Skip to main content
Surgical Infections logoLink to Surgical Infections
. 2023 Mar 30;24(3):250–257. doi: 10.1089/sur.2022.420

Gut Microbiome in Sepsis

Nathan J Klingensmith 1, Craig M Coopersmith 2,
PMCID: PMC10061326  PMID: 37010964

Abstract

The gut has been hypothesized to be the “motor” of multiple organ dysfunction in sepsis. Although there are multiple ways in which the gut can drive systemic inflammation, increasing evidence suggests that the intestinal microbiome plays a more substantial role than previously appreciated. An English language literature review was performed to summarize the current knowledge of sepsis-induced gut microbiome dysbiosis. Conversion of a normal microbiome to a pathobiome in the setting of sepsis is associated with worsened mortality. Changes in microbiome composition and diversity signal the intestinal epithelium and immune system resulting in increased intestinal permeability and a dysregulated immune response to sepsis. Clinical approaches to return to microbiome homeostasis may be theoretically possible through a variety of methods including probiotics, prebiotics, fecal microbial transplant, and selective decontamination of the digestive tract. However, more research is required to determine the efficacy (if any) of targeting the microbiome for therapeutic gain. The gut microbiome rapidly loses diversity with emergence of virulent bacteria in sepsis. Restoring normal commensal bacterial diversity through various therapies may be an avenue to improve sepsis mortality.

Keywords: gut, intestine, microbiome, pathobiome, probiotics, sepsis


Sepsis is a dysregulated host response to infection resulting in life-threatening organ dysfunction.1 Sepsis results in a significant burden to the healthcare system with high mortality and a cost of more than $60 billion annually in the United States alone.2 Unfortunately, antibiotic agents represent the only targeted therapy for sepsis that is otherwise managed by supportive therapy.3 The gut has long been hypothesized to be the “motor” of critical illness.4,5 The gut consists of three compartments: the microbiome, the epithelium, and the immune system. Each of these components is separately altered in sepsis, but crosstalk occurs between the three components.6–8 Accordingly, we review sepsis-induced changes of the gut microbiome as well as its interactions with intestinal epithelium and immune system. Additionally, we discuss data on targeting the microbiome as a therapeutic strategy in sepsis.

Gut Microbiome

The intestinal microbiome has the same number of cells as the human body (40 trillion) and at least two orders of magnitude more functional genes.9,10 It is shaped from the earliest time points in life and changes repeatedly based on environmental interactions.11 As the microbiome develops, influences ranging from diet to siblings to disease shape the predominant bacteria12–14 creating a highly diverse and unique microbial community.11 The predominant bacteria in the gut microbiome are within the phyla of Bacteriodetes and Firmicutes and are commonly referred to as commensal organisms reflecting their ability to support a wide variety of normal functions to maintain homeostasis.15–18

The developing and mature microbiome also plays a role in shaping local immune responses. Sampling of the intestinal lumen is constant, allowing for normal development of the immune system, as well as providing an opportunity for the mature immune system to detect new antigens. This is highlighted in germ-free animal models. These animals do not acquire an endogenous microbiome and as a result, have less adaptive immune development and function.19–21

Bacterial products also play a role in health and disease. Short-chain fatty acids (SCFA) are the primary end-products of fermentation of non-digestible carbohydrates available to the gut microbiota.17,22 Short-chain fatty acids such as acetate, propionate, and butyrate have been shown to play a role in restoring intestinal epithelial gut barrier dysfunction,23,24 as well as signaling between bacteria and the immune system.25

Sepsis results in a reduction of diversity and collapse of the normal intestinal microbiome. These changes have been referred to as a conversion from a microbiome to a pathobiome and occur almost immediately after a septic insult26 (Fig. 1). The pathobiome can demonstrate a striking lack of diversity with some patients having an ultra-low diversity group of pathogens that number as few as four taxa. These changes are associated with higher morbidity and mortality in intensive care unit (ICU) patients.27,28 Non-survivors of critical illness have been shown to have a higher abundance of Staphylococcus haemolyticus, Clostridiales sp., Campylobacter ureolyticus, Akkermansia sp., Malassezia sympodialis, Malassezia dermatis, and Saccharomyces cerevisiae, whereas survivors have a greater abundance of Collinsella aerofaciens, Blautia sp., Streptococcus sp., Faecalibacterium prausnitzii, and Bifidobacterium sp.29 The causes of the transition from a microbiome to a pathobiome are multifactorial. Critical illness in and of itself alters gut microbial composition. In addition, unintended effects of several treatments aimed at improving outcomes in critically ill patients—including antibiotics, opiates, proton pump inhibitors, and nutrition support route—also play a factor in this transition.30–32

FIG. 1.

FIG. 1.

Microbiome to pathobiome transformation. This figure demonstrates how stress events trigger a transformation from a healthy microbiome to a pathobiome.

The microbiome composition prior to the onset of sepsis may also impact outcomes after the onset of critical illness. In a recent pre-clinical study, genetically identical animals with different baseline microbiome diversity based on vendor of origin had divergent mortality after polymicrobial intra-abdominal sepsis. Animals with greater baseline diversity had improved survival and an altered immune response. This survival advantage was mechanistic because of the microbiome as co-housing the animals, resulting in animals having a similar microbiome regardless of their vendor of origin, led to abolition of mortality and immune differences.33 Co-housing represents a unique approach to adoptive immunity that may be readily explored using animal models and begins to provide a mechanistic understanding for microbiome transfer therapies such as fecal microbiota transplantation.

As bacterial composition changes and diversity decreases in the setting of sepsis, SCFA production also changes. Stool samples in critically ill ICU patients demonstrate decreased SCFA levels with lower butyric and isobutyric acid production; these findings are associated with higher rates of infection.34 Although convincing clinical data do not exist for SCFA administration in patients, pre-clinical models show promise with rescue of sepsis-induced mortality from pneumonia with oral SCFA administration after intestinal microbiome depletion after oral antibiotic administration.35

Intestinal Epithelium

The intestinal epithelium is a single-cell–layer inner lining that separates the microbiome residing in the gut lumen from the sterile host.9 The epithelial barrier is semipermeable, controlled by intercellular tight junction proteins that allow passage of water, nutrients, and antigens to sustain the host and enable appropriate immune system function.36–38 During sepsis, intestinal hyperpermeability and enterocyte apoptosis occur and result in barrier dysfunction that increases the antigens and bacterial translocation.39,40 In pre-clinical models, both reducing apoptosis and barrier permeability improve survival after sepsis.41,42 Furthermore, decreasing enterocyte apoptosis improves intestinal hyperpermeability, and suggests a link between enterocyte survival and gut barrier function competence.43

Evidence also links the microbiome to the intestinal epithelium. Mice lacking CYP1A1, a cytochrome produced by intestinal epithelial cells, have an altered gut microbiome, but improved survival after intra-peritoneal methicillin-resistant Staphylococcus aureus (MRSA) injection.44 This altered microbiome results in decreased cadaverine production, a compound that worsens intestinal permeability by activating the histamine H4 receptor/nfκB/MLCK signaling pathway. Mechanistically, the altered microbiome identified in septic knockout mice demonstrates low abundance of Enterococcus faecalis, which is a primary species responsible for cadaverine conversion from lysine. Importantly, re-introduction of that bacteria worsened intestinal permeability and abrogated the survival benefit seen in knockout mice. Notably, increased stool and blood cadaverine levels were also detected in critically ill patients infected with MRSA compared with healthy controls.

Short-chain fatty acids have also been shown to act as energy substances to protect intestinal barrier integrity.45 Acetate, propionate, and butyrate alone or in combination significantly increase transepithelial resistance of CACO-2 cells in vitro, and stimulate tight junction formation. Further, SCFAs decrease lipopolysaccharide (LPS)-induced hyperpermeability and prevent morphological disruption of the tight junction proteins ZO-1 and occludin while simultaneously inhibiting LPS-induced activation of NLRP3 inflammasome and autophagy. Clearly, SCFAs appear beneficial with regard to intestinal barrier function.

Mucosal Immune System

The mucosal immune system provides protection to the host in multiple ways.46 This protection is conferred, in part, through constant surveillance of the intestinal lumen and effector action to maintain luminal homeostasis or act if there is barrier breach.47–49 Furthermore, there is a symbiotic relation between the gut microbiome with the immune system across the intestinal epithelium whereby gut microbes influence and shape the immune system milieu and vice versa.50,51

The first line of immunologic defense is the innate immune system. Resident microbes interact with the innate immune system in an antigen non-specific manner through pathogen recognition receptors.51 Other innate system cells are then activated, releasing cytokines, and producing antimicrobial molecules. For the adaptive immune system, antigen presenting cells, such as dendritic cells, sample contents from the intestinal lumen and present them to lymphocytes in the mesenteric lymph nodes to stimulate differentiated T cells and B cells.52

Sepsis alters the immune system, and in turn, its ability to respond to gut dysbiosis. Neutrophils are a key effector of the innate immune system and act as a first line of defense against infection through an arsenal of phagocytosis, release of chemoattractants, and creation of neutrophil extracellular traps. When neutrophils have early access to the site of infection, sepsis mortality is reduced, at least in pre-clinical studies.53 Another population of innate immune cells that have been recently described are innate lymphoid cells (ILC). These cells are predominantly found at mucosal sites and have the ability to respond to bacterial exposure. Innate lymphoid cells are technically lymphocytes because they have a common lymphoid progenitor within the bone marrow.54 Furthermore, ILCs have been shown to be induced by the microbiome,55 and depletion of the microbiome in pre-clinical studies results in a loss of intestinal ILCs with subsequent disappearance of neutrophils and an increased susceptibility to neonatal sepsis.56,57 Additionally, when ILCs are lost, systemic dissemination of commensal bacteria results.58

Sepsis also induces adaptive immune dysfunction with increased sepsis-induced T cell apoptosis playing a crucial role in mediating mortality.58,59 The microbiome's relation with the adaptive immune system may also play a role in regulating intestinal epithelial cell apoptosis and gut barrier dysfunction. Septic RAG-/- animals that lack mature T lymphocytes have worsened enterocyte apoptosis, but the absence of a gut microbiome reverses the apoptosis back to the level noted in wild-type controls.60 B cells are the other component of the adaptive immune system and have a relation with the microbiome through immunoglobulin A (IgA) induction. Through sampling of the luminal contents, the microbiota stimulate IgA production that, in turn, feeds back and prevents pathogenic bacteria from overgrowing while supporting commensal microbiota proliferation.61,62 In the setting of critical illness, IgA also plays a protective role. Mice exposed to bacteria in the phylum Proteobacteria produce more IgA specific to these pathogenic bacteria and are protected against intra-abdominal sepsis.63

Recent pre-clinical mechanistic experiments demonstrate crosstalk connecting the gut barrier with the immune system, and the gut microbiome.64 Junctional adhesion molecule A (JAM-A) is a tight junction protein upregulated during sepsis.39 When JAM-A is constitutively deleted from mice, intestinal permeability is increased.65 When sepsis is induced in knockout animals, intestinal permeability worsens, whereas survival paradoxically improves as a reflection of reduced systemic inflammation and bacteremia. Additionally, the microbiome of JAM-A-/- animal's reveals increases in the phylum Proteobacteria. To determine potential mechanisms underlying the improved survival in knockout mice, a survey of the immune system disclosed increased T cells with decreased cytokine production, as well as higher circulating and gut IgA. However, JAM-A-/- x RAG-/- mice have improved survival compared with RAG-/- mice suggesting that additional mechanisms beyond the adaptive immune response are responsible for the survival benefit. Examination of the innate immune system demonstrates that neutrophil phagocytosis is increased in JAM-A-/- animals. Importantly the overall survival benefit is lost after neutrophil depletion. Thus, increased neutrophil phagocytosis of opsonized pathogenic microbiome bacteria in the setting of baseline intestinal hyperpermeability represents an integrated mechanism through which multiple components of the gut potentially drive the host response to sepsis.

Targeting the Microbiome in the ICU

In light of evidence demonstrating an association between the pathobiome and poor outcomes in critical illness, multiple approaches have taken aim at targeting the intestinal microbiome as a potential therapeutic intervention. Current methods include probiotics, prebiotics, fecal microbial transplantation (FMT), and selective decontamination of the digestive tract (SDD). Although evidence supporting each of these approaches varies, data are not compelling enough to recommend any of them as standard of care for microbiome manipulation to improve outcomes from critical illness.

Probiotics are enteral supplements containing live micro-organisms with the goal of diversifying the gut microbiome to improve health or outcome from disease. Probiotics have been explored extensively as part of the care approach for critically ill patients. The largest study in adult patients was a randomized placebo-controlled trial across 44 ICUs with more than 2,500 patients examining the effects of Lactobacillus rhamnosus GG on patients expected to require mechanical ventilation for greater than 72 hours.66 No difference was noted in the primary outcome of ventilator-associated pneumonia incidence. Additionally, none of 20 pre-specified secondary outcomes demonstrated any differences after probiotic supplementation. Notably, 15 patients had the adverse effect of the administered probiotic being recovered from a sterile site or as the sole bacteria identified in a non-sterile site. These data suggest that enterally supplemented bacteria may escape the gastrointestinal tract lumen during critical illness as a manifestation of increased intestinal permeability, reduced barrier function, and impeded immune competence.

These results are somewhat disparate from a number of smaller studies that showed that probiotics decrease ventilator-associated pneumonia in patients receiving invasive mechanical ventilation with variable impact on secondary outcomes. In light of the conflicting data often derived from low-quality studies, multiple meta-analyses have been recently published on the impact or probiotics.67–70 The largest of these includes 65 randomized controlled trials of 8,483 patients.67 This meta-analysis shows that probiotics may reduce ventilator-associated pneumonia, healthcare-associated pneumonia, ICU length of stay, hospital length of stay, and duration of mechanical ventilation without altering mortality. Notably, the benefits identified with probiotics were negated in post hoc sensitivity analyses when excluding studies with a high risk of bias. It is important to note that if there is a benefit of probiotics, numerous uncertainties remain including optimal strain(s) of bacteria, concentration, timing of initiation, duration of administration, and patient population who might benefit. Additionally, patient populations are also subject to substantial heterogeneity because baseline microbiome characteristics that are individually unique prior to probiotic administration may influence study outcomes.71 It should also be noted that several case reports and case series have reported evidence of bacteremia with organisms administered as probiotics.72,73 Therefore, even therapeutics that appear relatively benign may have deleterious impact on those with altered gut and immune function. Nonetheless, a closely related therapeutic, prebiotics have been well explored as a perhaps less risky microbiome support approach.

Prebiotics are indigestible materials that promote commensal bacterial growth. Prebiotics differ from probiotics in that they do not contain living organisms. Fiber is the most common prebiotic investigated. Fiber acts as a microbial food source for SCFA-producing organisms. Pre-clinical sepsis models have shown promise as they demonstrate decreased mortality in mice treated with a high-fiber diet for two weeks prior to sepsis onset compared with mice receiving low to normal fiber intake.74 Although the clinical relevance of a pre-treatment model is limited, this is potentially mechanistically important as the survival benefit induced by fiber was both microbiome-mediated and negated by antibiotic administration. A meta-analysis of 19 studies of fiber use in critically ill patients showed that enteral formulas with fiber may help reduce diarrhea incidence and severity as well as overall gastrointestinal complications without impacting mortality, ICU length of stay, or hospital length of stay.75 The certainty of these conclusions is low due to lower quality of many of the included studies. Additionally, a pilot study including 20 critically ill patients on broad-spectrum antibiotic agents given enteral fiber compared with no fiber demonstrated a trend toward increased relative abundance of SCFA-producing bacteria and increased SCFA levels in fiber receiving patients.76 Butyrate also rescues LPS-induced decreases in mitochondrial respiration in vitro, suggesting a potential additional mechanism through which SCFAs may help to mitigate the immunoinflammatory response in sepsis.77 Prebiotics may be most effective when a relatively normal microbiome remains resident and functional within a patient's gastrointestinal tract. For those with a substantially deranged microbiome, one well documented consequence is acute illness from Clostridium difficile overgrowth.

Fecal microbiota transplantation is a technique by which the entire native intestinal microbiome is replaced with a donor stool sample. This procedure is theoretically beneficial when there has been substantial alteration of the normal host diversity with an increase in normally low concentration pathogens. The ultimate goal is to restore normal microbial diversity by providing high volumes of commensals and preventing re-colonization of pathogenic bacteria. This dual approach theoretically allows for restoration of normal adaptive host metabolism and immune function.

Fecal microbiota transplantation has been studied most extensively in patients with recurrent as opposed to initial Clostridium difficile infection. Whereas oral antimicrobials remain the first-line treatment of Clostridium difficile, treatment failure occurs in a subset of patients.78 Based on robust efficacy data, FMT has become part of medical professional organization recommendations for recurrent disease management.79,80 With an increased understanding of sepsis-induced dysbiosis, there has been increased interest in the use of FMT in the ICU. Mechanistically, recent pre-clinical work suggests that FMT restores SCFA-producing microbial communities that, in turn, stimulate immune function to clear overgrown pathogens. Mortality is reduced after FMT in mice in an interferon regulatory factor 3-dependent manner with an associated increase in SCFA-producing Bacteriodetes.81 Additionally, FMT after murine sepsis improves gut barrier function by reducing epithelial cell apoptosis, improving the composition of the mucus layer, reducing intestinal permeability with upregulation of tight junction proteins, and improving the host inflammatory response.82

Despite the conceptual appeal of FMT and pre-clinical data supporting its use, numerous barriers currently exist to use FMT routinely in critically ill patients. A major concern is that most critically ill patients receive antibiotic agents during the ICU stay, a therapy that would be expected to maladaptively alter the transplanted flora after FMT. Furthermore, the implications of transplanting live microbes into a host often encumbered by sepsis-induced immunosuppression is unknown. Additionally, the lack of standardization of dose, frequency, and bacterial composition make therapy generalization quite difficult. Finally, there have been case reports of multi-drug–resistant organism bacteremia in patients receiving FMT including one attributable death.83 Although such a dire complication is extremely rare, it reinforces the need to focus on safety if this approach is to be used for critically ill patients who demonstrate compromised immune competence that is related, at least in part, to the gastrointestinal tract microbiome and gut barrier function. Accordingly, despite promising case reports documenting benefit, FMT as a routine therapy for critically ill patients remains experimental.84 Instead, a durably investigated approach leverages a common therapeutic, namely oral antibiotic therapy to alter gastrointestinal tract bacterial bioburden.

Loosely related to colonic preparation in advance of resectional surgery with a planned anastomosis, SDD eliminates bacteria from the intestinal tract using oral antibiotic therapy. This strategy relies on removing pathogenic bacteria that would sense a host stressor and become more virulent. Unfortunately, the process is non-specific and also removes commensal organisms. The most common application for this technique is for patients undergoing elective bowel surgery because pre-operative bowel preparation that includes antibiotic agents has been shown to reduce anastomotic leaks and surgical site infections.85 In critically ill patients, there is robust evidence regarding SDD. A meta-analysis of 41 trials (most with poor reporting quality) and more than 11,000 patients receiving invasive mechanical ventilation for at least 48 hours showed that SDD (using both oral and system antibiotic agents) decreased mortality and respiratory infections.86

Furthermore, oral antibiotic agents alone may decrease mortality and respiratory infection. It is important to note, however, that these studies were performed mostly in a single country characterized by a low baseline antibiotic resistance profile and may not be directly applicable to developed nations with substantial resistance profiles. In contrast, a randomized controlled trial of more than 8,600 patients in ICUs with moderate to high antimicrobial resistance failed to find any SDD benefit in decreasing infections or mortality.87 Given the conflicting data on SDD and the concern for amplifying antibiotic resistance as a result of using SDD, this therapy has not become routinely used throughout most of the world. Nonetheless, it should be noted that a recent study of more than 6,000 patients receiving either oral antibiotic agents or no therapy over a five-year period revealed no differences in the incidence of multi-drug–resistant bacteria, except for a lower rate of ESBL-producing Klebsiella pneumoniae and a higher rate of vancomycin-resistant Enterococcus faecium in the treatment group.88 Therefore, SDD remains a tantalizing but untargeted therapeutic that may benefit from specific refinement in a precision fashion that targets uniquely identified pathogenic bacteria present in a specific patient. The ability to provide such precise targeting remains elusive at present.

Conclusions

Our understanding of the interface of the gut microbiome with the immune system during health and disease continues to expand. Communication between the intestinal microbiome, the intestinal epithelium, and the immune system prior to and after sepsis impact intestinal barrier function and lead to local and systemic changes that influence outcome. Although targeting the pathobiome has conceptual appeal, more research needs to be done to determine the best approach (if any) toward manipulating the microbiome in critically ill patients for therapeutic gain.

Funding Information

This work was supported by funding from the National Institutes of Health (GM072808, GM104323, AA027396)

Author Disclosure Statement

The authors have no conflicts of interest to disclose.

References

  • 1. Singer M, Deutschman CS, Seymour C, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA 2016;315(8):801–810; doi: 10.1001/jama.2016.0287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Rhee C, Dantes R, Epstein L, et al. Incidence and trends of sepsis in US hospitals using clinical vs claims data, 2009–2014. JAMA 2017;318(13):1241–1249; doi: 10.1001/jama.2017.13836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Evans L, Rhodes A, Alhazzani W, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Crit Care Med 2021;49(11):e1063–e1143; doi: 10.1097/CCM.0000000000005337 [DOI] [PubMed] [Google Scholar]
  • 4. Carrico CJ, Meakins JL, Marshall JC, et al. Multiple-organ-failure syndrome. Arch Surg 1986;121(2):196–208; doi: 10.1001/archsurg.1986.01400020082010 [DOI] [PubMed] [Google Scholar]
  • 5. Klingensmith NJ, Coopersmith CM. The gut as the motor of multiple organ dysfunction in critical illness. Crit Care Clin 2016;32(2):203–212; doi: 10.1016/j.ccc.2015.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Clark JA, Coopersmith CM. Intestinal crosstalk: A new paradigm for understanding the gut as the “motor” of critical illness. Shock 2007;28(4):384–393; doi: 10.1097/shk.0b013e31805569df [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Mittal R, Coopersmith CM. Redefining the gut as the motor of critical illness. Trends Mol Med 2014;20(4):214–223; doi: 10.1016/j.molmed.2013.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Miniet A, Grunwell J, Coopersmith C. The microbiome and the immune system in critical illness. Curr Opin Crit Care 2021;27(2):157–163; doi: 10.1097/MCC.0000000000000800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016;164(3):337–340; doi: 10.1016/j.cell.2016.01.013 [DOI] [PubMed] [Google Scholar]
  • 10. International Human Genome Sequencing Consortium. Finishing the euchromatic sequence of the human genome. Nature 2004;431(7011):931–945; doi: 10.1038/nature03001 [DOI] [PubMed] [Google Scholar]
  • 11. Dominguez-Bello MG, Godoy-Vitorino F, Knight R, et al. Role of the microbiome in human development. Gut 2019;68(6):1108–1114; doi: 10.1136/gutjnl-2018-317503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Martin R, Makino H, Cetinyurek Yavuz A, et al. Early-life events, including mode of delivery and type of feeding, Siblings and gender, shape the developing gut microbiota. PLoS One 2016;11(6):e0158498; doi: 10.1371/journal.pone.0158498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Scott KP, Gratz SW, Sheridan PO, et al. The influence of diet on the gut microbiota. Pharmacol Res 2013;69(1):52–60; doi: 10.1016/j.phrs.2012.10.020 [DOI] [PubMed] [Google Scholar]
  • 14. Palleja A, Mikkelsen KH, Forslund SK, et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat Microbiol 2018;3(11):1255–1265; doi: 10.1038/s41564-018-0257-9 [DOI] [PubMed] [Google Scholar]
  • 15. Dethlefsen L, Eckburg PB, Bik EM, et al. Assembly of the human intestinal microbiota. Trends Ecol Evol 2006;21(9):517–523; doi: 10.1016/j.tree.2006.06.013 [DOI] [PubMed] [Google Scholar]
  • 16. Qin J, Li R, Raes J, et al. A human gut microbial gene catalog established by metagenomic sequencing. Nature 2010;464(7285):59–65; doi: 10.1038/nature08821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016;7(3):189–200; doi: 10.1080/19490976.2015.1134082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Zegarra-Ruiz DF, Kim DV, Norwood K, et al. Thymic development of gut-microbiota-specific T cells. Nature 2021;1–5; doi: 10.1038/s41586-021-03531-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Smith K, McCoy KD, Macpherson AJ. Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin Immunol 2007;19(2):59–69; doi: 10.1016/j.smim.2006.10.002 [DOI] [PubMed] [Google Scholar]
  • 20. Round JL, Mazmanian SK. The gut microbiome shapes intestinal immune responses during health and disease. Nat Rev Immunol 2009;9(5):313–323; doi: 10.1038/nri2515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Cabrera-Perez J, Badovinac VP, Griffith TS. Enteric immunity, the gut microbiome, and sepsis: Rethinking the germ theory of disease. Exp Biol Med 2017;242(2):127–139; doi: 10.1177/1535370216669610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Pickard JM, Zeng MY, Caruso R, et al. Gut microbiota: Role in pathogen colonization, immune responses and inflammatory disease. Immunol Rev 2017;279(1):70–89; doi: 10.1111/imr.12567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Yan H, Ajuwon KM. Butyrate modifies intestinal barrier function in IPEC-J2 cells through a selective upregulation of tight junction proteins and activation of the Akt signaling pathway. PLoS One 2017;12(6):e0179586; doi: 10.1371/journal.pone.0179586 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Miao W, Wu X, Wang K, et al. sodium butyrate promotes reassembly of tight junctions in Caco-2 monolayers involving inhibition of MLCK/MLC2 pathway and phosphorylation of PKCβ2. Int J Mol Sci 2016;17(10):1696; doi: 10.3390/ijms17101696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Corrêa-Oliveira R, Fachi JL, Vieira A, et al. Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunol 2016;5(4):e73; doi: 10.1038/cti.2016.17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Krezalek MA, DeFazio J, Zaborina O, et al. The shift of an intestinal “microbiome” to a “pathobiome” governs the course and outcome of sepsis following surgical injury: Shock 2016;45(5):475–482; doi: 10.1097/SHK.0000000000000534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Babrowski T, Holbrook C, Moss J, et al. Pseudomonas aeruginosa virulence expression is directly activated by morphine and is capable of causing lethal gut derived sepsis in mice during chronic morphine administration. Ann Surg 2012;255(2):386–393; doi: 10.1097/SLA.0b013e3182331870 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Freedberg DE, Zhou MJ, Cohen ME, 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):1203–1211; doi: 10.1007/s00134-018-5268-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Prevel R, Enaud R, Orieux A, et al. Gut bacteriobiota and mycobiota are both associated with Day-28 mortality among critically ill patients. Crit Care 2022;26(1):105; doi: 10.1186/s13054-022-03980-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Miller WD, Keskey R, Alverdy JC. Sepsis and the microbiome: A vicious cycle. J Infect Dis 2020;223(Suppl 3):S264–S269; doi: 10.1093/infdis/jiaa682 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Suez J, Zmora N, Zilberman-Schapira G, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 2018;174(6):1406-1423.e16; doi: 10.1016/j.cell.2018.08.047 [DOI] [PubMed] [Google Scholar]
  • 32. David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505(7484):559–563; doi: 10.1038/nature12820 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Fay KT, Klingensmith NJ, Chen C-W, et al. The gut microbiome alters immunophenotype and survival from sepsis. FASEB J 2019;33(10):11258–11269; doi: 10.1096/fj.201802188R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Valdés-Duque BE, Giraldo-Giraldo NA, Jaillier-Ramírez AM, et al. Stool short-chain fatty acids in critically ill patients with sepsis. J Am Coll Nutr 2020;39(8):706–712; doi: 10.1080/07315724.2020.1727379 [DOI] [PubMed] [Google Scholar]
  • 35. Wu T, Li H, Su C, et al. Microbiota-derived short-chain fatty acids promote LAMTOR2-mediated immune responses in macrophages. mSystems 2020;5(6); doi: 10.1128/mSystems.00587-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 2009;9(11):799–809; doi: 10.1038/nri2653 [DOI] [PubMed] [Google Scholar]
  • 37. Garcia-Hernandez V, Quiros M, Nusrat A. Intestinal epithelial claudins: Expression and regulation in homeostasis and inflammation. Ann NY Acad Sci 2017;1397(1):66–79; doi: 10.1111/nyas.13360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Tsukita S, Tanaka H, Tamura A. The claudins: From tight junctions to biological systems. Trends Biochem Sci 2019;44(2):141–152; doi: 10.1016/j.tibs.2018.09.008 [DOI] [PubMed] [Google Scholar]
  • 39. Yoseph BP, Klingensmith NJ, Liang Z, et al. Mechanisms of intestinal barrier dysfunction in sepsis. Shock 2016;46(1):52–59; doi: 10.1097/SHK.0000000000000565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Coopersmith CM, Stromberg PE, Dunne W, et al. Inhibition of intestinal epithelial apoptosis and survival in a murine model of pneumonia-induced sepsis. JAMA 2002;287(13):1716–1721; doi: 10.1001/jama.287.13.1716 [DOI] [PubMed] [Google Scholar]
  • 41. Lorentz CA, Liang Z, Meng M, et al. Myosin light chain kinase knockout improves gut barrier function and confers a survival advantage in polymicrobial sepsis. Mol Med 2017;23:155–165; doi: 10.2119/molmed.2016.00256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Klingensmith NJ, Yoseph BP, Liang Z, et al. Epidermal growth factor improves intestinal integrity and survival in murine sepsis following chronic alcohol ingestion. Shock 2017;47(2):184–192; doi: 10.1097/SHK.0000000000000709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Otani S, Oami T, Yoseph BP, et al. Overexpression of BCL-2 in the intestinal epithelium prevents sepsis-induced gut barrier function via altering tight junction protein expression. Shock 2020;54(3):330–336; doi: 10.1097/SHK.0000000000001463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Ma X, Jin H, Chu X, et al. The Host CYP1A1-microbiota metabolic axis promotes gut barrier disruption in methicillin-resistant Staphylococcus aureus-induced abdominal sepsis. Front Microbiol 2022;13:802409; doi: 10.3389/fmicb.2022.802409 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Feng Y, Wang Y, Wang P, et al. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell Physiol Biochem 2018;49(1):190–205; doi: 10.1159/000492853 [DOI] [PubMed] [Google Scholar]
  • 46. Otani S, Coopersmith CM. Gut integrity in critical illness. J Intensive Care 2019;7:17; doi: 10.1186/s40560-019-0372-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Pabst O, Cerovic V, Hornef M. Secretory IgA in the coordination of establishment and maintenance of the microbiota. Trends Immunol 2016;37(5):287–296; doi: 10.1016/j.it.2016.03.002 [DOI] [PubMed] [Google Scholar]
  • 48. Gutzeit C, Magri G, Cerutti A. Intestinal IgA production and its role in host-microbe interaction. Immunol Rev 2014;260(1):76–85; doi: 10.1111/imr.12189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Jangid A, Fukuda S, Seki M, et al. Gut microbiota alternation under the intestinal epithelium-specific knockout of mouse Piga gene. Sci Rep 2022;12(1):10812; doi: 10.1038/s41598-022-15150-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Thaiss CA, Levy M, Suez J, et al. The interplay between the innate immune system and the microbiota. Curr Opin Immunol 2014;26:41–48; doi: 10.1016/j.coi.2013.10.016 [DOI] [PubMed] [Google Scholar]
  • 51. Thaiss CA, Zmora N, Levy M, et al. The microbiome and innate immunity. Nature 2016;535(7610):65–74; doi: 10.1038/nature18847 [DOI] [PubMed] [Google Scholar]
  • 52. Zhao Q, Elson CO. Adaptive immune education by gut microbiota antigens. Immunology 2018;154(1):28–37; doi: 10.1111/imm.12896 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Craciun FL, Schuller ER, Remick DG. Early enhanced local neutrophil recruitment in peritonitis-induced sepsis improves bacterial clearance and survival. J Immunol 2010;185(11):6930–6938; doi: 10.4049/jimmunol.1002300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Ganal-Vonarburg SC, Duerr CU. The interaction of intestinal microbiota and innate lymphoid cells in health and disease throughout life. Immunology 2020;159(1):39–51; doi: 10.1111/imm.13138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Satoh-Takayama N, Vosshenrich CAJ, Lesjean-Pottier S, et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 2008;29(6):958–970; doi: 10.1016/j.immuni.2008.11.001 [DOI] [PubMed] [Google Scholar]
  • 56. Deshmukh HS, Liu Y, Menkiti OR, et al. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat Med 2014;20(5):524–530; doi: 10.1038/nm.3542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Niu X, Daniel S, Kumar D, et al. Transient neonatal antibiotic exposure increases susceptibility to late-onset sepsis driven by microbiota-dependent suppression of type 3 innate lymphoid cells. Sci Rep 2020;10:12974; doi: 10.1038/s41598-020-69797-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Hotchkiss RS, Swanson PE, Freeman BD, et al. Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Crit Care Med 1999;27(7):1230–1251. [DOI] [PubMed] [Google Scholar]
  • 59. Coopersmith CM, Chang KC, Swanson PE, et al. Overexpression of Bcl-2 in the intestinal epithelium improves survival in septic mice. Crit Care Med 2002;30(1):195–201; doi: 10.1097/00003246-200201000-00028 [DOI] [PubMed] [Google Scholar]
  • 60. Fox AC, McConnell KW, Yoseph BP, et al. The endogenous bacteria alter gut epithelial apoptosis and decrease mortality following Pseudomonas aeruginosa pneumonia. Shock 2012;38(5):508–514; doi: 10.1097/SHK.0b013e31826e47e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Bunker JJ, Bendelac A. IgA responses to microbiota. Immunity 2018;49(2):211–224; doi: 10.1016/j.immuni.2018.08.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Donaldson GP, Ladinsky MS, Yu KB, et al. Gut microbiota utilize immunoglobulin A for mucosal colonization. Science 2018;360(6390):795–800; doi: 10.1126/science.aaq0926 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Wilmore JR, Gaudette BT, Atria DG, et al. Commensal microbes induce serum IgA responses that protect against polymicrobial sepsis. Cell Host Microbe 2018;23(3):302-311.e3; doi: 10.1016/j.chom.2018.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Klingensmith NJ, Fay KT, Swift DA, et al. Junctional adhesion molecule-A deletion increases phagocytosis and improves survival in a murine model of sepsis. JCI Insight 2022;7(16); doi: 10.1172/jci.insight.156255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Laukoetter MG, Nava P, Lee WY, et al. JAM-A regulates permeability and inflammation in the intestine in vivo. J Exp Med 2007;204(13):3067–3076; doi: 10.1084/jem.20071416 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Johnstone J, Meade M, Lauzier F, et al. Effect of probiotics on incident ventilator-associated pneumonia in critically ill patients: A randomized clinical trial. JAMA 2021;326(11):1024–1033; doi: 10.1001/jama.2021.13355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Sharif S, Greer A, Skorupski C, et al. Probiotics in critical illness: A systematic review and meta-analysis of randomized controlled trials. Crit Care Med 2022;50(8):1175–1186; doi: 10.1097/CCM.0000000000005580 [DOI] [PubMed] [Google Scholar]
  • 68. Song H, Hu W, Zhou X, et al. Clinical benefits from administering probiotics to mechanical ventilated patients in intensive care unit: A PRISMA-guided meta-analysis. Front Nutr 2021;8:798827; doi: 10.3389/fnut.2021.798827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Zhao J, Li L-Q, Chen C-Y, et al. Do probiotics help prevent ventilator-associated pneumonia in critically ill patients? A systematic review with meta-analysis. ERJ Open Res 2021;7(1):00302–02020; doi: 10.1183/23120541.00302-2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Su M, Jia Y, Li Y, et al. Probiotics for the prevention of ventilator-associated pneumonia: A meta-analysis of randomized controlled trials. Respir Care 2020;65(5):673–685; doi: 10.4187/respcare.07097 [DOI] [PubMed] [Google Scholar]
  • 71. Zmora N, Zilberman-Schapira G, Suez J, et al. personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell 2018;174(6):1388-1405.e21; doi: 10.1016/j.cell.2018.08.041 [DOI] [PubMed] [Google Scholar]
  • 72. Yelin I, Flett KB, Merakou C, et al. Genomic and epidemiological evidence of bacterial transmission from probiotic capsule to blood in ICU patients. Nat Med 2019;25(11):1728–1732; doi: 10.1038/s41591-019-0626-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Sakurai Y, Watanabe T, Miura Y, et al. Clinical and bacteriologic characteristics of six cases of Bifidobacterium breve bacteremia due to probiotic administration in the neonatal intensive care unit. Pediatr Infect Dis J 2022;41(1):62–65; doi: 10.1097/INF.0000000000003232 [DOI] [PubMed] [Google Scholar]
  • 74. Morowitz M, Di Caro V, Pang D, et al. Dietary supplementation with non-fermentable fiber alters the gut microbiota and confers protection in a murine model of sepsis. Crit Care Med 2017;45(5):e516–e523; doi: 10.1097/CCM.0000000000002291 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Cara KC, Beauchesne AR, Wallace TC, et al. Safety of using enteral nutrition formulations containing dietary fiber in hospitalized critical care patients: A systematic review and meta-analysis. J Parenter Enter Nutr 2021;45(5):882–906; doi: 10.1002/jpen.2210 [DOI] [PubMed] [Google Scholar]
  • 76. Freedberg DE, Messina M, Lynch E, et al. Impact of fiber-based enteral nutrition on the gut microbiome of icu patients receiving broad-spectrum antibiotics: A randomized pilot trial. Crit Care Explor 2020;2(6); doi: 10.1097/CCE.0000000000000135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Weiss SL, Zhang D, Farooqi S, et al. Sodium butyrate reverses lipopolysaccharide-induced mitochondrial dysfunction in lymphoblasts. J Cell Mol Med 2022;26(11):3290–3293; doi: 10.1111/jcmm.17342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Cornely OA, Crook DW, Esposito R, et al. Fidaxomicin versus vancomycin for infection with Clostridium difficile in Europe, Canada, and the USA: A double-blind, non-inferiority, randomised controlled trial. Lancet Infect Dis 2012;12(4):281–289; doi: 10.1016/S1473-3099(11)70374-7 [DOI] [PubMed] [Google Scholar]
  • 79. Debast SB, Bauer MP, Kuijper EJ. European Society of Clinical Microbiology and Infectious Diseases: Update of the treatment guidance document for Clostridium difficile infection. Clin Microbiol Infect 2014;20:1–26; doi: 10.1111/1469-0691.12418 [DOI] [PubMed] [Google Scholar]
  • 80. McDonald LC, Gerding DN, Johnson S, et al. Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis 2018;66(7):e1–e48; doi: 10.1093/cid/cix1085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Kim SM, DeFazio JR, Hyoju SK, et al. Fecal microbiota transplant rescues mice from human pathogen mediated sepsis by restoring systemic immunity. Nat Commun 2020;11(1):2354; doi: 10.1038/s41467-020-15545-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Gai X, Wang H, Li Y, et al. Fecal microbiota transplantation protects the intestinal mucosal barrier by reconstructing the gut microbiota in a murine model of sepsis. Front Cell Infect Microbiol 2021;11:736204; doi: 10.3389/fcimb.2021.736204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. DeFilipp Z, Bloom PP, Soto MT, et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med 2019; doi: 10.1056/NEJMoa1910437 [DOI] [PubMed] [Google Scholar]
  • 84. McClave SA, Patel J, Bhutiani N. Should fecal microbial transplantation be used in the ICU? Curr Opin Crit Care 2018;24(2):105–111; doi: 10.1097/MCC.0000000000000489 [DOI] [PubMed] [Google Scholar]
  • 85. Klinger AL, Green H, Monlezun DJ, et al. The role of bowel preparation in colorectal surgery: Results of the 2012-2015 ACS-NSQIP Data. Ann Surg 2019;269(4):671–677; doi: 10.1097/SLA.0000000000002568 [DOI] [PubMed] [Google Scholar]
  • 86. Minozzi S, Pifferi S, Brazzi L, et al. Topical antibiotic prophylaxis to reduce respiratory tract infections and mortality in adults receiving mechanical ventilation. Cochrane Database Syst Rev 2021;1:CD000022; doi: 10.1002/14651858.CD000022.pub4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Wittekamp BH, Plantinga NL, Cooper BS, et al. Decontamination strategies and bloodstream infections with antibiotic-resistant microorganisms in ventilated patients. JAMA 2018;320(20):2087–2098; doi: 10.1001/jama.2018.13765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Wang B, Briegel J, Krueger WA, et al. Ecological effects of selective oral decontamination on multidrug-resistance bacteria acquired in the intensive care unit: A case-control study over 5 years. Intensive Care Med 2022;48(9):1165–1175; doi: 10.1007/s00134-022-06826-7 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Surgical Infections are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES