Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 720
Filter
1.
Microbes Infect ; 25(8): 105210, 2023.
Article in English | MEDLINE | ID: mdl-37634661

ABSTRACT

Clostridioides difficile (CD) is a major cause of antibiotic-associated diarrhea and pseudomembranous enteritis. C. difficile infection (CDI) is increasingly present in the community and represents a significant burden on the healthcare system. Identification of novel immune-based therapeutic targets from a better understanding of their molecular pathogenesis is urgently required. Toll-like receptor 7 (TLR7) is an important pattern recognition receptor and function as an immune sensor that can trigger host defenses against pathogens, but the relationship between TLR7 and CDI remains unknown. Here, we reported that the expression levels of TLR7 increased significantly in patients and mice with CDI. Absence of TLR7 in mice with CDI demonstrated enhanced bacterial clearance of intestinal contents and reduced intestinal inflammation, edema, injury and prolonged the survival. TLR7 loss decreased the concentrations of tumor necrosis factor (TNF)-α, interferon (IFN)-γ and IFN-α1 in the intestine and improved tissue damage and inflammation. Flow cytometry and immunofluorescence results indicated that TLR7 enhanced leukocyte recruitment in the infected intestine. In-vitro results have shown that TLR7 impairs the phagocytosis and killing ability of macrophages to CD, prompts reactive oxygen species (ROS) production and accelerates apoptosis. To our knowledge, our study first identified TLR7 as a critical factor that contributes to the immunopathology of CDI, suggesting that targeting TLR7 might serve as a potential treatment for CDI.


Subject(s)
Clostridioides difficile , Clostridium Infections , Enterocolitis, Pseudomembranous , Animals , Humans , Mice , Clostridium Infections/microbiology , Enterocolitis, Pseudomembranous/pathology , Inflammation , Toll-Like Receptor 7
2.
Gut Microbes ; 13(1): 1966277, 2021.
Article in English | MEDLINE | ID: mdl-34486488

ABSTRACT

Clostridioides difficile is the causative agent of antibiotic-associated diarrhea, a worldwide public health problem. Different factors can promote the progression of C. difficile infection (CDI), mainly altered intestinal microbiota composition. Microbial species belonging to different domains (i.e., bacteria, archaea, eukaryotes, and even viruses) are synergistically and antagonistically associated with CDI. This review was aimed at updating changes regarding CDI-related human microbiota composition using recent data and an integral approach that included the different microorganism domains. The three domains of life contribute to intestinal microbiota homeostasis at different levels in which relationships among microorganisms could explain the wide range of clinical manifestations. A holistic understanding of intestinal ecosystem functioning will facilitate identifying new predictive factors for infection and developing better treatment and new diagnostic tools, thereby reducing this disease's morbidity and mortality.


Subject(s)
Archaea/classification , Clostridioides difficile/classification , Eukaryota/classification , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/microbiology , Archaea/isolation & purification , Clostridioides difficile/growth & development , Enterocolitis, Pseudomembranous/pathology , Eukaryota/isolation & purification , Humans
3.
Gut Microbes ; 13(1): 1966255, 2021.
Article in English | MEDLINE | ID: mdl-34432545

ABSTRACT

BACKGROUND: Clostridioides difficile is a serious problem for the aging population. Aged mouse model of C. difficile infection (CDI) has emerged as a valuable tool to evaluate the mechanism of aging in CDI. METHODS: We reviewed five published studies utilizing aged mice (7-28 months) for CDI model for findings that may advance our understanding of how aging influences outcome from CDI. RESULTS: Aged mouse models of CDI uniformly demonstrated more severe disease in the old compared to young mice. Diminished neutrophil recruitment to intestinal tissue in aged mice is the most consistent finding. Differences in innate and humoral immune responses were also observed. The effects of aging on the outcome of infection were reversed by pharmacologic or microbiota-targeted interventions. CONCLUSION: The aged mouse presents an important in vivo model to study CDI and elucidate the mechanisms underlying advanced age as an important risk factor for severe disease.


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/pathology , Intestinal Mucosa/immunology , Neutrophil Infiltration/immunology , Aging , Animals , Disease Models, Animal , Enterocolitis, Pseudomembranous/microbiology , Gastrointestinal Microbiome/physiology , Germ-Free Life , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Mice , Neutrophils/immunology , Risk Factors , Severity of Illness Index
4.
Nat Commun ; 12(1): 3748, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145250

ABSTRACT

C. difficile is a major cause of antibiotic-associated gastrointestinal infections. Two C. difficile exotoxins (TcdA and TcdB) are major virulence factors associated with these infections, and chondroitin sulfate proteoglycan 4 (CSPG4) is a potential receptor for TcdB, but its pathophysiological relevance and the molecular details that govern recognition remain unknown. Here, we determine the cryo-EM structure of a TcdB-CSPG4 complex, revealing a unique binding site spatially composed of multiple discontinuous regions across TcdB. Mutations that selectively disrupt CSPG4 binding reduce TcdB toxicity in mice, while CSPG4-knockout mice show reduced damage to colonic tissues during C. difficile infections. We further show that bezlotoxumab, the only FDA approved anti-TcdB antibody, blocks CSPG4 binding via an allosteric mechanism, but it displays low neutralizing potency on many TcdB variants from epidemic hypervirulent strains due to sequence variations in its epitopes. In contrast, a CSPG4-mimicking decoy neutralizes major TcdB variants, suggesting a strategy to develop broad-spectrum therapeutics against TcdB.


Subject(s)
Antigens/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/pathology , Proteoglycans/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antigens/genetics , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Binding Sites/physiology , Broadly Neutralizing Antibodies/pharmacology , Cryoelectron Microscopy , Enterocolitis, Pseudomembranous/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Protein Binding/drug effects , Protein Conformation , Proteoglycans/genetics
5.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805767

ABSTRACT

Novel therapeutics are needed to treat pathologies associated with the Clostridioides difficile binary toxin (CDT), particularly when C. difficile infection (CDI) occurs in the elderly or in hospitalized patients having illnesses, in addition to CDI, such as cancer. While therapies are available to block toxicities associated with the large clostridial toxins (TcdA and TcdB) in this nosocomial disease, nothing is available yet to treat toxicities arising from strains of CDI having the binary toxin. Like other binary toxins, the active CDTa catalytic subunit of CDT is delivered into host cells together with an oligomeric assembly of CDTb subunits via host cell receptor-mediated endocytosis. Once CDT arrives in the host cell's cytoplasm, CDTa catalyzes the ADP-ribosylation of G-actin leading to degradation of the cytoskeleton and rapid cell death. Although a detailed molecular mechanism for CDT entry and host cell toxicity is not yet fully established, structural and functional resemblances to other binary toxins are described. Additionally, unique conformational assemblies of individual CDT components are highlighted herein to refine our mechanistic understanding of this deadly toxin as is needed to develop effective new therapeutic strategies for treating some of the most hypervirulent and lethal strains of CDT-containing strains of CDI.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridioides difficile/pathogenicity , Cross Infection/drug therapy , Enterocolitis, Pseudomembranous/drug therapy , Enterotoxins/antagonists & inhibitors , ADP-Ribosylation/drug effects , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/deficiency , Actins/genetics , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Binding Sites , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Cross Infection/metabolism , Cross Infection/microbiology , Cross Infection/pathology , Endocytosis/drug effects , Enterocolitis, Pseudomembranous/metabolism , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Enterotoxins/chemistry , Enterotoxins/genetics , Enterotoxins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , Protein Structure, Secondary
7.
PLoS Pathog ; 16(9): e1008852, 2020 09.
Article in English | MEDLINE | ID: mdl-32960931

ABSTRACT

Enzymatic inactivation of Rho-family GTPases by the glucosyltransferase domain of Clostridioides difficile Toxin B (TcdB) gives rise to various pathogenic effects in cells that are classically thought to be responsible for the disease symptoms associated with C. difficile infection (CDI). Recent in vitro studies have shown that TcdB can, under certain circumstances, induce cellular toxicities that are independent of glucosyltransferase (GT) activity, calling into question the precise role of GT activity. Here, to establish the importance of GT activity in CDI disease pathogenesis, we generated the first described mutant strain of C. difficile producing glucosyltransferase-defective (GT-defective) toxin. Using allelic exchange (AE) technology, we first deleted tcdA in C. difficile 630Δerm and subsequently introduced a deactivating D270N substitution in the GT domain of TcdB. To examine the role of GT activity in vivo, we tested each strain in two different animal models of CDI pathogenesis. In the non-lethal murine model of infection, the GT-defective mutant induced minimal pathology in host tissues as compared to the profound caecal inflammation seen in the wild-type and 630ΔermΔtcdA (ΔtcdA) strains. In the more sensitive hamster model of CDI, whereas hamsters in the wild-type or ΔtcdA groups succumbed to fulminant infection within 4 days, all hamsters infected with the GT-defective mutant survived the 10-day infection period without primary symptoms of CDI or evidence of caecal inflammation. These data demonstrate that GT activity is indispensable for disease pathogenesis and reaffirm its central role in disease and its importance as a therapeutic target for small-molecule inhibition.


Subject(s)
Bacterial Proteins , Bacterial Toxins , Clostridioides difficile , Enterocolitis, Pseudomembranous , Glucosyltransferases , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Clostridioides difficile/enzymology , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Cricetinae , Disease Models, Animal , Enterocolitis, Pseudomembranous/enzymology , Enterocolitis, Pseudomembranous/genetics , Enterocolitis, Pseudomembranous/pathology , Female , Gene Deletion , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Male , Mice
8.
Gastroenterology ; 159(4): 1431-1443.e6, 2020 10.
Article in English | MEDLINE | ID: mdl-32574621

ABSTRACT

BACKGROUND & AIMS: The protease plasmin is an important wound healing factor, but it is not clear how it affects gastrointestinal infection-mediated damage, such as that resulting from Clostridioides difficile. We investigated the role of plasmin in C difficile-associated disease. This bacterium produces a spore form that is required for infection, so we also investigated the effects of plasmin on spores. METHODS: C57BL/6J mice expressing the precursor to plasmin, the zymogen human plasminogen (hPLG), or infused with hPLG were infected with C difficile, and disease progression was monitored. Gut tissues were collected, and cytokine production and tissue damage were analyzed by using proteomic and cytokine arrays. Antibodies that inhibit either hPLG activation or plasmin activity were developed and structurally characterized, and their effects were tested in mice. Spores were isolated from infected patients or mice and visualized using super-resolution microscopy; the functional consequences of hPLG binding to spores were determined. RESULTS: hPLG localized to the toxin-damaged gut, resulting in immune dysregulation with an increased abundance of cytokines (such as interleukin [IL] 1A, IL1B, IL3, IL10, IL12B, MCP1, MP1A, MP1B, GCSF, GMCSF, KC, TIMP-1), tissue degradation, and reduced survival. Administration of antibodies that inhibit plasminogen activation reduced disease severity in mice. C difficile spores bound specifically to hPLG and active plasmin degraded their surface, facilitating rapid germination. CONCLUSIONS: We found that hPLG is recruited to the damaged gut, exacerbating C difficile disease in mice. hPLG binds to C difficile spores, and, upon activation to plasmin, remodels the spore surface, facilitating rapid spore germination. Inhibitors of plasminogen activation might be developed for treatment of C difficile or other infection-mediated gastrointestinal diseases.


Subject(s)
Clostridioides difficile/drug effects , Enterocolitis, Pseudomembranous/etiology , Enterocolitis, Pseudomembranous/pathology , Plasminogen/pharmacology , Spores, Bacterial/drug effects , Animals , Disease Models, Animal , Humans , Intestine, Small , Mice , Mice, Inbred C57BL
9.
Int Microbiol ; 23(4): 575-587, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32388701

ABSTRACT

PURPOSE: Clostridioides difficile toxin B (TcdB) plays a critical role in C. difficile infection (CDI), a common and costly healthcare-associated disease. The aim of the current study was to explore the intracellular and potent systemic effects of TcdB on human colon epithelial cells utilizing Gene Expression Omnibus and bioinformatic methods. METHODS: Two datasets (GSE63880 and GSE29008) were collected to extract data components of mRNA of TcdB-treated human colon epithelial cells; "limma" package of "R" software was used to screen the differential genes, and "pheatmap" package was applied to construct heat maps for the differential genes; Metascape website was utilized for protein-protein interaction network and Molecular Complex Detection analysis, and Genome Ontology (GO) was used to analyze the selected differential genes. Quantitative real-time PCR (qRT-PCR) and Western blot were performed to validate the expression of hub genes. RESULTS: GO terms involved in DNA replication and cell cycle were identified significantly enriched in TcdB-treated human colon epithelial cells. Moreover, the decreased expression of DNA replication-related genes, MCM complex, and CDC45 in C. difficile (TcdA-/TcdB+)-infected Caco-2 cells were validated via qRT-PCR and Western blot assays. CONCLUSIONS: In conclusion, the integrated analysis of different gene expression datasets allowed us to identify a set of genes and GO terms underlying the mechanisms of CDI induced by TcdB. It would aid in understanding of the molecular mechanisms underlying TcdB-exposed colon epithelial cells and provide the basis for developing diagnosis biomarkers, treatment, and prevention strategies.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/pathogenicity , Computational Biology/methods , Enterocolitis, Pseudomembranous/pathology , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Caco-2 Cells , Cell Line, Tumor , Clostridioides difficile/genetics , Enterocolitis, Pseudomembranous/microbiology , Epithelial Cells/pathology , Gene Expression/genetics , Humans , Intestinal Mucosa/pathology , Protein Array Analysis , Protein Interaction Maps
10.
Gut Microbes ; 12(1): 1725220, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32129694

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation that includes Crohn´s disease (CD) and ulcerative colitis (UC). Although the etiology is still unknown, some specific factors have been directly related to IBD, including genetic factors, abnormal intestinal immunity, and/or gut microbiota modifications. Recent findings highlight the primary role of the gut microbiota closely associated with a persistent inappropriate inflammatory response. This gut environment of dysbiosis in a susceptible IBD host can increasingly worsen and lead to colonization and infection with some opportunistic pathogens, especially Clostridium difficile. C. difficile is an intestinal pathogen considered the main cause of antibiotic-associated diarrhea and colitis and an important complication of IBD, which can trigger or worsen an IBD flare. Recent findings have highlighted the loss of bacterial cooperation in the gut ecosystem, as well as the pronounced intestinal dysbiosis, in patients suffering from IBD and concomitant C. difficile infection (CDI). The results of intestinal microbiota studies are still limited and often difficult to compare because of the variety of disease conditions. However, these data provide important clues regarding the main modifications and interrelations in the complicated gut ecosystem to better understand both diseases and to take advantage of the development of new therapeutic strategies. In this review, we analyze in depth the gut microbiota changes associated with both forms of IBD and CDI and their similarity with the dysbiosis that occurs in CDI. We also discuss the metabolic pathways that favor the proliferation or decrease in several important taxa directly related to the disease.


Subject(s)
Dysbiosis/microbiology , Enterocolitis, Pseudomembranous/microbiology , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/microbiology , Clostridioides difficile/isolation & purification , Enterocolitis, Pseudomembranous/pathology , Fecal Microbiota Transplantation/methods , Humans , Intestines/microbiology , Intestines/pathology
11.
J Clin Invest ; 130(5): 2377-2390, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31990686

ABSTRACT

Colitis caused by Clostridium difficile infection is a growing cause of human morbidity and mortality, especially after antibiotic use in health care settings. The natural immunity of newborn infants and protective host immune mediators against C. difficile infection are not fully understood, with data suggesting that inflammation can be either protective or pathogenic. Here, we show an essential role for IL-17A produced by γδ T cells in host defense against C. difficile infection. Fecal extracts from children with C. difficile infection showed increased IL-17A and T cell receptor γ chain expression, and IL-17 production by intestinal γδ T cells was efficiently induced after infection in mice. C. difficile-induced tissue inflammation and mortality were markedly increased in mice deficient in IL-17A or γδ T cells. Neonatal mice, with naturally expanded RORγt+ γδ T cells poised for IL-17 production were resistant to C. difficile infection, whereas elimination of γδ T cells or IL-17A each efficiently overturned neonatal resistance against infection. These results reveal an expanded role for IL-17-producing γδ T cells in neonatal host defense against infection and provide a mechanistic explanation for the clinically observed resistance of infants to C. difficile colitis.


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/prevention & control , Interleukin-17/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Animals , Enterocolitis, Pseudomembranous/genetics , Enterocolitis, Pseudomembranous/pathology , Female , Humans , Interleukin-17/genetics , Male , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/genetics
12.
FASEB J ; 34(2): 2198-2212, 2020 02.
Article in English | MEDLINE | ID: mdl-31907988

ABSTRACT

Clostridioides difficile (formerly Clostridium difficile; C difficile), the leading cause of nosocomial antibiotic-associated colitis and diarrhea in the industrialized world, triggers colonic disease through the release two toxins, toxin A (TcdA) and toxin B (TcdB), glucosyltransferases that modulate monomeric G-protein function and alter cytoskeletal function. The initial degree of the host immune response to C difficile and its pathogenic toxins is a common indicator of disease severity and infection recurrence. Thus, targeting the intestinal inflammatory response during infection could significantly decrease disease morbidity and mortality. In the current study, we sought to interrogate the influence of the pregnane X receptor (PXR), a modulator of xenobiotic and detoxification responses, which can sense and respond to microbial metabolites and modulates inflammatory activity, during exposure to TcdA and TcdB. Following intrarectal exposure to TcdA/B, PXR-deficient mice (Nr1i2-/- ) exhibited reduced survival, an effect that was associated with increased levels of innate immune cell influx. This exacerbated response was associated with a twofold increase in the expression of Tlr4. Furthermore, while broad-spectrum antibiotic treatment (to deplete the intestinal microbiota) did not alter the responses in Nr1i2-/- mice, blocking TLR4 signaling significantly reduced TcdA/B-induced disease severity and immune responses in these mice. Lastly, to assess the therapeutic potential of targeting the PXR, we activated the PXR with pregnenolone 16α-carbonitrile (PCN) in wild-type mice, which greatly reduced the severity of TcdA/B-induced damage and intestinal inflammation. Taken together, these data suggest that the PXR plays a role in the host's response to TcdA/B and may provide a novel target to dampen the inflammatory tissue damage in C difficile infections.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile , Enterocolitis, Pseudomembranous/metabolism , Enterotoxins/metabolism , Pregnane X Receptor/metabolism , Signal Transduction , Animals , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/genetics , Enterocolitis, Pseudomembranous/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Pregnane X Receptor/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
13.
J Infect Chemother ; 26(3): 305-308, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31822448

ABSTRACT

A 74-year-old male was referred to our critical care department for refractory severe watery diarrhea with advanced leukocytosis (over 70,000/µl) after multiple administrations of eradication therapy against Helicobacter pylori (HP). He was diagnosed as having fulminant colitis due to Clostridioides difficile after antimicrobial eradication therapy. He was given intravenous metronidazole and oral vancomycin. He also received supportive therapy including continuous hemodiafiltration for severe metabolic acidosis. However, despite emergency open sigmoidectomy, he died. The C. difficile isolate recovered was PCR-ribotype 002, which was positive for toxins A and B but negative for binary toxin. HP eradication therapy for prevention of chronic gastritis and stomach cancer is now in widespread use. Although such secondary severe complications are rare, we consider it to be necessary to pay sufficient attention when administering HP eradication therapy.


Subject(s)
Anti-Bacterial Agents/adverse effects , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/chemically induced , Helicobacter Infections/drug therapy , Proton Pump Inhibitors/adverse effects , Acute Disease , Aged , Clostridioides difficile/isolation & purification , Colectomy , Colon, Sigmoid/pathology , Colon, Sigmoid/surgery , Drug Therapy, Combination/adverse effects , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Enterocolitis, Pseudomembranous/therapy , Fatal Outcome , Humans , Male
14.
Vet Pathol ; 56(6): 959-963, 2019 11.
Article in English | MEDLINE | ID: mdl-31382854

ABSTRACT

Clostridium difficile is a well-documented cause of enterocolitis in several species, including humans, with limited documentation in New World nonhuman primates. We report several cases of C. difficile-associated pseudomembranous enterocolitis, including a case in a Geoffroy's spider monkey (Ateles geoffroyi) and several cases in common marmosets (Callithrix jacchus). The histologic lesions included a spectrum of severity, with most cases characterized by the classic "volcano" lesions described in humans and several other animal species. C. difficile was isolated from the colon of the spider monkey, while the presence of toxin A or toxin B or of the genes of toxin A or B by polymerase chain reaction served as corroborative evidence in several affected marmosets. C. difficile should be considered a cause of enterocolitis in these species.


Subject(s)
Ateles geoffroyi/microbiology , Callithrix/microbiology , Clostridioides difficile/isolation & purification , Enterocolitis, Pseudomembranous/veterinary , Monkey Diseases/microbiology , Animals , Clostridioides difficile/genetics , Colon/microbiology , Colon/pathology , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Female , Male , Monkey Diseases/pathology
17.
Proc Natl Acad Sci U S A ; 116(9): 3774-3783, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808763

ABSTRACT

Establishing causal links between bacterial metabolites and human intestinal disease is a significant challenge. This study reveals the molecular basis of antibiotic-associated hemorrhagic colitis (AAHC) caused by intestinal resident Klebsiella oxytoca Colitogenic strains produce the nonribosomal peptides tilivalline and tilimycin. Here, we verify that these enterotoxins are present in the human intestine during active colitis and determine their concentrations in a murine disease model. Although both toxins share a pyrrolobenzodiazepine structure, they have distinct molecular targets. Tilimycin acts as a genotoxin. Its interaction with DNA activates damage repair mechanisms in cultured cells and causes DNA strand breakage and an increased lesion burden in cecal enterocytes of colonized mice. In contrast, tilivalline binds tubulin and stabilizes microtubules leading to mitotic arrest. To our knowledge, this activity is unique for microbiota-derived metabolites of the human intestine. The capacity of both toxins to induce apoptosis in intestinal epithelial cells-a hallmark feature of AAHC-by independent modes of action, strengthens our proposal that these metabolites act collectively in the pathogenicity of colitis.


Subject(s)
Enterocolitis, Pseudomembranous/genetics , Enterotoxins/metabolism , Host Microbial Interactions/genetics , Klebsiella oxytoca/genetics , Animals , Benzodiazepinones/metabolism , Benzodiazepinones/toxicity , DNA Damage/drug effects , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Enterotoxins/biosynthesis , Epithelial Cells/microbiology , Epithelial Cells/pathology , Humans , Intestines/microbiology , Intestines/pathology , Klebsiella Infections/genetics , Klebsiella Infections/microbiology , Klebsiella oxytoca/metabolism , Klebsiella oxytoca/pathogenicity , Mice , Microtubules/drug effects , Oxyquinoline/analogs & derivatives , Oxyquinoline/metabolism , Oxyquinoline/toxicity , Peptides/metabolism , Peptides/toxicity
19.
Front Immunol ; 10: 2866, 2019.
Article in English | MEDLINE | ID: mdl-31921134

ABSTRACT

Colonization with Clostridioides difficile occurs in up to half of infants under the age of 3 months, is strongly influenced by feeding modality and is largely asymptomatic. In spite of this, C. difficile's presence has been associated with susceptibility to chronic disease later in childhood, perhaps by promoting or benefiting from changes in infant gut microbiome development, including colonization with pathogenic bacteria and disrupted production of microbial bioactive metabolites and proteins. In this study, the microbiomes of 1554 infants from the CHILD Cohort Study were described according to C. difficile colonization status and feeding mode at 3-4 months of age. C. difficile colonization was associated with a different gut microbiome profile in exclusively breastfed (EBF) vs. exclusively formula fed (EFF) infants. EBF infants colonized with C. difficile had an increased relative abundance of Firmicutes and Proteobacteria, decreased relative abundance of Bifidobacteriaceae, greater microbiota alpha-diversity, greater detectable fecal short chain fatty acids (SCFA), and lower detectable fecal secretory Immunoglobulin A (sIgA) than those not colonized. Similar but less pronounced differences were seen among partially breastfed infants (PBF) but EFF infants did not possess these differences in the gut microbiome according to colonization status. Thus, breastfed infants colonized with C. difficile appear to possess a gut microbiome that differs from non-colonized infants and resembles that of EFF infants, but the driving force and direction of this association remains unknown. Understanding these compositional differences as drivers of C. difficile colonization may be important to ensure future childhood health.


Subject(s)
Breast Feeding , Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/mortality , Gastrointestinal Microbiome/immunology , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/pathology , Female , Humans , Infant , Infant, Newborn , Male , Prospective Studies
20.
Pathog Dis ; 76(3)2018 04 01.
Article in English | MEDLINE | ID: mdl-29718184

ABSTRACT

Numerous pathogens including Clostridium difficile and Yersinia pestis have evolved toxins or effectors targeting GTPases from the RhoA subfamily (RhoA/B/C) to inhibit or hijack the host cytoskeleton dynamics. The resulting impairment of RhoA GTPases activity is sensed by the host via an innate immune complex termed the pyrin inflammasome in which caspase-1 is activated. The cascade leading to activation of the pyrin inflammasome has been recently uncovered. In this review, following a brief presentation of RhoA GTPases-modulating toxins, we present the pyrin inflammasome and its regulatory mechanisms. Furthermore, we discuss how some pathogens have developed strategies to escape detection by the pyrin inflammasome. Finally, we present five monogenic autoinflammatory diseases associated with pyrin inflammasome deregulation. The molecular insights provided by the study of these diseases and the corresponding mutations on pyrin inflammasome regulation and activation are presented.


Subject(s)
Bacterial Toxins/immunology , Enterocolitis, Pseudomembranous/immunology , Inflammasomes/immunology , Plague/immunology , Pyrin/immunology , rhoA GTP-Binding Protein/immunology , Animals , Autoimmunity , Bacterial Toxins/biosynthesis , Clostridioides difficile/immunology , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/genetics , Inflammation , Isoenzymes/genetics , Isoenzymes/immunology , Plague/microbiology , Plague/pathology , Pyrin/genetics , Syndrome , Yersinia pestis/immunology , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , rhoA GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...