Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Clin Infect Dis ; 72(11): e868-e871, 2021 06 01.
Article in English | MEDLINE | ID: mdl-32940644

ABSTRACT

In a cohort of infants, we found that lack of the Lewis histo-blood group antigen was associated with increased susceptibility to shigellosis. Broadly inhibiting fucosylation in epithelial cells in vitro decreased invasion by Shigella flexneri. These results support a role for fucosylated glycans in susceptibility to shigellosis.


Subject(s)
Dysentery, Bacillary , Humans , Infant , Lewis Blood Group Antigens
2.
Anaerobe ; 37: 25-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26549696

ABSTRACT

Recent advances in developmental and stem cell biology have resulted in techniques that enable the generation and maintenance of complex epithelium in vitro. While these models have been utilized to study host development and disease, a renewed appreciation of host-microbe interactions has sparked interest in employing these new techniques to study microbes at the epithelial interface. Here we review the current advances in host-microbe interactions that have resulted from experiments using these complex epithelia. Furthermore we highlight aspects of these techniques that warrant further development to facilitate the study of host-microbe interactions.


Subject(s)
Intestinal Mucosa/microbiology , Stem Cells , Animals , Bacteriological Techniques , Clostridioides difficile/physiology , Helicobacter pylori/physiology , Humans , In Vitro Techniques , Salmonella enterica/physiology , Stem Cells/microbiology
3.
Infect Immun ; 83(1): 138-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25312952

ABSTRACT

Clostridium difficile is the leading cause of infectious nosocomial diarrhea. The pathogenesis of C. difficile infection (CDI) results from the interactions between the pathogen, intestinal epithelium, host immune system, and gastrointestinal microbiota. Previous studies of the host-pathogen interaction in CDI have utilized either simple cell monolayers or in vivo models. While much has been learned by utilizing these approaches, little is known about the direct interaction of the bacterium with a complex host epithelium. Here, we asked if human intestinal organoids (HIOs), which are derived from pluripotent stem cells and demonstrate small intestinal morphology and physiology, could be used to study the pathogenesis of the obligate anaerobe C. difficile. Vegetative C. difficile, microinjected into the lumen of HIOs, persisted in a viable state for up to 12 h. Upon colonization with C. difficile VPI 10463, the HIO epithelium is markedly disrupted, resulting in the loss of paracellular barrier function. Since similar effects were not observed when HIOs were colonized with the nontoxigenic C. difficile strain F200, we directly tested the role of toxin using TcdA and TcdB purified from VPI 10463. We show that the injection of TcdA replicates the disruption of the epithelial barrier function and structure observed in HIOs colonized with viable C. difficile.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Clostridioides difficile/metabolism , Enterotoxins/toxicity , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Organoids/microbiology , Organoids/physiology , Humans , Intestinal Mucosa/drug effects , Organ Culture Techniques
5.
Anaerobe ; 30: 90-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25230329

ABSTRACT

Clostridium difficile is the major cause of antibiotic-associated diarrhea and pseudomembranous colitis in healthcare settings. However, the host factors involved in the intestinal inflammatory response and pathogenesis of C. difficile infection (CDI) are largely unknown. Here we investigated the role of leukotrienes (LTs), a group of pro-inflammatory lipid mediators, in CDI. Notably, the neutrophil chemoattractant LTB4, but not cysteinyl (cys) LTs, was induced in the intestine of C57BL/6 mice infected with either C. difficile strain VPI 10463 or strain 630. Genetic or pharmacological ablation of LT production did not ameliorate C. difficile colitis or clinical signs of disease in infected mice. Histological analysis demonstrated that intestinal neutrophilic inflammation, edema and tissue damage in mice during acute and severe CDI were not modulated in the absence of LTs. In addition, CDI induced a burst of cytokines in the intestine of infected mice in a LT-independent manner. Serum levels of anti-toxin A immunoglobulin (Ig) G levels were also not modulated by endogenous LTs. Collectively, our results do not support a role for LTs in modulating host susceptibility to CDI in mice.


Subject(s)
Clostridioides difficile/growth & development , Clostridium Infections/microbiology , Clostridium Infections/pathology , Colitis/microbiology , Colitis/pathology , Leukotrienes/metabolism , Animals , Clostridioides difficile/immunology , Disease Models, Animal , Female , Histocytochemistry , Mice, Inbred C57BL
6.
mBio ; 15(3): e0333823, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38376154

ABSTRACT

Innate lymphoid cells (ILCs) play a critical role in maintaining intestinal health in homeostatic and diseased conditions. During Clostridium difficile infection (CDI), IL-33 activates ILC2 to protect from colonic damage and mortality. The function of IL-33 and ILC is tightly regulated by the intestinal microbiota. We set out to determine the impact of antibiotic-induced disruption of the microbiome on ILC function. Our goal was to understand antibiotic-induced changes in ILC function on susceptibility to C. difficile colitis in a mouse model. We utilized high-throughput single-cell RNAseq to investigate the phenotypic features of colonic ILC at baseline, after antibiotic administration with or without IL-33 treatment. We identified a heterogeneous landscape of colonic ILCs with gene signatures of inflammatory, anti-inflammatory, migratory, progenitor, plastic, and antigen-presenting ILCs. Antibiotic treatment decreased ILC2 while coordinately increasing ILC1 and ILC3 phenotypes. Notably, Ifng+, Ccl5+, and Il23r+ ILC increased after antibiotics. IL-33 treatment counteracted the antibiotic effect by downregulating ILC1 and ILC3 and activating ILC2. In addition, IL-33 treatment markedly induced the expression of type 2 genes, including Areg and Il5. Finally, we identified amphiregulin, produced by ILC2, as protective during C. difficile infection. Together, our data expand our understanding of how antibiotics induce susceptibility to C. difficile colitis through their impact on ILC subsets and function.IMPORTANCEClostridium difficile infection (CDI) accounts for around 500,000 symptomatic cases and over 20,000 deaths annually in the United States alone. A major risk factor of CDI is antibiotic-induced dysbiosis of the gut. Microbiota-regulated IL-33 and innate lymphoid cells (ILCs) are important in determining the outcomes of C. difficile infection. Understanding how antibiotic and IL-33 treatment alter the phenotype of colon ILCs is important to identify potential therapeutics. Here, we performed single-cell RNAseq of mouse colon ILCs collected at baseline, after antibiotic treatment, and after IL-33 treatment. We identified heterogeneous subpopulations of all three ILC subtypes in the mouse colon. Our analysis revealed several potential pathways of antibiotic-mediated increased susceptibility to intestinal infection. Our discovery that Areg is abundantly expressed by ILCs, and the protection of mice from CDI by amphiregulin treatment, suggests that the amphiregulin-epidermal growth factor receptor pathway is a potential therapeutic target for treating intestinal colitis.


Subject(s)
Clostridioides difficile , Clostridium Infections , Colitis , Enterocolitis, Pseudomembranous , Mice , Animals , Immunity, Innate , Lymphocytes , Anti-Bacterial Agents/pharmacology , Interleukin-33/metabolism , Interleukin-33/pharmacology , Amphiregulin/metabolism , Amphiregulin/pharmacology , Dysbiosis , Clostridium Infections/metabolism
7.
J Clin Microbiol ; 51(1): 278-80, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23052320

ABSTRACT

In a large prospective comparison, the illumigene test detected Clostridium difficile in 98% of toxin-positive and 58% of toxin-negative samples confirmed positive by other methods. The Xpert was uniformly sensitive. Most samples with discrepant results had C. difficile concentrations below the illumigene limit of detection. The significance of low-level C. difficile detection needs investigation.


Subject(s)
Bacterial Load/methods , Clostridioides difficile/isolation & purification , Feces/microbiology , Molecular Diagnostic Techniques/methods , Adult , Humans , Limit of Detection
8.
bioRxiv ; 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37609255

ABSTRACT

Fecal Microbiota Transplant (FMT) is an emerging therapy that has had remarkable success in treatment and prevention of recurrent Clostridioides difficile infection (rCDI). FMT has recently been associated with adverse outcomes such as inadvertent transfer of antimicrobial resistance, necessitating development of more targeted bacteriotherapies. To address this challenge, we developed a novel systems biology pipeline to identify candidate probiotic strains that would be predicted to interrupt C. difficile pathogenesis. Utilizing metagenomic characterization of human FMT donor samples, we identified those metabolic pathways most associated with successful FMTs and reconstructed the metabolism of encoding species to simulate interactions with C. difficile . This analysis resulted in predictions of high levels of cross-feeding for amino acids in species most associated with FMT success. Guided by these in silico models, we assembled consortia of bacteria with increased amino acid cross-feeding which were then validated in vitro . We subsequently tested the consortia in a murine model of CDI, demonstrating total protection from severe CDI through decreased toxin levels, recovered gut microbiota, and increased intestinal eosinophils. These results support the novel framework that amino acid cross-feeding is likely a critical mechanism in the initial resolution of CDI by FMT. Importantly, we conclude that our predictive platform based on predicted and testable metabolic interactions between the microbiota and C. difficile led to a rationally designed biotherapeutic framework that may be extended to other enteric infections.

9.
mBio ; 13(4): e0118322, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35856563

ABSTRACT

The severity of Clostridioides difficile infections (CDI) has increased over the last few decades. Patient age, white blood cell count, and creatinine levels as well as C. difficile ribotype and toxin genes have been associated with disease severity. However, it is unclear whether specific members of the gut microbiota are associated with variations in disease severity. The gut microbiota is known to interact with C. difficile during infection. Perturbations to the gut microbiota are necessary for C. difficile to colonize the gut. The gut microbiota can inhibit C. difficile colonization through bile acid metabolism, nutrient consumption, and bacteriocin production. Here, we sought to demonstrate that members of the gut bacterial communities can also contribute to disease severity. We derived diverse gut communities by colonizing germfree mice with different human fecal communities. The mice were then infected with a single C. difficile ribotype 027 clinical isolate, which resulted in moribundity and histopathologic differences. The variation in severity was associated with the human fecal community that the mice received. Generally, bacterial populations with pathogenic potential, such as Enterococcus, Helicobacter, and Klebsiella, were associated with more-severe outcomes. Bacterial groups associated with fiber degradation and bile acid metabolism, such as Anaerotignum, Blautia, Lactonifactor, and Monoglobus, were associated with less-severe outcomes. These data indicate that, in addition to the host and C. difficile subtype, populations of gut bacteria can influence CDI disease severity. IMPORTANCE Clostridioides difficile colonization can be asymptomatic or develop into an infection ranging in severity from mild diarrhea to toxic megacolon, sepsis, and death. Models that predict severity and guide treatment decisions are based on clinical factors and C. difficile characteristics. Although the gut microbiome plays a role in protecting against CDI, its effect on CDI disease severity is unclear and has not been incorporated into disease severity models. We demonstrated that variation in the microbiome of mice colonized with human feces yielded a range of disease outcomes. These results revealed groups of bacteria associated with both severe and mild C. difficile infection outcomes. Gut bacterial community data from patients with CDI could improve our ability to identify patients at risk of developing more severe disease and improve interventions that target C. difficile and the gut bacteria to reduce host damage.


Subject(s)
Clostridioides difficile , Clostridium Infections , Gastrointestinal Microbiome , Animals , Bacteria/genetics , Bile Acids and Salts , Clostridium Infections/microbiology , Feces/microbiology , Humans , Mice
10.
Open Forum Infect Dis ; 9(3): ofac001, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35146046

ABSTRACT

BACKGROUND: The incidence of Clostridioides difficile infection (CDI) has increased over the past 2 decades and is considered an urgent threat by the Centers for Disease Control and Prevention. Hypervirulent strains such as ribotype 027, which possess genes for the additional toxin C. difficile binary toxin (CDT), are contributing to increased morbidity and mortality. METHODS: We retrospectively tested stool from 215 CDI patients for CDT by enzyme-linked immunosorbent assay (ELISA). Stratifying patients by CDT status, we assessed if disease severity and clinical outcomes correlated with CDT positivity. Additionally, we completed quantitative PCR (PCR) DNA extracted from patient stool to detect cdtB gene. Lastly, we performed 16 S rRNA gene sequencing to examine if CDT-positive samples had an altered fecal microbiota. RESULTS: We found that patients with CdtB, the pore-forming component of CDT, detected in their stool by ELISA, were more likely to have severe disease with higher 90-day mortality. CDT-positive patients also had higher C. difficile bacterial burden and white blood cell counts. There was no significant difference in gut microbiome diversity between CDT-positive and -negative patients. CONCLUSIONS: Patients with fecal samples that were positive for CDT had increased disease severity and worse clinical outcomes. Utilization of PCR and testing for C. difficile toxins A and B may not reveal the entire picture when diagnosing CDI; detection of CDT-expressing strains is valuable in identifying patients at risk of more severe disease.

11.
Mucosal Immunol ; 15(1): 165-175, 2022 01.
Article in English | MEDLINE | ID: mdl-34400793

ABSTRACT

Entamoeba histolytica is a pathogenic protozoan parasite that causes intestinal colitis, diarrhea, and in some cases, liver abscess. Through transcriptomics analysis, we observed that E. histolytica infection was associated with increased expression of IL-33 mRNA in both the human and murine colon. IL-33, the IL-1 family cytokine, is released after cell injury to alert the immune system of tissue damage. Treatment with recombinant IL-33 protected mice from amebic infection and intestinal tissue damage; moreover, blocking IL-33 signaling made mice more susceptible to amebiasis. IL-33 limited the recruitment of inflammatory immune cells and decreased the pro-inflammatory cytokine IL-6 in the cecum. Type 2 immune responses were upregulated by IL-33 treatment during amebic infection. Interestingly, administration of IL-33 protected RAG2-/- mice but not RAG2-/-γc-/- mice, demonstrating that IL-33-mediated protection required the presence of innate lymphoid cells (ILCs). IL-33 induced recruitment of ILC2 but not ILC1 and ILC3 in RAG2-/- mice. At baseline and after amebic infection, there was a significantly higher IL13+ILC2s in C57BL/J mice, which are naturally resistant to amebiasis, than CBA/J mice. Adoptive transfer of ILC2s to RAG2-/-γc-/- mice restored IL-33-mediated protection. These data reveal that the IL-33-ILC2 pathway is an important host defense mechanism against amebic colitis.


Subject(s)
Colon/physiology , Dysentery, Amebic/immunology , Entamoeba histolytica/physiology , Entamoebiasis/immunology , Interleukin-33/genetics , Lymphocytes/immunology , RNA, Messenger/genetics , Animals , Cell Movement , Colon/parasitology , DNA-Binding Proteins/genetics , Disease Resistance , Gene Expression Profiling , Genetic Background , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Signal Transduction , Th1 Cells/immunology , Th2 Cells/immunology
12.
Trends Parasitol ; 37(2): 165-175, 2021 02.
Article in English | MEDLINE | ID: mdl-33502317

ABSTRACT

The protozoan parasite Entamoeba histolytica is the causative agent of amebiasis, an infection that manifests as colitis and, in some cases, liver abscess. A better understanding of host protective factors is key to developing an effective remedy. Recently, significant advances have been made in understanding the mechanisms of MUC2 production by goblet cells upon amebic infection, regulation of antimicrobial peptide production by Paneth cells, the interaction of commensal microbiota with immune stimulation, and host genetics in conferring protection from amebiasis. In addition to host pathways that may serve as potential therapeutic targets, significant progress has also been made with respect to development of a vaccine against amebiasis. Here, we aim to highlight the current understanding and knowledge gaps critically.


Subject(s)
Entamoebiasis/immunology , Host-Parasite Interactions/immunology , Entamoeba histolytica , Entamoebiasis/genetics , Entamoebiasis/parasitology , Entamoebiasis/prevention & control , Goblet Cells/immunology , Goblet Cells/parasitology , Humans , Mucin-2/immunology , Paneth Cells/immunology , Pore Forming Cytotoxic Proteins/immunology , Protozoan Vaccines
13.
mSystems ; 6(5): e0091921, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34609164

ABSTRACT

The pathogen Clostridioides difficile causes toxin-mediated diarrhea and is the leading cause of hospital-acquired infection in the United States. Due to growing antibiotic resistance and recurrent infection, targeting C. difficile metabolism presents a new approach to combat this infection. Genome-scale metabolic network reconstructions (GENREs) have been used to identify therapeutic targets and uncover properties that determine cellular behaviors. Thus, we constructed C. difficile GENREs for a hypervirulent isolate (strain [str.] R20291) and a historic strain (str. 630), validating both with in vitro and in vivo data sets. Growth simulations revealed significant correlations with measured carbon source usage (positive predictive value [PPV] ≥ 92.7%), and single-gene deletion analysis showed >89.0% accuracy. Next, we utilized each GENRE to identify metabolic drivers of both sporulation and biofilm formation. Through contextualization of each model using transcriptomes generated from in vitro and infection conditions, we discovered reliance on the pentose phosphate pathway as well as increased usage of cytidine and N-acetylneuraminate when virulence expression is reduced, which was subsequently supported experimentally. Our results highlight the ability of GENREs to identify novel metabolite signals in higher-order phenotypes like bacterial pathogenesis. IMPORTANCE Clostridioides difficile has become the leading single cause of hospital-acquired infections. Numerous studies have demonstrated the importance of specific metabolic pathways in aspects of C. difficile pathophysiology, from initial colonization to regulation of virulence factors. In the past, genome-scale metabolic network reconstruction (GENRE) analysis of bacteria has enabled systematic investigation of the genetic and metabolic properties that contribute to downstream virulence phenotypes. With this in mind, we generated and extensively curated C. difficile GENREs for both a well-studied laboratory strain (str. 630) and a more recently characterized hypervirulent isolate (str. R20291). In silico validation of both GENREs revealed high degrees of agreement with experimental gene essentiality and carbon source utilization data sets. Subsequent exploration of context-specific metabolism during both in vitro growth and infection revealed consistent patterns of metabolism which corresponded with experimentally measured increases in virulence factor expression. Our results support that differential C. difficile virulence is associated with distinct metabolic programs related to use of carbon sources and provide a platform for identification of novel therapeutic targets.

14.
mBio ; 12(2)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785619

ABSTRACT

Clostridioides difficile, a Gram-positive, spore-forming bacterium, is the primary cause of infectious nosocomial diarrhea. Antibiotics are a major risk factor for C. difficile infection (CDI), as they disrupt the gut microbial community, enabling increased germination of spores and growth of vegetative C. difficile To date, the only single-species bacterial preparation that has demonstrated efficacy in reducing recurrent CDI in humans is nontoxigenic C. difficile Using multiple infection models, we determined that precolonization with a less virulent strain is sufficient to protect from challenge with a lethal strain of C. difficile, surprisingly even in the absence of adaptive immunity. Additionally, we showed that protection is dependent on high levels of colonization by the less virulent strain and that it is mediated by exclusion of the invading strain. Our results suggest that reduction of amino acids, specifically glycine following colonization by the first strain of C. difficile, is sufficient to decrease germination of the second strain, thereby limiting colonization by the lethal strain.IMPORTANCE Antibiotic-associated colitis is often caused by infection with the bacterium Clostridioides difficile In this study, we found that reduction of the amino acid glycine by precolonization with a less virulent strain of C. difficile is sufficient to decrease germination of a second strain. This finding demonstrates that the axis of competition for nutrients can include multiple life stages. This work is important, as it is the first to identify a possible mechanism through which precolonization with C. difficile, a current clinical therapy, provides protection from reinfection. Furthermore, our work suggests that targeting nutrients utilized by all life stages could be an improved strategy for bacterial therapeutics that aim to restore colonization resistance in the gut.


Subject(s)
Antibiosis , Biological Therapy , Clostridioides difficile/physiology , Clostridium Infections/prevention & control , Animals , Clostridioides difficile/classification , Clostridioides difficile/growth & development , Clostridioides difficile/pathogenicity , Clostridium Infections/microbiology , Female , Glycine/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microbiota , Spores, Bacterial/classification , Spores, Bacterial/genetics , Spores, Bacterial/growth & development , Spores, Bacterial/physiology , Virulence
15.
Front Immunol ; 12: 683157, 2021.
Article in English | MEDLINE | ID: mdl-34248966

ABSTRACT

Amebiasis is a neglected tropical disease caused by Entamoeba histolytica. Although the disease burden varies geographically, amebiasis is estimated to account for some 55,000 deaths and millions of infections globally per year. Children and travelers are among the groups with the greatest risk of infection. There are currently no licensed vaccines for prevention of amebiasis, although key immune correlates for protection have been proposed from observational studies in humans. We previously described the development of a liposomal adjuvant formulation containing two synthetic TLR ligands (GLA and 3M-052) that enhanced antigen-specific fecal IgA, serum IgG2a, a mixed IFNγ and IL-17A cytokine profile from splenocytes, and protective efficacy following intranasal administration with the LecA antigen. By applying a statistical design of experiments (DOE) and desirability function approach, we now describe the optimization of the dose of each vaccine formulation component (LecA, GLA, 3M-052, and liposome) as well as the excipient composition (acyl chain length and saturation; PEGylated lipid:phospholipid ratio; and presence of antioxidant, tonicity, or viscosity agents) to maximize desired immunogenicity characteristics while maintaining physicochemical stability. This DOE/desirability index approach led to the identification of a lead candidate composition that demonstrated immune response durability and protective efficacy in the mouse model, as well as an assessment of the impact of each active vaccine formulation component on protection. Thus, we demonstrate that both GLA and 3M-052 are required for statistically significant protective efficacy. We also show that immunogenicity and efficacy results differ in female vs male mice, and the differences appear to be at least partly associated with adjuvant formulation composition.


Subject(s)
Antigens, Protozoan/immunology , Entamoeba histolytica/immunology , Entamoebiasis/immunology , Entamoebiasis/prevention & control , Protozoan Vaccines/immunology , Adjuvants, Immunologic/chemistry , Administration, Intranasal , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Chemical Phenomena , Cytokines/metabolism , Drug Compounding , Entamoebiasis/parasitology , Enzyme-Linked Immunosorbent Assay , Humans , Immunogenicity, Vaccine , Immunoglobulin G/immunology , Liposomes , Mice , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/chemistry , Vaccination
16.
J Clin Invest ; 130(8): 4019-4024, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32369444

ABSTRACT

The microbiome provides resistance to infection. However, the underlying mechanisms are poorly understood. We demonstrate that colonization with the intestinal bacterium Clostridium scindens protects from Entamoeba histolytica colitis via innate immunity. Introduction of C. scindens into the gut microbiota epigenetically altered and expanded bone marrow granulocyte-monocyte progenitors (GMPs) and resulted in increased intestinal neutrophils with subsequent challenge with E. histolytica. Introduction of C. scindens alone was sufficient to expand GMPs in gnotobiotic mice. Adoptive transfer of bone marrow from C. scindens-colonized mice into naive mice protected against amebic colitis and increased intestinal neutrophils. Children without E. histolytica diarrhea also had a higher abundance of Lachnoclostridia. Lachnoclostridia C. scindens can metabolize the bile salt cholate, so we measured deoxycholate and discovered that it was increased in the sera of C. scindens-colonized specific pathogen-free and gnotobiotic mice, as well as in children protected from amebiasis. Administration of deoxycholate alone increased GMPs and provided protection from amebiasis. We elucidated a mechanism by which C. scindens and the microbially metabolized bile salt deoxycholic acid alter hematopoietic precursors and provide innate protection from later infection with E. histolytica.


Subject(s)
Bone Marrow/immunology , Clostridiales/immunology , Dysentery, Amebic/immunology , Entamoeba histolytica/immunology , Gastrointestinal Microbiome/immunology , Animals , Bone Marrow/pathology , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Dysentery, Amebic/microbiology , Dysentery, Amebic/pathology , Humans , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Mice
17.
Nat Med ; 26(4): 608-617, 2020 04.
Article in English | MEDLINE | ID: mdl-32066975

ABSTRACT

The involvement of host immunity in the gut microbiota-mediated colonization resistance to Clostridioides difficile infection (CDI) is incompletely understood. Here, we show that interleukin (IL)-22, induced by colonization of the gut microbiota, is crucial for the prevention of CDI in human microbiota-associated (HMA) mice. IL-22 signaling in HMA mice regulated host glycosylation, which enabled the growth of succinate-consuming bacteria Phascolarctobacterium spp. within the gut microbiome. Phascolarctobacterium reduced the availability of luminal succinate, a crucial metabolite for the growth of C. difficile, and therefore prevented the growth of C. difficile. IL-22-mediated host N-glycosylation is likely impaired in patients with ulcerative colitis (UC) and renders UC-HMA mice more susceptible to CDI. Transplantation of healthy human-derived microbiota or Phascolarctobacterium reduced luminal succinate levels and restored colonization resistance in UC-HMA mice. IL-22-mediated host glycosylation thus fosters the growth of commensal bacteria that compete with C. difficile for the nutritional niche.


Subject(s)
Bacteria/growth & development , Bacteria/metabolism , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Gastrointestinal Microbiome/physiology , Interleukins/physiology , Animals , Bacteria/drug effects , Clostridioides difficile/drug effects , Clostridium Infections/immunology , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/metabolism , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/prevention & control , Female , Gastrointestinal Microbiome/drug effects , Glycosylation/drug effects , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Interleukins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Veillonellaceae/drug effects , Veillonellaceae/growth & development , Veillonellaceae/metabolism , Interleukin-22
18.
mSphere ; 4(1)2019 01 30.
Article in English | MEDLINE | ID: mdl-30700514

ABSTRACT

Clostridium (Clostridioides) difficile, a Gram-positive, anaerobic bacterium, is the leading single cause of nosocomial infections in the United States. A major risk factor for Clostridium difficile infection (CDI) is prior exposure to antibiotics, as they increase susceptibility to CDI by altering the membership of the microbial community enabling colonization. The importance of the gut microbiota in providing protection from CDI is underscored by the reported 80 to 90% success rate of fecal microbial transplants in treating recurrent infections. Adaptive immunity, specifically humoral immunity, is also sufficient to protect from both acute and recurrent CDI. However, the role of the adaptive immune system in mediating clearance of C. difficile has yet to be resolved. Using murine models of CDI, we found that adaptive immunity is dispensable for clearance of C. difficile However, random forest analysis using only two members of the resident bacterial community correctly identified animals that would go on to clear the infection with 66.7% accuracy. These findings indicate that the indigenous gut microbiota independent of adaptive immunity facilitates clearance of C. difficile from the murine gastrointestinal tract.IMPORTANCEClostridium difficile infection is a major cause of morbidity and mortality in hospitalized patients in the United States. Currently, the role of the adaptive immune response in modulating levels of C. difficile colonization is unresolved. This work suggests that the indigenous gut microbiota is a main factor that promotes clearance of C. difficile from the GI tract. Our results show that clearance of C. difficile can occur without contributions from the adaptive immune response. This study also has implications for the design of preclinical studies testing the efficacy of vaccines on clearance of bacterial pathogens, as inherent differences in the baseline community structure of animals may bias findings.


Subject(s)
Adaptive Immunity , Clostridioides difficile/growth & development , Clostridium Infections/immunology , Clostridium Infections/microbiology , Gastrointestinal Microbiome , Microbial Interactions , Animals , Antibody Formation , Disease Models, Animal , Immunity, Cellular , Mice
19.
Cell Host Microbe ; 25(5): 756-765.e5, 2019 May 08.
Article in English | MEDLINE | ID: mdl-31003940

ABSTRACT

Clostridium difficile infection (CDI) is the number one hospital-acquired infection in the United States. CDI is more common and severe in inflammatory bowel disease patients. Here, we studied the mechanism by which prior colitis exacerbates CDI. Mice were given dextran sulfate sodium (DSS) colitis, recovered for 2 weeks, and then were infected with C. difficile. Mortality and CDI severity were increased in DSS-treated mice compared to controls. Severe CDI is dependent on CD4+ T cells, which persist after colitis-associated inflammation subsides. Adoptive transfer of Th17 cells to naive mice is sufficient to increase CDI-associated mortality through elevated IL-17 production. Finally, in humans, the Th17 cytokines IL-6 and IL-23 associate with severe CDI, and patients with high serum IL-6 are 7.6 times more likely to die post infection. These findings establish a central role for Th17 cells in CDI pathogenesis following colitis and identify them as a potential target for preventing severe disease.


Subject(s)
Clostridioides difficile/growth & development , Clostridium Infections/immunology , Colitis/complications , Colitis/pathology , Disease Susceptibility , Th17 Cells/immunology , Adolescent , Adoptive Transfer , Adult , Aged , Animals , Child , Clostridium Infections/mortality , Clostridium Infections/pathology , Colitis/chemically induced , Disease Models, Animal , Female , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/pathology , Interleukin-23 Subunit p19/blood , Interleukin-6/blood , Male , Mice , Middle Aged , Risk Assessment , Survival Analysis , Young Adult
20.
Nat Commun ; 10(1): 2712, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31221971

ABSTRACT

Clostridium difficile (C. difficile) incidence has tripled over the past 15 years and is attributed to the emergence of hypervirulent strains. While it is clear that C. difficile toxins cause damaging colonic inflammation, the immune mechanisms protecting from tissue damage require further investigation. Through a transcriptome analysis, we identify IL-33 as an immune target upregulated in response to hypervirulent C. difficile. We demonstrate that IL-33 prevents C. difficile-associated mortality and epithelial disruption independently of bacterial burden or toxin expression. IL-33 drives colonic group 2 innate lymphoid cell (ILC2) activation during infection and IL-33 activated ILC2s are sufficient to prevent disease. Furthermore, intestinal IL-33 expression is regulated by the microbiota as fecal microbiota transplantation (FMT) rescues antibiotic-associated depletion of IL-33. Lastly, dysregulated IL-33 signaling via the decoy receptor, sST2, predicts C. difficile-associated mortality in human patients. Thus, IL-33 signaling to ILC2s is an important mechanism of defense from C. difficile colitis.


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Immunity, Innate , Interleukin-33/metabolism , Lymphocytes/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Anti-Bacterial Agents/adverse effects , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Clostridioides difficile/pathogenicity , Colon/cytology , Colon/immunology , Colon/microbiology , Colon/pathology , Disease Models, Animal , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/mortality , Enterocolitis, Pseudomembranous/therapy , Fecal Microbiota Transplantation , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Gene Expression Profiling , Humans , Interleukin-33/immunology , Lymphocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Up-Regulation/drug effects , Up-Regulation/immunology , Virulence/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL