Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 442
1.
Anaerobe ; 86: 102841, 2024 Apr.
Article En | MEDLINE | ID: mdl-38521227

OBJECTIVES: Clostridioides difficile infection (CDI) is the leading hospital-acquired infection in North America. While previous work on fecal microbiota transplantation (FMT), a highly effective treatment for CDI, has focused on colonization resistance mounted against C. difficile by FMT-delivered commensals, the effects of FMT on host gene expression are relatively unexplored. This study aims to identify transcriptional changes associated with FMT, particularly changes associated with protective immune responses. METHODS: Gene expression was assessed on day 2 and day 7 after FMT in mice after antibiotic-induced dysbiosis. Flow cytometry was also performed on colon and mesenteric lymph nodes at day 7 to investigate changes in immune cell populations. RESULTS: FMT administration after antibiotic-induced dysbiosis successfully restored microbial alpha diversity to levels of donor mice by day 7 post-FMT. Bulk RNA sequencing of cecal tissue at day 2 identified immune genes, including both pro-inflammatory and Type 2 immune pathways as upregulated after FMT. RNA sequencing was repeated on day 7 post-FMT, and expression of these immune genes was decreased along with upregulation of genes associated with restoration of intestinal homeostasis. Immunoprofiling on day 7 identified increased colonic CD45+ immune cells that exhibited dampened Type 1 and heightened regulatory and Type 2 responses. These include an increased abundance of eosinophils, alternatively activated macrophages, Th2, and T regulatory cell populations. CONCLUSION: These results highlight the impact of FMT on host gene expression, providing evidence that FMT restores intestinal homeostasis after antibiotic treatment and facilitates tolerogenic and Type 2 immune responses.


Clostridium Infections , Disease Models, Animal , Fecal Microbiota Transplantation , Animals , Fecal Microbiota Transplantation/methods , Mice , Clostridium Infections/therapy , Clostridium Infections/immunology , Clostridium Infections/microbiology , Gastrointestinal Microbiome , Dysbiosis/therapy , Clostridioides difficile/immunology , Immune Tolerance , Mice, Inbred C57BL
2.
Nature ; 623(7989): 1009-1016, 2023 Nov.
Article En | MEDLINE | ID: mdl-37968387

Iron is indispensable for almost all forms of life but toxic at elevated levels1-4. To survive within their hosts, bacterial pathogens have evolved iron uptake, storage and detoxification strategies to maintain iron homeostasis1,5,6. Recent studies showed that three Gram-negative environmental anaerobes produce iron-containing ferrosome granules7,8. However, it remains unclear whether ferrosomes are generated exclusively by Gram-negative bacteria. The Gram-positive bacterium Clostridioides difficile is the leading cause of nosocomial and antibiotic-associated infections in the USA9. Here we report that C. difficile undergoes an intracellular iron biomineralization process and stores iron in membrane-bound ferrosome organelles containing non-crystalline iron phosphate biominerals. We found that a membrane protein (FezA) and a P1B6-ATPase transporter (FezB), repressed by both iron and the ferric uptake regulator Fur, are required for ferrosome formation and play an important role in iron homeostasis during transition from iron deficiency to excess. Additionally, ferrosomes are often localized adjacent to cellular membranes as shown by cryo-electron tomography. Furthermore, using two mouse models of C. difficile infection, we demonstrated that the ferrosome system is activated in the inflamed gut to combat calprotectin-mediated iron sequestration and is important for bacterial colonization and survival during C. difficile infection.


Clostridioides difficile , Clostridium Infections , Ferric Compounds , Host Microbial Interactions , Iron , Organelles , Animals , Mice , Clostridioides difficile/growth & development , Clostridioides difficile/immunology , Clostridioides difficile/metabolism , Clostridium Infections/immunology , Clostridium Infections/metabolism , Clostridium Infections/microbiology , Iron/metabolism , Organelles/metabolism , Homeostasis , Ferric Compounds/metabolism , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cryoelectron Microscopy , Electron Microscope Tomography , Disease Models, Animal , Leukocyte L1 Antigen Complex/metabolism , Microbial Viability , Inflammation/metabolism , Inflammation/microbiology , Intestines/metabolism , Intestines/microbiology
3.
Anaerobe ; 72: 102475, 2021 Dec.
Article En | MEDLINE | ID: mdl-34752901

BACKGROUND: Determination of the humoral response to Clostridioides difficile (CD) toxins could be of great value in the management of patients with CD infection (CDI). METHODS: A prospective study was conducted on the clinical characteristics and humoral response in patients with CDI. Determination of ELISA IgG CD anti-toxin B (tgcBiomics, Germany) was performed. The following dilutions were planned for each patient, 1:100, 1: 200, 1: 400, 1: 800: 1: 1600. A significant concentration of antibody was considered to be present in each dilution if an optical density 0.2 units higher than the negative control of the technique was evident. RESULTS: Eighty-five patients were included during the study period, November 2018-February 2020. The median age was 73 years (interquartile range: 62.5-85 years), with female predominance (45 patients, 52.9%). Thirty-nine patients (45.9%) had a severe infection. Seven patients (8.2%) had suffered an episode of CDI in the previous three months. Seventeen patients (20%) had one or more recurrent episodes during the three-month follow-up: No patient died during admission or required surgery for severe-complicated infection. The incidence of recurrence in patients with no antibody detected at 1:400 dilution was 25.4% (16 patients) while it was 4.3% (one patient) in patients with antibody present at that dilution (p = 0.03). Liver cirrhosis was associated with higher humoral response against CD. CONCLUSIONS: Antibodies IgG CD anti-toxin B detection at a dilution of 1:400, using a B ELISA technique, effectively identified patients at increased risk of recurrence. This information could help assist in the management of patients.


Clostridioides difficile/immunology , Clostridium Infections/immunology , Clostridium Infections/microbiology , Host-Pathogen Interactions/immunology , Immunity, Humoral , Aged , Aged, 80 and over , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Comorbidity , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Middle Aged , Recurrence , Spain
4.
Gut Microbes ; 13(1): 1966255, 2021.
Article En | MEDLINE | ID: mdl-34432545

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.


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
5.
Gut Microbes ; 13(1): 1968258, 2021.
Article En | MEDLINE | ID: mdl-34432564

Clostridioides difficile infection is currently the leading cause of nosocomial antibiotic-associated diarrhea and pseudomembranous colitis worldwide. Cathelicidins, a major group of natural antimicrobial peptides, have antimicrobial and immunomodulatory activities in Clostridioides difficile infection. Here, we have shown that cytokine IL-27 induced human cathelicidin antimicrobial peptide (LL-37) expression in primary human colonic epithelial cells. IL-27 receptor-deficient mice had impaired expression of cathelicidin-related antimicrobial peptide (CRAMP, mouse homolog for human LL-37) after Clostridioides difficile infection, and restoration of CRAMP improved Clostridium difficile clearance and reduced mortality in IL-27 receptor-deficient mice after Clostridioides difficile challenge. In clinical samples from 119 patients with Clostridioides difficile infection, elevated levels of IL-27 were positively correlated with LL-37 in the sera and stools. These findings suggest that IL-27 may be involved in host immunity against Clostridioides difficile infection via induction of LL-37/CRAMP. Therefore, IL-27-LL-37 axis may be a valuable pathway in the development of immune-based therapy.


Cathelicidins/metabolism , Clostridioides difficile/immunology , Interleukins/genetics , Interleukins/metabolism , Animals , Cell Line , Enterocolitis, Pseudomembranous/therapy , Epithelial Cells/metabolism , Humans , Immunotherapy/methods , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL
6.
Infect Immun ; 89(11): e0043821, 2021 10 15.
Article En | MEDLINE | ID: mdl-34424751

All clinical Clostridioides difficile strains identified to date express a surface capsule-like polysaccharide structure known as polysaccharide II (PSII). The PSII antigen is immunogenic and, when conjugated to a protein carrier, induces a protective antibody response in animal models. Given that CD1d-restricted natural killer T (NKT) cells promote antibody responses, including those against carbohydrates, we tested the hypothesis that immunization with PSII and a CD1d-binding glycolipid adjuvant could lead to enhanced protection against a live C. difficile challenge. We purified PSII from a clinical isolate of C. difficile and immunized B6 mice with PSII alone or PSII plus the CD1d-binding glycolipid α-galactosylceramide (α-GC). PSII-specific IgM and IgG titers were evident in sera from immunized mice. The inclusion of α-GC had a modest influence on isotype switch but increased the IgG1/IgG2c ratio. Enhanced protection against C. difficile disease was achieved by inclusion of the α-GC ligand and was associated with reduced bacterial numbers in fecal pellets. In contrast, NKT-deficient Traj18-/- mice were not protected by the PSII/α-GC immunization modality. Absence of NKT cells similarly had a modest effect on isotype switch, but ratios of IgG1/IgG2c decreased. These results indicate that α-GC-driven NKT cells move the humoral immune response against C. difficile PSII antigen toward Th2-driven IgG1 and may contribute to augmented protection. This study suggests that NKT activation represents a pathway for additional B-cell help that could be used to supplement existing efforts to develop vaccines against polysaccharides derived from C. difficile and other pathogens.


Antigens, Bacterial/immunology , Clostridioides difficile/immunology , Galactosylceramides/immunology , Immunoglobulin G/blood , Natural Killer T-Cells/immunology , Polysaccharides, Bacterial/immunology , Animals , Antibodies, Bacterial/blood , Female , Immunization , Lymphocyte Activation , Mice , Mice, Inbred C57BL
7.
Sci Rep ; 11(1): 14215, 2021 07 09.
Article En | MEDLINE | ID: mdl-34244557

Clostridium difficile is a spore-forming gram-positive bacterium, recognized as the primary cause of antibiotic-associated nosocomial diarrhoea. Clostridium difficile infection (CDI) has emerged as a major health-associated infection with increased incidence and hospitalization over the years with high mortality rates. Contamination and infection occur after ingestion of vegetative spores, which germinate in the gastro-intestinal tract. The surface layer protein and flagellar proteins are responsible for the bacterial colonization while the spore coat protein, is associated with spore colonization. Both these factors are the main concern of the recurrence of CDI in hospitalized patients. In this study, the CotE, SlpA and FliC proteins are chosen to form a multivalent, multi-epitopic, chimeric vaccine candidate using the immunoinformatics approach. The overall reliability of the candidate vaccine was validated in silico and the molecular dynamics simulation verified the stability of the vaccine designed. Docking studies showed stable vaccine interactions with Toll-Like Receptors of innate immune cells and MHC receptors. In silico codon optimization of the vaccine and its insertion in the cloning vector indicates a competent expression of the modelled vaccine in E. coli expression system. An in silico immune simulation system evaluated the effectiveness of the candidate vaccine to trigger a protective immune response.


Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/drug therapy , Clostridium Infections/immunology , Computational Biology/methods , Escherichia coli/metabolism , Humans
8.
Anaerobe ; 70: 102381, 2021 Aug.
Article En | MEDLINE | ID: mdl-34082120

Clostridioides difficile is a gram-positive, spore-forming anaerobic bacterium, and the leading cause of antibiotic-associated diarrhea worldwide. During C. difficile infection, spores germinate in the presence of bile acids into vegetative cells that subsequently colonize the large intestine and produce toxins. In this study, we demonstrated that C. difficile spores can universally adhere to, and be phagocytosed by, murine macrophages. Only spores from toxigenic strains were able to significantly stimulate the production of inflammatory cytokines by macrophages and subsequently induce significant cytotoxicity. Spores from the isogenic TcdA and TcdB double mutant induced significantly lower inflammatory cytokines and cytotoxicity in macrophages, and these activities were restored by pre-exposure of the spores to either toxins. These findings suggest that during sporulation, spores might be coated with C. difficile toxins from the environment, which could affect C. difficile pathogenesis in vivo.


Clostridioides difficile/immunology , Clostridium Infections/immunology , Cytokines/immunology , Macrophages/immunology , Spores, Bacterial/immunology , Animals , Bacterial Toxins/immunology , Clostridioides difficile/genetics , Clostridium Infections/genetics , Clostridium Infections/microbiology , Cytokines/genetics , Humans , Macrophages/microbiology , Mice , RAW 264.7 Cells , Spores, Bacterial/genetics
9.
Infect Immun ; 89(10): e0027421, 2021 09 16.
Article En | MEDLINE | ID: mdl-34097471

The pathology associated with Clostridioides difficile disease is caused in large part by TcdB, an intracellular bacterial toxin that inactivates small GTPases. Despite C. difficile causing enteric disease, antitoxin IgG is a clear correlate of protection against infection-associated pathology. Immunization with TcdB-based immunogens or passive transfer of monoclonal antibodies specific for the TcdB carboxy-terminal domain (CTD) confers protection following C. difficile infection. Whether the mechanism by which circulating IgG is delivered to the gut depends on specific receptor-mediated transport or is solely reflective of infection-induced damage to the gut remains unclear. Here, we tested the hypothesis that neonatal Fc receptor (FcRn) is required for the delivery of systemic TcdB-specific IgG to the gut and protection against C. difficile-associated pathology. FcRn-expressing mice and FcRn-deficient littermates were immunized subcutaneously with Alhydrogel adjuvant-adsorbed CTD before challenge with live C. difficile spores. FcRn was required for the delivery of systemic TcdB-specific IgG to the gut and for vaccine-induced protection against C. difficile-associated disease. The lack of FcRn expression had minimal effects on the composition of the gut microbiome and did not affect susceptibility to C. difficile infection in nonimmunized mice. In further experiments, intraperitoneal injection of immune sera in FcRn-deficient mice led to the transport of protective IgG to the gut independently of infection, confirming a reported method of bypassing the FcRn. Our results reveal an FcRn-dependent mechanism by which systemic immunization-induced IgG protects the gut during enteric C. difficile infection. These findings may be beneficial for the targeting of C. difficile-specific IgG to the gut.


Clostridioides difficile/immunology , Clostridium Infections/immunology , Digestive System/immunology , Digestive System/microbiology , Disease Susceptibility/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antitoxins/immunology , Bacterial Toxins/immunology , Clostridium Infections/microbiology , Disease Susceptibility/microbiology , Enterotoxins/immunology , Female , Immunity/immunology , Immunization/methods , Male , Mice , Mice, Inbred C57BL , Vaccination/methods
10.
PLoS One ; 16(5): e0252211, 2021.
Article En | MEDLINE | ID: mdl-34043688

Clostridioides difficile (C. difficile) infection is a major public health problem worldwide. The current treatment of C. difficile-associated diarrhea relies on the use of antibacterial agents. However, recurrences are frequent. The main virulence factors of C. difficile are two secreted cytotoxic proteins toxin A and toxin B. Alternative research exploring toxin binding by resins found a reduced rate of recurrence by administration of tolevamer. Hence, binding of exotoxins may be useful in preventing a relapse provided that the adsorbent is innocuous. Here, we examined the toxin binding capacity of G-PUR®, a purified version of natural clinoptilolite-tuff. Our observations showed that the purified clinoptilolite-tuff adsorbed clinically relevant amounts of C. difficile toxins A and B in vitro and neutralized their action in a Caco-2 intestinal model. This conclusion is based on four independent sets of findings: G-PUR® abrogated toxin-induced (i) RAC1 glucosylation, (ii) redistribution of occludin, (iii) rarefaction of the brush border as visualized by scanning electron microscopy and (iv) breakdown of the epithelial barrier recorded by transepithelial electrical resistance monitoring. Finally, we confirmed that the epithelial monolayer tolerated G-PUR® over a wide range of particle densities. Our findings justify the further exploration of purified clinoptilolite-tuff as a safe agent in the treatment and/or prevention of C. difficile-associated diarrhea.


Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Enterotoxins/metabolism , Virulence Factors/metabolism , Zeolites/pharmacology , Caco-2 Cells , Humans , Protein Binding
11.
Sci Rep ; 11(1): 9940, 2021 05 11.
Article En | MEDLINE | ID: mdl-33976336

Clostridioides difficile (C. difficile) is an opportunistic anaerobic bacterium that causes severe diseases of the digestive tract of humans and animals. One of the possible methods of preventing C. difficile infection is to develop a vaccine. The most promising candidates for vaccine antigens are the proteins involved in the adhesion phenomena. Among them, the FliC and FliD are considered to be suitable candidates. In this paper, the FliC and FliD protein polypeptide epitopes were mapped in silico and by using PEPSCAN procedure. We identified four promising epitopes: 117QRMRTLS123, 205MSKAG209 of FliC and 226NKVAS230, 306TTKKPKD312 of FliD protein. We showed that 117QRMRTLS123 sequence is not only located in TLR5-binding and activating region, as previously shown, but forms an epitope recognized by C. difficile-infected patients' antibodies. 205MSKAG209 is a C. difficile-unique, immunogenic sequence that forms an exposed epitope on the polymerized flagella structure which makes it a suitable vaccine antigen. 226NKVAS230 and 306TTKKPKD312 are well exposed and possess potential protective properties according to VaxiJen analysis. Our results open the possibility to use these epitopes as suitable anti-C. difficile vaccine antigens.


Clostridioides difficile/immunology , Clostridium Infections/diagnosis , Flagella/immunology , Amino Acid Sequence/genetics , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Clostridioides/genetics , Clostridioides/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/immunology , Epitopes/immunology , Flagellin/immunology , Humans , Rabbits , Sequence Alignment/methods
12.
J Am Coll Surg ; 233(2): 223-231, 2021 08.
Article En | MEDLINE | ID: mdl-34015452

BACKGROUND: Immunosuppressant use increases risk of Clostridioides (Clostridium) difficile infection. To date, no studies have analyzed the relationship between immunosuppressant use and C difficile infections after metabolic and bariatric surgery (MBS). METHODS: A retrospective analysis of the 2015-2018 MBSAQIP data was conducted. The MBSAQIP data include information from 854 affiliated practices in the US and Canada. Initial sample size was 760,076 MBS patients. After excluding participants due to missing variables (n = 188,106) and the use of surgical procedures other than Roux-en-Y gastric bypass and sleeve gastroplasty (n = 129,712), final analyses were performed on 442,258 participants. Logistic regression models generated the odds of C difficile infection developing post MBS, according to immunosuppressant status (positive or negative). RESULTS: Unadjusted logistic regression analysis showed that patients using immunosuppressants were 95% more likely to have postoperative C difficile infection (odds ratio 1.945; 95% CI, 1.230 to 3.075; p < 0.001) vs MBS patients not taking immunosuppressants. After adjusting for age, sex, ethnicity, preoperative BMI, diabetes status, and surgical procedure type, the association remained unaffected (adjusted odds ratio 1.956; 95% CI, 1.236 to 3.095; p < 0.01). Patients who completed the laparoscopic Roux-en-Y gastric bypass procedure had more than double the odds of C difficile infection developing compared with those who completed the laparoscopic sleeve gastrectomy procedure (odds ratio 2.183; 95% CI, 1.842 to 2.587; p < 0.0001). CONCLUSIONS: Our results using a population-based sample of MBS patients showed that those taking immunosuppressants have a significantly higher risk of developing Clostridioides (Clostridium) difficile infection postoperatively. These findings suggest that patients using immunosuppressants should be closely monitored both pre and post procedure.


Clostridium Infections/epidemiology , Gastric Bypass/adverse effects , Gastroplasty/adverse effects , Immunosuppressive Agents/adverse effects , Postoperative Complications/epidemiology , Adult , Canada/epidemiology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Clostridium Infections/microbiology , Female , Gastric Bypass/statistics & numerical data , Gastroplasty/statistics & numerical data , Humans , Laparoscopy/adverse effects , Laparoscopy/statistics & numerical data , Male , Middle Aged , Obesity, Morbid/surgery , Postoperative Complications/immunology , Postoperative Complications/microbiology , Retrospective Studies , Risk Assessment/statistics & numerical data , Risk Factors , United States/epidemiology
13.
Gastroenterology ; 160(7): 2328-2339.e6, 2021 06.
Article En | MEDLINE | ID: mdl-33684427

BACKGROUND & AIMS: Although the role of gut microbiota in Clostridioides difficile infection (CDI) has been well established, little is known about the role of mycobiota in CDI. Here, we performed mycobiome data analysis in a well-characterized human cohort to evaluate the potential of using gut mycobiota features for CDI diagnosis. METHODS: Stool samples were collected from 118 hospital patients, divided into 3 groups: CDI (n = 58), asymptomatic carriers (Carrier, n = 28), and Control (n = 32). The nuclear ribosomal DNA internal transcribed spacer 2 was sequenced using the Illumina HiSeq platform to assess the fungal composition. Downstream statistical analyses (including Alpha diversity analysis, ordination analysis, differential abundance analysis, fungal correlation network analysis, and classification analysis) were then performed. RESULTS: Significant differences were observed in alpha and beta diversity between patients with CDI and Carrier (P < .05). Differential abundance analysis identified 2 genera (Cladosporium and Aspergillus) enriched in Carrier. The ratio of Ascomycota to Basidiomycota was dramatically higher in patients with CDI than in Carrier and Control (P < .05). Correlations between host immune factors and mycobiota features were weaker in patients with CDI than in Carrier. Using 4 fungal operational taxonomic units combined with 6 host immune markers in the random forest classifier can achieve very high performance (area under the curve ∼92.38%) in distinguishing patients with CDI from Carrier. CONCLUSIONS: Our study provides specific markers of stool fungi combined with host immune factors to distinguish patients with CDI from Carrier. It highlights the importance of gut mycobiome in CDI, which may have been underestimated. Further studies on the diagnostic applications and therapeutic potentials of these findings are warranted.


Carrier State/diagnosis , Clostridium Infections/diagnosis , Feces/microbiology , Immunologic Factors/analysis , Mycobiome/immunology , Carrier State/microbiology , Clostridioides difficile/immunology , Clostridium Infections/microbiology , Diagnosis, Differential , Female , Gastrointestinal Microbiome/immunology , Humans , Male , Middle Aged , Prospective Studies
14.
J Infect Dis ; 224(9): 1556-1565, 2021 11 16.
Article En | MEDLINE | ID: mdl-33780547

BACKGROUND: Inflammation is a crucial driver of host damage in patients with Clostridioides difficile colitis. We examined the potential for the intestinal microbiome to modify inflammation in patients with C. difficile colitis via the effects of gut-derived endotoxin on cytokine production. METHODS: Endotoxin from Escherichia coli and Pseudomonas aeruginosa as well as stool-derived endotoxin were tested for their ability to enhance interleukin 1ß (IL-1ß) and tumor necrosis factor alpha (TNF-α) production by toxin B-stimulated peripheral blood mononuclear cells. Inflammasome and Toll-like receptor 4 (TLR4) blocking studies were done to discern the importance of these pathways, while metagenomic studies were done to characterize predominant organisms from stool samples. RESULTS: Endotoxin significantly enhanced the ability of C. difficile toxin B to promote IL-1ß production but not TNF-α. The magnitude of this effect varied by endotoxin type and was dependent on combined inflammasome and TLR4 activation. Stool-derived endotoxin exhibited a similar synergistic effect on IL-1ß production with less synergy observed for stools that contained a high proportion of γ-proteobacteria. CONCLUSIONS: The ability of endotoxin to enhance IL-1ß production highlights a manner by which the microbiome can modify inflammation and severity of C. difficile disease. This information may be useful in devising new therapies for severe C. difficile colitis.


Bacterial Proteins/immunology , Bacterial Toxins/immunology , Clostridioides difficile/immunology , Endotoxins , Feces/microbiology , Gastrointestinal Microbiome , Inflammation Mediators/metabolism , Interleukin-1beta/blood , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Child , Child, Preschool , Clostridioides difficile/genetics , Colitis , Female , Humans , Inflammasomes/blood , Inflammation , Interleukin-1beta/metabolism , Leukocytes, Mononuclear , Male , Toll-Like Receptor 4/blood
15.
J Immunol ; 206(7): 1576-1585, 2021 04 01.
Article En | MEDLINE | ID: mdl-33597149

Short-chain fatty acids, such as butyrate, are major gut microbial metabolites that are beneficial for gastrointestinal health. Clostridium butyricum MIYAIRI588 (CBM588) is a bacterium that produces a robust amount of butyrate and therefore has been used as a live biotherapeutic probiotic in clinical settings. Clostridioides difficile causes life-threatening diarrhea and colitis. The gut resident microbiota plays a critical role in the prevention of C. difficile infection (CDI), as the disruption of the healthy microbiota by antibiotics greatly increases the risk for CDI. We report that CBM588 treatment in mice significantly improved clinical symptoms associated with CDI and increased the number of neutrophils and Th1 and Th17 cells in the colonic lamina propria in the early phase of CDI. The protective effect of CBM588 was abolished when neutrophils, IFN-γ, or IL-17A were depleted, suggesting that induction of the immune reactants is required to elicit the protective effect of the probiotic. The administration of tributyrin, which elevates the concentration of butyrate in the colon, also increased the number of neutrophils in the colonic lamina propria, indicating that butyrate is a potent booster of neutrophil activity during infection. However, GPR43 and GPR109a, two G protein-coupled receptors activated by butyrate, were dispensable for the protective effect of CBM588. These results indicate that CBM588 and butyrate suppress CDI, in part by boosting antimicrobial innate and cytokine-mediated immunity.


Clostridioides difficile/immunology , Clostridium Infections/immunology , Clostridium butyricum/physiology , Colon/immunology , Neutrophils/immunology , Receptors, G-Protein-Coupled/metabolism , Animals , Butyrates/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , alpha-Defensins/metabolism
16.
Nat Commun ; 12(1): 462, 2021 01 19.
Article En | MEDLINE | ID: mdl-33469019

Clostridioides difficile is a bacterial pathogen that causes a range of clinical disease from mild to moderate diarrhea, pseudomembranous colitis, and toxic megacolon. Typically, C. difficile infections (CDIs) occur after antibiotic treatment, which alters the gut microbiota, decreasing colonization resistance against C. difficile. Disease is mediated by two large toxins and the expression of their genes is induced upon nutrient depletion via the alternative sigma factor TcdR. Here, we use tcdR mutants in two strains of C. difficile and omics to investigate how toxin-induced inflammation alters C. difficile metabolism, tissue gene expression and the gut microbiota, and to determine how inflammation by the host may be beneficial to C. difficile. We show that C. difficile metabolism is significantly different in the face of inflammation, with changes in many carbohydrate and amino acid uptake and utilization pathways. Host gene expression signatures suggest that degradation of collagen and other components of the extracellular matrix by matrix metalloproteinases is a major source of peptides and amino acids that supports C. difficile growth in vivo. Lastly, the inflammation induced by C. difficile toxin activity alters the gut microbiota, excluding members from the genus Bacteroides that are able to utilize the same essential nutrients released from collagen degradation.


Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Clostridium Infections/immunology , Gastrointestinal Microbiome/immunology , Sigma Factor/metabolism , Animals , Anti-Bacterial Agents/adverse effects , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacteroides/drug effects , Bacteroides/metabolism , Clostridioides difficile/genetics , Clostridioides difficile/immunology , Clostridium Infections/microbiology , Clostridium Infections/pathology , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation, Bacterial/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Matrix Metalloproteinases/metabolism , Mice , Nutrients/metabolism , Proteolysis , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA-Seq , Sigma Factor/genetics , Sigma Factor/immunology , Transcriptome/immunology
17.
Front Immunol ; 12: 818734, 2021.
Article En | MEDLINE | ID: mdl-35095921

Adjuvant combinations may enhance or broaden the expression of immune responses to vaccine antigens. Information on whether established Alum type adjuvants can be combined with experimental CD1d ligand adjuvants is currently lacking. In this study, we used a murine Clostridioides difficile immunization and challenge model to evaluate Alum (Alhydrogel™), α-galactosylceramide (α-GC), and one of its analogs 7DW8-5 singly and in combination as vaccine adjuvants. We observed that the Alum/α-GC combination caused modest enhancement of vaccine antigen-specific IgG1 and IgG2b responses, and a broadening to include IgG2c that did not significantly impact overall protection. Similar observations were made using the Alum/7DW8-5 combination. Examination of the impact of adjuvants on NKT cells revealed expansion of invariant NKT (iNKT) cells with modest expansion of their iNKTfh subset and little effect on diverse NKT (dNKT) cells. Side effects of the adjuvants was determined and revealed transient hepatotoxicity when Alum/α-GC was used in combination but not singly. In summary these results showed that the Alum/α-GC or the Alum/7DW8-5 combination could exert distinct effects on the NKT cell compartment and on isotype switch to produce Th1-driven IgG subclasses in addition to Alum/Th2-driven subclasses. While Alum alone was efficacious in stimulating IgG-mediated protection, and α-GC offered no apparent additional benefit in the C. difficile challenge model, the work herein reveals immune response features that could be optimized and harnessed in other vaccine contexts.


Adjuvants, Vaccine , Alum Compounds , Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Clostridium Infections/prevention & control , Natural Killer T-Cells/immunology , Adjuvants, Vaccine/administration & dosage , Adjuvants, Vaccine/chemistry , Animals , Bacterial Vaccines/administration & dosage , Biomarkers , Disease Models, Animal , Dose-Response Relationship, Immunologic , Female , Immunization , Immunophenotyping , Ligands , Lymphocyte Activation/immunology , Mice , Natural Killer T-Cells/metabolism
18.
J Leukoc Biol ; 109(1): 195-210, 2021 01.
Article En | MEDLINE | ID: mdl-32829520

Clostridioides difficile (C. difficile) is the major cause of hospital-acquired gastrointestinal infections in individuals following antibiotics treatment. The pathogenesis of C. difficile infection (CDI) is mediated mainly by the production of toxins that induce tissue damage and host inflammatory responses. While innate immunity is well characterized in human and animal models of CDI, adaptive immune responses remain poorly understood. In this review, the current understanding of adaptive immunity is summarized and its influence on pathogenesis and disease outcome is discussed. The perspectives on what we believe to be the main pending questions and the focus of future research are also provided. There is no doubt that the innate immune response provides a first line of defense to CDI. But, is the adaptive immune response a friend or a foe? Probably it depends on the course of the disease. Adaptive immunity is essential for pathogen eradication, but may also trigger uncontrolled or pathological inflammation. Most of the understanding of the role of T cells is based on findings from experimental models. While they are a very valuable tool for research studies, more studies in human are needed to translate these findings into human disease. Another main challenge is to unravel the role of the different T cell populations on protection or induction of immunopathogenesis.


Adaptive Immunity/immunology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Animals , Humans
19.
J Immunol Methods ; 492: 112932, 2021 05.
Article En | MEDLINE | ID: mdl-33221459

BACKGROUND: Clostridioides difficile infections (CDI) have been a challenging and increasingly serious concern in recent years. While early and accurate diagnosis is crucial, available assays have frustrating limitations. OBJECTIVE: Develop a simple, blood-based immunoassay to accurately diagnose patients suffering from active CDI. MATERIALS AND METHODS: Uninfected controls (N = 95) and CDI patients (N = 167) were recruited from Atlanta area hospitals. Blood samples were collected from patients within twelve days of a positive CDI test and processed to yield serum and PBMCs cultured to yield medium enriched for newly synthesized antibodies (MENSA). Multiplex immunoassays measured Ig responses to ten recombinant C. difficile antigens. RESULTS: Sixty-six percent of CDI patients produced measurable responses to C. difficile antigens in their serum or MENSA within twelve days of a positive CDI test. Fifty-two of the 167 CDI patients (31%) were detectable in both serum and MENSA, but 32/167 (19%) were detectable only in MENSA, and 27/167 (16%) were detectable only in serum. DISCUSSION: We describe the results of a multiplex immunoassay for the diagnosis of ongoing CDI in hospitalized patients. Our assay resolved patients into four categories: MENSA-positive only, serum-positive only, MENSA- and serum-positive, and MENSA- and serum-negative. The 30% of patients who were MENSA-positive only may be accounted for by nascent antibody secretion prior to seroconversion. Conversely, the serum-positive only subset may have been more advanced in their disease course. Immunocompromise and misdiagnosis may have contributed to the 34% of CDI patients who were not identified using MENSA or serum immunoassays. IMPORTANCE: While there was considerable overlap between patients identified through MENSA and serum, each method detected a distinctive patient group. The combined use of both MENSA and serum to detect CDI patients resulted in the greatest identification of CDI patients. Together, longitudinal analysis of MENSA and serum will provide a more accurate evaluation of successful host humoral immune responses in CDI patients.


Antibodies, Bacterial/analysis , Clostridioides difficile/isolation & purification , Clostridium Infections/diagnosis , Serologic Tests/methods , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Case-Control Studies , Cell Culture Techniques , Clostridioides difficile/immunology , Clostridium Infections/blood , Clostridium Infections/microbiology , Culture Media/metabolism , Female , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
20.
J Immunol Methods ; 488: 112917, 2021 01.
Article En | MEDLINE | ID: mdl-33221460

Clostridioides difficile (C. difficile) is the most common cause of nosocomial antibiotic associated diarrhoea. The incidence of C. difficile infection (CDI) has been rising worldwide over the last 20 years with consequent rises in morbidity, mortality and healthcare costs, although the incidence has fallen in the UK over the last few years. Confirmation of diagnosis and early intervention are critical to the management of CDI. The standard treatment for CDI is the administration of antibiotics. However, vaccination has been recognized as the most cost-effective treatment for the prevention and possible long-term protection against CDI episode. There are several promising vaccine candidates in various stages of development. Many of these vaccines have displayed good efficacy for CDI under laboratory conditions or in clinical trials. With the emergence of vaccines against C. difficile, here we describe the development and verification of an Enzyme Linked Immunosorbent Assay (ELISA) that can be used for the quality control testing of candidate vaccines against C. difficile through the measurement of vaccine antigen content. Verification of the assay was performed by assessment of specificity, sensitivity, intermediate precision and relative accuracy. The ELISAs were specific for the toxoids being detected and the detection limit of the assay for toxoid A was 4.88 ng/mL and 3.91 ng/mL for toxoid B. The geometric coefficients of variation for intermediate precision did not exceed 25% and relative accuracy was within 77-130%. We therefore conclude that the ELISA described here is sufficiently sensitive, specific, precise and accurate for use for the quality control testing of candidate C. difficile vaccines.


Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bacterial Vaccines/metabolism , Clostridioides difficile/metabolism , Enterotoxins/metabolism , Enzyme-Linked Immunosorbent Assay , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Enterotoxins/immunology , Limit of Detection , Quality Control , Reproducibility of Results
...