
25.
go back to reference Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJ, de Boer JD, Hoogendijk AJ, de Beer R, de Vos A, Belzer C, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 2016, 65(4):575–583. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJ, de Boer JD, Hoogendijk AJ, de Beer R, de Vos A, Belzer C, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 2016, 65(4):575–583.
26.
go back to reference Lankelma JM, Cranendonk DR, Belzer C, de Vos AF, de Vos WM, van der Poll T, Wiersinga WJ. Antibiotic-induced gut microbiota disruption during human endotoxemia: a randomised controlled study. Gut. 2017;66(9):1623–30.PubMedCrossRef Lankelma JM, Cranendonk DR, Belzer C, de Vos AF, de Vos WM, van der Poll T, Wiersinga WJ. Antibiotic-induced gut microbiota disruption during human endotoxemia: a randomised controlled study. Gut. 2017;66(9):1623–30.PubMedCrossRef
27.
28.
go back to reference Woodworth MH, Hayden MK, Young VB, Kwon JH. The role of fecal microbiota transplantation in reducing intestinal colonization with antibiotic resistant organisms: the current landscape and future directions. Open Forum Infect Dis. 2019;6(7):ofz288. Woodworth MH, Hayden MK, Young VB, Kwon JH. The role of fecal microbiota transplantation in reducing intestinal colonization with antibiotic resistant organisms: the current landscape and future directions. Open Forum Infect Dis. 2019;6(7):ofz288.
29.
go back to reference Kim SG, Becattini S, Moody TU, Shliaha PV, Littmann ER, Seok R, Gjonbalaj M, Eaton V, Fontana E, Amoretti L, et al. Microbiota-derived lantibiotic restores resistance against vancomycin-resistant Enterococcus. Nature. 2019;572(7771):665–9.PubMedPubMedCentralCrossRef Kim SG, Becattini S, Moody TU, Shliaha PV, Littmann ER, Seok R, Gjonbalaj M, Eaton V, Fontana E, Amoretti L, et al. Microbiota-derived lantibiotic restores resistance against vancomycin-resistant Enterococcus. Nature. 2019;572(7771):665–9.PubMedPubMedCentralCrossRef
30.
go back to reference Ayres JS, Trinidad NJ, Vance RE. Lethal inflammasome activation by a multidrug-resistant pathobiont upon antibiotic disruption of the microbiota. Nat Med. 2012;18(5):799–806.PubMedPubMedCentralCrossRef Ayres JS, Trinidad NJ, Vance RE. Lethal inflammasome activation by a multidrug-resistant pathobiont upon antibiotic disruption of the microbiota. Nat Med. 2012;18(5):799–806.PubMedPubMedCentralCrossRef
31.
go back to reference Hyoju SK, Zaborin A, Keskey R, Sharma A, Arnold W, van den Berg F, Kim SM, Gottel N, Bethel C, Charnot-Katsikas A, et al. Mice fed an obesogenic western diet, administered antibiotics, and subjected to a sterile surgical procedure develop lethal septicemia with multidrug-resistant pathobionts. MBio. 2019;10(4):e00903-19. Hyoju SK, Zaborin A, Keskey R, Sharma A, Arnold W, van den Berg F, Kim SM, Gottel N, Bethel C, Charnot-Katsikas A, et al. Mice fed an obesogenic western diet, administered antibiotics, and subjected to a sterile surgical procedure develop lethal septicemia with multidrug-resistant pathobionts. MBio. 2019;10(4):e00903-19.
33.
go back to reference Deshmukh HS, Liu Y, Menkiti OR, Mei J, Dai N, O'Leary CE, Oliver PM, Kolls JK, Weiser JN, Worthen GS. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat Med. 2014;20(5):524–30.PubMedPubMedCentralCrossRef Deshmukh HS, Liu Y, Menkiti OR, Mei J, Dai N, O'Leary CE, Oliver PM, Kolls JK, Weiser JN, Worthen GS. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat Med. 2014;20(5):524–30.PubMedPubMedCentralCrossRef
34.
go back to reference Khosravi A, Yanez A, Price JG, Chow A, Merad M, Goodridge HS, Mazmanian SK. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe. 2014;15(3):374–81.PubMedPubMedCentralCrossRef Khosravi A, Yanez A, Price JG, Chow A, Merad M, Goodridge HS, Mazmanian SK. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe. 2014;15(3):374–81.PubMedPubMedCentralCrossRef
35.
go back to reference Cabrera-Perez J, Babcock JC, Dileepan T, Murphy KA, Kucaba TA, Badovinac VP, Griffith TS. Gut microbial membership modulates CD4 T cell reconstitution and function after sepsis. J Immunol. 2016;197(5):1692–8.PubMedCrossRef Cabrera-Perez J, Babcock JC, Dileepan T, Murphy KA, Kucaba TA, Badovinac VP, Griffith TS. Gut microbial membership modulates CD4 T cell reconstitution and function after sepsis. J Immunol. 2016;197(5):1692–8.PubMedCrossRef
36.
go back to reference Wilmore JR, Gaudette BT, Gomez Atria D, Hashemi T, Jones DD, Gardner CA, Cole SD, Misic AM, Beiting DP, Allman D. Commensal microbes induce serum IgA responses that protect against polymicrobial sepsis. Cell Host Microbe. 2018;23(3):302–11 e303.PubMedPubMedCentralCrossRef Wilmore JR, Gaudette BT, Gomez Atria D, Hashemi T, Jones DD, Gardner CA, Cole SD, Misic AM, Beiting DP, Allman D. Commensal microbes induce serum IgA responses that protect against polymicrobial sepsis. Cell Host Microbe. 2018;23(3):302–11 e303.PubMedPubMedCentralCrossRef
37.
go back to reference Morowitz MJ, Di Caro V, Pang D, Cummings J, Firek B, Rogers MB, Ranganathan S, Clark RSB, Aneja RK. Dietary supplementation with nonfermentable fiber alters the gut microbiota and confers protection in murine models of sepsis. Crit Care Med. 2017;45(5):e516–23.PubMedPubMedCentralCrossRef Morowitz MJ, Di Caro V, Pang D, Cummings J, Firek B, Rogers MB, Ranganathan S, Clark RSB, Aneja RK. Dietary supplementation with nonfermentable fiber alters the gut microbiota and confers protection in murine models of sepsis. Crit Care Med. 2017;45(5):e516–23.PubMedPubMedCentralCrossRef
38.
go back to reference Ikeda M, Shimizu K, Ogura H, Kurakawa T, Umemoto E, Motooka D, Nakamura S, Ichimaru N, Takeda K, Takahara S, et al. Hydrogen-rich saline regulates intestinal barrier dysfunction, dysbiosis, and bacterial translocation in a murine model of sepsis. Shock. 2018;50(6):640–7.PubMedCrossRef Ikeda M, Shimizu K, Ogura H, Kurakawa T, Umemoto E, Motooka D, Nakamura S, Ichimaru N, Takeda K, Takahara S, et al. Hydrogen-rich saline regulates intestinal barrier dysfunction, dysbiosis, and bacterial translocation in a murine model of sepsis. Shock. 2018;50(6):640–7.PubMedCrossRef
39.
go back to reference Liu Z, Li N, Fang H, Chen X, Guo Y, Gong S, Niu M, Zhou H, Jiang Y, Chang P, et al. Enteric dysbiosis is associated with sepsis in patients. FASEB J. 2019;33(11):12299-12310. Liu Z, Li N, Fang H, Chen X, Guo Y, Gong S, Niu M, Zhou H, Jiang Y, Chang P, et al. Enteric dysbiosis is associated with sepsis in patients. FASEB J. 2019;33(11):12299-12310.
40.
go back to reference Kim D, Kang H. Exercise training modifies gut microbiota with attenuated host responses to sepsis in wild-type mice. FASEB J. 2019;33(4):5772–81.PubMedCrossRef Kim D, Kang H. Exercise training modifies gut microbiota with attenuated host responses to sepsis in wild-type mice. FASEB J. 2019;33(4):5772–81.PubMedCrossRef
41.
go back to reference Li S, Lv J, Li J, Zhao Z, Guo H, Zhang Y, Cheng S, Sun J, Pan H, Fan S, et al. Intestinal microbiota impact sepsis associated encephalopathy via the vagus nerve. Neurosci Lett. 2018;662:98–104.PubMedCrossRef Li S, Lv J, Li J, Zhao Z, Guo H, Zhang Y, Cheng S, Sun J, Pan H, Fan S, et al. Intestinal microbiota impact sepsis associated encephalopathy via the vagus nerve. Neurosci Lett. 2018;662:98–104.PubMedCrossRef
42.
go back to reference Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167(4):1125–36 e1128.PubMedPubMedCentralCrossRef Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167(4):1125–36 e1128.PubMedPubMedCentralCrossRef
43.
go back to reference Li Q, Wang C, Tang C, He Q, Zhao X, Li N, Li J. Successful treatment of severe sepsis and diarrhea after vagotomy utilizing fecal microbiota transplantation: a case report. Crit Care. 2015;19:37.PubMedPubMedCentralCrossRef Li Q, Wang C, Tang C, He Q, Zhao X, Li N, Li J. Successful treatment of severe sepsis and diarrhea after vagotomy utilizing fecal microbiota transplantation: a case report. Crit Care. 2015;19:37.PubMedPubMedCentralCrossRef
44.
go back to reference Singer BH, Dickson RP, Denstaedt SJ, Newstead MW, Kim K, Falkowski NR, Erb-Downward JR, Schmidt TM, Huffnagle GB, Standiford TJ. Bacterial dissemination to the brain in sepsis. Am J Respir Crit Care Med. 2018;197(6):747–56.PubMedPubMedCentralCrossRef Singer BH, Dickson RP, Denstaedt SJ, Newstead MW, Kim K, Falkowski NR, Erb-Downward JR, Schmidt TM, Huffnagle GB, Standiford TJ. Bacterial dissemination to the brain in sepsis. Am J Respir Crit Care Med. 2018;197(6):747–56.PubMedPubMedCentralCrossRef
45.
go back to reference Cernada M, Bauerl C, Serna E, Collado MC, Martinez GP, Vento M. Sepsis in preterm infants causes alterations in mucosal gene expression and microbiota profiles compared to non-septic twins. Sci Rep. 2016;6:25497.PubMedPubMedCentralCrossRef Cernada M, Bauerl C, Serna E, Collado MC, Martinez GP, Vento M. Sepsis in preterm infants causes alterations in mucosal gene expression and microbiota profiles compared to non-septic twins. Sci Rep. 2016;6:25497.PubMedPubMedCentralCrossRef
46.
go back to reference Shimizu K, Ogura H, Goto M, Asahara T, Nomoto K, Morotomi M, Yoshiya K, Matsushima A, Sumi Y, Kuwagata Y, et al. Altered gut flora and environment in patients with severe SIRS. J Trauma. 2006;60(1):126–33.PubMedCrossRef Shimizu K, Ogura H, Goto M, Asahara T, Nomoto K, Morotomi M, Yoshiya K, Matsushima A, Sumi Y, Kuwagata Y, et al. Altered gut flora and environment in patients with severe SIRS. J Trauma. 2006;60(1):126–33.PubMedCrossRef
47.
go back to reference Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260–8.PubMedPubMedCentralCrossRef Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis. 2013;13(3):260–8.PubMedPubMedCentralCrossRef
48.
go back to reference Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. Proc Nutr Soc. 2003;62(1):67–72.PubMedCrossRef Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. Proc Nutr Soc. 2003;62(1):67–72.PubMedCrossRef
49.
go back to reference Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50.PubMedCrossRef Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50.PubMedCrossRef
50.
go back to reference Parada Venegas D, De la Fuente MK, Landskron G, Gonzalez MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277.PubMedPubMedCentralCrossRef Parada Venegas D, De la Fuente MK, Landskron G, Gonzalez MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277.PubMedPubMedCentralCrossRef
51.
go back to reference Zhao Y, Chen F, Wu W, Sun M, Bilotta AJ, Yao S, Xiao Y, Huang X, Eaves-Pyles TD, Golovko G, et al. GPR43 mediates microbiota metabolite SCFA regulation of antimicrobial peptide expression in intestinal epithelial cells via activation of mTOR and STAT3. Mucosal Immunol. 2018;11(3):752–62.PubMedPubMedCentralCrossRef Zhao Y, Chen F, Wu W, Sun M, Bilotta AJ, Yao S, Xiao Y, Huang X, Eaves-Pyles TD, Golovko G, et al. GPR43 mediates microbiota metabolite SCFA regulation of antimicrobial peptide expression in intestinal epithelial cells via activation of mTOR and STAT3. Mucosal Immunol. 2018;11(3):752–62.PubMedPubMedCentralCrossRef
52.
go back to reference Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469(7331):543–7.PubMedCrossRef Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469(7331):543–7.PubMedCrossRef
53.
go back to reference Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe. 2015;17(5):662–71.PubMedPubMedCentralCrossRef Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe. 2015;17(5):662–71.PubMedPubMedCentralCrossRef
54.
go back to reference Stevens BR, Goel R, Seungbum K, Richards EM, Holbert RC, Pepine CJ, Raizada MK. Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut. 2018;67(8):1555–7.PubMedCrossRef Stevens BR, Goel R, Seungbum K, Richards EM, Holbert RC, Pepine CJ, Raizada MK. Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut. 2018;67(8):1555–7.PubMedCrossRef
55.
go back to reference Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother. 2014;69(4):881–91.PubMedCrossRef Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother. 2014;69(4):881–91.PubMedCrossRef
56.
go back to reference Zaborin A, Smith D, Garfield K, Quensen J, Shakhsheer B, Kade M, Tirrell M, Tiedje J, Gilbert JA, Zaborina O, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio. 2014;5(5):e01361–14.PubMedPubMedCentralCrossRef Zaborin A, Smith D, Garfield K, Quensen J, Shakhsheer B, Kade M, Tirrell M, Tiedje J, Gilbert JA, Zaborina O, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio. 2014;5(5):e01361–14.PubMedPubMedCentralCrossRef
57.
go back to reference Dahlgren AF, Pan A, Lam V, Gouthro KC, Simpson PM, Salzman NH, Nghiem-Rao TH. Longitudinal changes in the gut microbiome of infants on total parenteral nutrition. Pediatr Res. 2019;86(1):107–14.PubMedPubMedCentralCrossRef Dahlgren AF, Pan A, Lam V, Gouthro KC, Simpson PM, Salzman NH, Nghiem-Rao TH. Longitudinal changes in the gut microbiome of infants on total parenteral nutrition. Pediatr Res. 2019;86(1):107–14.PubMedPubMedCentralCrossRef
58.
go back to reference Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, Vork L, Tigchelaar EF, Jankipersadsing SA, Cenit MC, Harmsen HJ, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65(5):740–8.PubMedCrossRef Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, Vork L, Tigchelaar EF, Jankipersadsing SA, Cenit MC, Harmsen HJ, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65(5):740–8.PubMedCrossRef
59.
go back to reference Lankelma JM, van Vught LA, Belzer C, Schultz MJ, van der Poll T, de Vos WM, Wiersinga WJ. Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation: a pilot study. Intensive Care Med. 2017;43(1):59–68.PubMedCrossRef Lankelma JM, van Vught LA, Belzer C, Schultz MJ, van der Poll T, de Vos WM, Wiersinga WJ. Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation: a pilot study. Intensive Care Med. 2017;43(1):59–68.PubMedCrossRef
60.
go back to reference McDonald D, Ackermann G, Khailova L, Baird C, Heyland D, Kozar R, Lemieux M, Derenski K, King J, Vis-Kampen C, et al. Extreme Dysbiosis of the Microbiome in Critical Illness. mSphere. 2016;1(4):e00199-16. McDonald D, Ackermann G, Khailova L, Baird C, Heyland D, Kozar R, Lemieux M, Derenski K, King J, Vis-Kampen C, et al. Extreme Dysbiosis of the Microbiome in Critical Illness. mSphere. 2016;1(4):e00199-16.
61.
go back to reference Wan YD, Zhu RX, Wu ZQ, Lyu SY, Zhao LX, Du ZJ, Pan XT. Gut microbiota disruption in septic shock patients: a pilot study. Med Sci Monit. 2018;24:8639–46.PubMedPubMedCentralCrossRef Wan YD, Zhu RX, Wu ZQ, Lyu SY, Zhao LX, Du ZJ, Pan XT. Gut microbiota disruption in septic shock patients: a pilot study. Med Sci Monit. 2018;24:8639–46.PubMedPubMedCentralCrossRef
62.
go back to reference Ravi A, Halstead FD, Bamford A, Casey A, Thomson NM, van Schaik W, Snelson C, Goulden R, Foster-Nyarko E, Savva GM, et al. Loss of microbial diversity and pathogen domination of the gut microbiota in critically ill patients. Microb Genom. 2019;5(9):e000293. Ravi A, Halstead FD, Bamford A, Casey A, Thomson NM, van Schaik W, Snelson C, Goulden R, Foster-Nyarko E, Savva GM, et al. Loss of microbial diversity and pathogen domination of the gut microbiota in critically ill patients. Microb Genom. 2019;5(9):e000293.
63.
go back to reference Shimizu K, Ogura H, Hamasaki T, Goto M, Tasaki O, Asahara T, Nomoto K, Morotomi M, Matsushima A, Kuwagata Y, et al. Altered gut flora are associated with septic complications and death in critically ill patients with systemic inflammatory response syndrome. Dig Dis Sci. 2011;56(4):1171–7.PubMedCrossRef Shimizu K, Ogura H, Hamasaki T, Goto M, Tasaki O, Asahara T, Nomoto K, Morotomi M, Matsushima A, Kuwagata Y, et al. Altered gut flora are associated with septic complications and death in critically ill patients with systemic inflammatory response syndrome. Dig Dis Sci. 2011;56(4):1171–7.PubMedCrossRef
64.
go back to reference Yamada T, Shimizu K, Ogura H, Asahara T, Nomoto K, Yamakawa K, Hamasaki T, Nakahori Y, Ohnishi M, Kuwagata Y, et al. Rapid and sustained long-term decrease of fecal short-chain fatty acids in critically ill patients with systemic inflammatory response syndrome. JPEN J Parenter Enteral Nutr. 2015;39(5):569–77.PubMedCrossRef Yamada T, Shimizu K, Ogura H, Asahara T, Nomoto K, Yamakawa K, Hamasaki T, Nakahori Y, Ohnishi M, Kuwagata Y, et al. Rapid and sustained long-term decrease of fecal short-chain fatty acids in critically ill patients with systemic inflammatory response syndrome. JPEN J Parenter Enteral Nutr. 2015;39(5):569–77.PubMedCrossRef
65.
go back to reference Al-Harbi NO, Nadeem A, Ahmad SF, Alotaibi MR, AlAsmari AF, Alanazi WA, Al-Harbi MM, El-Sherbeeny AM, Ibrahim KE. Short chain fatty acid, acetate ameliorates sepsis-induced acute kidney injury by inhibition of NADPH oxidase signaling in T cells. Int Immunopharmacol. 2018;58:24–31.PubMedCrossRef Al-Harbi NO, Nadeem A, Ahmad SF, Alotaibi MR, AlAsmari AF, Alanazi WA, Al-Harbi MM, El-Sherbeeny AM, Ibrahim KE. Short chain fatty acid, acetate ameliorates sepsis-induced acute kidney injury by inhibition of NADPH oxidase signaling in T cells. Int Immunopharmacol. 2018;58:24–31.PubMedCrossRef
66.
go back to reference Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ, Huffnagle GB. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1(10):16113.PubMedPubMedCentralCrossRef Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ, Huffnagle GB. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1(10):16113.PubMedPubMedCentralCrossRef
67.
go back to reference Harris B, Morjaria SM, Littmann ER, Geyer AI, Stover DE, Barker JN, Giralt SA, Taur Y, Pamer EG. Gut microbiota predict pulmonary infiltrates after allogeneic hematopoietic cell transplantation. Am J Respir Crit Care Med. 2016;194(4):450–63.PubMedPubMedCentralCrossRef Harris B, Morjaria SM, Littmann ER, Geyer AI, Stover DE, Barker JN, Giralt SA, Taur Y, Pamer EG. Gut microbiota predict pulmonary infiltrates after allogeneic hematopoietic cell transplantation. Am J Respir Crit Care Med. 2016;194(4):450–63.PubMedPubMedCentralCrossRef
68.
69.
go back to reference de Smet AM, Kluytmans JA, Cooper BS, Mascini EM, Benus RF, van der Werf TS, van der Hoeven JG, Pickkers P, Bogaers-Hofman D, van der Meer NJ, et al. Decontamination of the digestive tract and oropharynx in ICU patients. N Engl J Med. 2009;360(1):20–31.PubMedCrossRef de Smet AM, Kluytmans JA, Cooper BS, Mascini EM, Benus RF, van der Werf TS, van der Hoeven JG, Pickkers P, Bogaers-Hofman D, van der Meer NJ, et al. Decontamination of the digestive tract and oropharynx in ICU patients. N Engl J Med. 2009;360(1):20–31.PubMedCrossRef
70.
go back to reference Huttner B, Haustein T, Uckay I, Renzi G, Stewardson A, Schaerrer D, Agostinho A, Andremont A, Schrenzel J, Pittet D, et al. Decolonization of intestinal carriage of extended-spectrum beta-lactamase-producing Enterobacteriaceae with oral colistin and neomycin: a randomized, double-blind, placebo-controlled trial. J Antimicrob Chemother. 2013;68(10):2375–82.PubMedCrossRef Huttner B, Haustein T, Uckay I, Renzi G, Stewardson A, Schaerrer D, Agostinho A, Andremont A, Schrenzel J, Pittet D, et al. Decolonization of intestinal carriage of extended-spectrum beta-lactamase-producing Enterobacteriaceae with oral colistin and neomycin: a randomized, double-blind, placebo-controlled trial. J Antimicrob Chemother. 2013;68(10):2375–82.PubMedCrossRef
71.
go back to reference de Jonge E, de Wilde RBP, Juffermans NP, Oostdijk EAN, Bernards AT, van Essen EHR, Kuijper EJ, Visser CE, Kesecioglu J, Bonten MJM. Carriage of antibiotic-resistant Gram-negative bacteria after discontinuation of selective decontamination of the digestive tract (SDD) or selective oropharyngeal decontamination (SOD). Crit Care. 2018;22(1):243.PubMedPubMedCentralCrossRef de Jonge E, de Wilde RBP, Juffermans NP, Oostdijk EAN, Bernards AT, van Essen EHR, Kuijper EJ, Visser CE, Kesecioglu J, Bonten MJM. Carriage of antibiotic-resistant Gram-negative bacteria after discontinuation of selective decontamination of the digestive tract (SDD) or selective oropharyngeal decontamination (SOD). Crit Care. 2018;22(1):243.PubMedPubMedCentralCrossRef
72.
go back to reference Oostdijk EA, de Smet AM, Blok HE, Thieme Groen ES, van Asselt GJ, Benus RF, Bernards SA, Frenay IH, Jansz AR, de Jongh BM, et al. Ecological effects of selective decontamination on resistant gram-negative bacterial colonization. Am J Respir Crit Care Med. 2010;181(5):452–7.PubMedCrossRef Oostdijk EA, de Smet AM, Blok HE, Thieme Groen ES, van Asselt GJ, Benus RF, Bernards SA, Frenay IH, Jansz AR, de Jongh BM, et al. Ecological effects of selective decontamination on resistant gram-negative bacterial colonization. Am J Respir Crit Care Med. 2010;181(5):452–7.PubMedCrossRef
73.
go back to reference Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, Baccaglini L, Mohapatra A, Mohapatra SS, Misra PR, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548(7668):407–12.PubMedCrossRef Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, Baccaglini L, Mohapatra A, Mohapatra SS, Misra PR, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548(7668):407–12.PubMedCrossRef
74.
go back to reference Costeloe K, Hardy P, Juszczak E, Wilks M, Millar MR. Probiotics in preterm infants study collaborative G: Bifidobacterium breve BBG-001 in very preterm infants: a randomised controlled phase 3 trial. Lancet. 2016;387(10019):649–60.PubMedCrossRef Costeloe K, Hardy P, Juszczak E, Wilks M, Millar MR. Probiotics in preterm infants study collaborative G: Bifidobacterium breve BBG-001 in very preterm infants: a randomised controlled phase 3 trial. Lancet. 2016;387(10019):649–60.PubMedCrossRef
75.
go back to reference Millar M, Seale J, Greenland M, Hardy P, Juszczak E, Wilks M, Panton N, Costeloe K, Wade WG. The microbiome of infants recruited to a randomised placebo-controlled probiotic trial (PiPS trial). EBioMedicine. 2017;20:255–62.PubMedPubMedCentralCrossRef Millar M, Seale J, Greenland M, Hardy P, Juszczak E, Wilks M, Panton N, Costeloe K, Wade WG. The microbiome of infants recruited to a randomised placebo-controlled probiotic trial (PiPS trial). EBioMedicine. 2017;20:255–62.PubMedPubMedCentralCrossRef
76.
go back to reference Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Crit Care. 2016;19:262.PubMedCrossRef Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Crit Care. 2016;19:262.PubMedCrossRef
77.
go back to reference Yelin I, Flett KB, Merakou C, Mehrotra P, Stam J, Snesrud E, Hinkle M, Lesho E, McGann P, McAdam AJ, et al. Genomic and epidemiological evidence of bacterial transmission from probiotic capsule to blood in ICU patients. Nat Med. 2019. Yelin I, Flett KB, Merakou C, Mehrotra P, Stam J, Snesrud E, Hinkle M, Lesho E, McGann P, McAdam AJ, et al. Genomic and epidemiological evidence of bacterial transmission from probiotic capsule to blood in ICU patients. Nat Med. 2019.
78.
go back to reference Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, Brown EM, Graham DB, Xavier RJ, Moon JJ, et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science. 2019;364(6446):1179–84.PubMedPubMedCentralCrossRef Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, Brown EM, Graham DB, Xavier RJ, Moon JJ, et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science. 2019;364(6446):1179–84.PubMedPubMedCentralCrossRef
79.
go back to reference Caballero S, Kim S, Carter RA, Leiner IM, Susac B, Miller L, Kim GJ, Ling L, Pamer EG. Cooperating commensals restore colonization resistance to vancomycin-resistant Enterococcus faecium. Cell Host Microbe. 2017;21(5):592–602 e594.PubMedPubMedCentralCrossRef Caballero S, Kim S, Carter RA, Leiner IM, Susac B, Miller L, Kim GJ, Ling L, Pamer EG. Cooperating commensals restore colonization resistance to vancomycin-resistant Enterococcus faecium. Cell Host Microbe. 2017;21(5):592–602 e594.PubMedPubMedCentralCrossRef
80.
go back to reference Li Q, Wang C, Tang C, He Q, Zhao X, Li N, Li J. Therapeutic modulation and reestablishment of the intestinal microbiota with fecal microbiota transplantation resolves sepsis and diarrhea in a patient. Am J Gastroenterol. 2014;109(11):1832–4. Li Q, Wang C, Tang C, He Q, Zhao X, Li N, Li J. Therapeutic modulation and reestablishment of the intestinal microbiota with fecal microbiota transplantation resolves sepsis and diarrhea in a patient. Am J Gastroenterol. 2014;109(11):1832–4.
81.
go back to reference Gopalsamy SN, Sherman A, Woodworth MH, Lutgring JD, Kraft CS. Fecal microbiota transplant for multidrug-resistant organism decolonization administered during septic shock. Infect Control Hosp Epidemiol. 2018;39(4):490–2.PubMedPubMedCentralCrossRef Gopalsamy SN, Sherman A, Woodworth MH, Lutgring JD, Kraft CS. Fecal microbiota transplant for multidrug-resistant organism decolonization administered during septic shock. Infect Control Hosp Epidemiol. 2018;39(4):490–2.PubMedPubMedCentralCrossRef
82.
go back to reference Wei Y, Yang J, Wang J, Yang Y, Huang J, Gong H, Cui H, Chen D. Successful treatment with fecal microbiota transplantation in patients with multiple organ dysfunction syndrome and diarrhea following severe sepsis. Crit Care. 2016;20(1):332.PubMedPubMedCentralCrossRef Wei Y, Yang J, Wang J, Yang Y, Huang J, Gong H, Cui H, Chen D. Successful treatment with fecal microbiota transplantation in patients with multiple organ dysfunction syndrome and diarrhea following severe sepsis. Crit Care. 2016;20(1):332.PubMedPubMedCentralCrossRef
83.
go back to reference DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, Turbett S, Chung RT, Chen YB, Hohmann EL. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381(21):2043-050. DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, Turbett S, Chung RT, Chen YB, Hohmann EL. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381(21):2043-050.
Metadata
Title
The gut microbiome’s role in the development, maintenance, and outcomes of sepsis
Authors
Max W. Adelman
Michael H. Woodworth
Charles Langelier
Lindsay M. Busch
Jordan A. Kempker
Colleen S. Kraft
Greg S. Martin
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Critical Care / Issue 1/2020
Electronic ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-02989-1

Other articles of this Issue 1/2020

Critical Care 1/2020 Go to the issue