Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Antibiotics (Basel) ; 11(5)2022 May 05.
Article in English | MEDLINE | ID: mdl-35625268

ABSTRACT

Clostridioides difficile infection (CDI) is the primary cause of health-care-associated infectious diarrhea. Treatment requires mostly specific antibiotics such as metronidazole (MTZ), vancomycin or fidaxomicin. However, approximately 20% of treated patients experience recurrences. Treatment with MTZ is complicated by reduced susceptibility to this molecule, which could result in high failure and recurrence rates. However, the mechanism remains unclear. In this study, we investigated the impact of subinhibitory concentrations of MTZ on morphology, motility, biofilm formation, bacterial adherence to the intestinal Caco-2/TC7 differentiated monolayers, and colonization in monoxenic and conventional mouse models of two C. difficile strains (VPI 10463 and CD17-146), showing different susceptibility profiles to MTZ. Our results revealed that in addition to the inhibition of motility and the downregulation of flagellar genes for both strains, sub-inhibitory concentrations of MTZ induced various in vitro phenotypes for the strain CD17-146 exhibiting a reduced susceptibility to this antibiotic: elongated morphology, enhanced biofilm production and increased adherence to Caco-2/TC7 cells. Weak doses of MTZ induced higher level of colonization in the conventional mouse model and a trend to thicker 3-D structures entrapping bacteria in monoxenic mouse model. Thus, sub-inhibitory concentrations of MTZ can have a wide range of physiological effects on bacteria, which may contribute to their persistence after treatment.

2.
J Clin Med ; 9(11)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202811

ABSTRACT

In a previous monocentric study in preterm neonates (PN), we described a high Clostridioides difficile colonization rate (74%) with two uncommon non-toxigenic strains (NTCD) belonging to PCR-ribotype (RT) (CE)847 and (CE)032. To determine the extent of carriage of both NTCD in other spatio-temporal settings, strains isolated in PN stools from two multicenter cohorts were characterized by PCR-ribotyping, MLVA and MLST. We also evaluated the protective role of two NTCD from these RT against C. difficile infection in a hamster caecitis model. Animals were administered either each NTCD alone (n = 7), or followed by a 027 strain (n = 9). A control group received only the 027 strain (n = 8). Clinical activity and colonization by C. difficile in stools were monitored daily until death or sacrifice at D20. We isolated 18 RT(CE)032 (ST-83) strains and 2 RT(CE)847 (ST-26) strains among 247 PN from both cohorts. Within each RT, strains were genetically related. The survival rate was significantly increased when animals received a RT(CE)847 or (CE)032 strain before the 027 strain (4/9 deaths, p = 0.029; 1/9 death, p = 0.0004, respectively). We describe two predominant uncommon NTCD strains, in a PN population from different healthcare facilities. Both NTCD provide a potential protection against C. difficile infection.

3.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31085703

ABSTRACT

Clostridium difficile is the leading cause of antibiotic-associated diarrhea in adults. During infection, C. difficile must detect the host environment and induce an appropriate survival strategy. Signal transduction networks involving serine/threonine kinases (STKs) play key roles in adaptation, as they regulate numerous physiological processes. PrkC of C. difficile is an STK with two PASTA domains. We showed that PrkC is membrane associated and is found at the septum. We observed that deletion of prkC affects cell morphology with an increase in mean size, cell length heterogeneity, and presence of abnormal septa. A ΔprkC mutant was able to sporulate and germinate but was less motile and formed more biofilm than the wild-type strain. Moreover, a ΔprkC mutant was more sensitive to antimicrobial compounds that target the cell envelope, such as the secondary bile salt deoxycholate, cephalosporins, cationic antimicrobial peptides, and lysozyme. This increased susceptibility was not associated with differences in peptidoglycan or polysaccharide II composition. However, the ΔprkC mutant had less peptidoglycan and released more polysaccharide II into the supernatant. A proteomic analysis showed that the majority of C. difficile proteins associated with the cell wall were less abundant in the ΔprkC mutant than the wild-type strain. Finally, in a hamster model of infection, the ΔprkC mutant had a colonization delay that did not significantly affect overall virulence.


Subject(s)
Bacterial Proteins/physiology , Clostridioides difficile/drug effects , Protein Serine-Threonine Kinases/physiology , Animals , Cell Wall/metabolism , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Cricetinae , Drug Resistance, Bacterial , Homeostasis , Mesocricetus , Microbial Sensitivity Tests , Peptidoglycan/metabolism , Protein Serine-Threonine Kinases/genetics , Virulence
4.
Front Microbiol ; 9: 2849, 2018.
Article in English | MEDLINE | ID: mdl-30524414

ABSTRACT

Introduction: Bile acids (BA) influence germination and growth of Clostridium difficile. Ursodeoxycholic acid (UDCA), a BA minor in human, used for cholestatic liver diseases, inhibits germination and growth of C. difficile in vitro, but was never tested in vivo with an infectious challenge versus control. We hypothesized that UDCA could prevent CDI. We evaluated the effects of UDCA on C. difficile in vitro and in hamsters, with pharmacokinetics study and with an infectious challenge. Then, we studied CDI incidence in UDCA-treated patients. Methods: We evaluated germination and growth of C. difficile, with 0.01, 0.05, and 0.1% UDCA. We analyzed fecal BA of hamsters receiving antibiotics and UDCA (50 mg/kg/day), antibiotics, or UDCA alone. Then, we challenged with spores of C. difficile at D6 hamsters treated with UDCA (50 mg/kg/day) from D1 to D13, versus control. In human, we analyzed the database of a cohort on CDI in acute flares of inflammatory bowel disease (IBD). As PSC-IBD patients were under UDCA treatment, we compared PSC-IBD patients to IBD patients without PSC. Results: In vitro, UDCA inhibited germination and growth of C. difficile at 0.05 and 0.1%, competing with 0.1% TCA (with 0.1%: 0.05% ± 0.05% colony forming unit versus 100% ± 0%, P < 0.0001). In hamsters, UDCA reached high levels only when administered with antibiotics (43.5% UDCA at D5). Without antibiotics, UDCA was in small amount in feces (max. 4.28%), probably because of UDCA transformation into LCA by gut microbiota. During infectious challenge, mortality was similar in animals treated or not with UDCA (62.5%, n = 5/8, P = 0.78). UDCA percentage was high, similar and with the same kinetics in dead and surviving hamsters. However, dead hamsters had a higher ratio of primary over secondary BA compared to surviving hamsters. 9% (n = 41/404) of IBD patients without PSC had a CDI, versus 25% (n = 4/12) of PSC-IBD patients treated with UDCA. Conclusion: We confirmed the inhibitory effect of UDCA on growth and germination of C. difficile in vitro, with 0.05 or 0.1% UDCA. However, in our hamster model, UDCA was inefficient to prevent CDI, despite high levels of UDCA in feces. Patients with PSC-IBD treated with UDCA did not have less CDI than IBD patients.

5.
PLoS One ; 12(11): e0187212, 2017.
Article in English | MEDLINE | ID: mdl-29176760

ABSTRACT

The immunogenicity of bacterial flagellin has been reported in different studies. By its close interaction with the immune system, the flagellin represents an interesting adjuvant and vaccine candidate. Salmonella Typhimurium flagellin has already been tested as adjuvant to stimulate mucosal immunity. Here, we assessed the ability of Clostridium difficile flagellin FliC to act as a mucosal adjuvant, first combined with ovalbumin as antigen and second with a C. difficile surface protein, the precursor of the S-layer proteins SlpA. Using ovalbumin as antigen, we compared the gut mucosal adjuvanticity of FliC to Salmonella Typhimurium flagellin and cholera toxin. Two routes of immunization were tested in a mouse model: intra-rectal and intra-peritoneal, following which, gut mucosal and systemic antibody responses against ovalbumin (Immunoglobulins G and Immunoglobulins A) were analyzed by Enzyme-Linked Immuno Assay in intestinal contents and in sera. In addition, ovalbumin-specific immunoglobulin producing cells were detected in the intestinal lamina propria by Enzyme-Linked Immunospot. Results showed that FliC as adjuvant for immunization targeting ovalbumin was able to stimulate a gut mucosal and systemic antibody response independently of the immunization route. In order to develop a mucosal vaccine to prevent C. difficile intestinal colonization, we assessed in a mouse model the efficacy of FliC as adjuvant compared with cholera toxin co-administrated with the C. difficile S-layer precursor SlpA as antigen. After challenge, a significant decrease of C. difficile intestinal colonization was observed in immunized groups compared to the control group. Our results showed that C. difficile FliC could be used as adjuvant in mucosal vaccination strategy against C. difficile infections.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Flagellin/metabolism , Immunity, Mucosal/drug effects , Animals , Antibodies, Bacterial/immunology , Cell Count , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Colony Count, Microbial , Enterocolitis, Pseudomembranous/blood , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/microbiology , Female , Immunity/drug effects , Immunization , Immunoglobulin G/blood , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Ovalbumin/immunology , Rectum/immunology , Vaccination
6.
Front Microbiol ; 8: 2086, 2017.
Article in English | MEDLINE | ID: mdl-29118745

ABSTRACT

Clostridium difficile infection (CDI) is a major healthcare-associated disease with high recurrence rates. Host colonization is critical for the infectious process, both in first episodes and in recurrent disease, with biofilm formation playing a key role. The ability of C. difficile to form a biofilm on abiotic surfaces is established, but has not yet been confirmed in the intestinal tract. Here, four different isolates of C. difficile, which are in vitro biofilm producers, were studied for their ability to colonize germ-free mice. The level of colonization achieved was similar for all isolates in the different parts of the murine gastrointestinal tract, but pathogen burden was higher in the cecum and colon. Confocal laser scanning microscopy revealed that C. difficile bacteria were distributed heterogeneously over the intestinal tissue, without contact with epithelial cells. The R20291 strain, which belongs to the Ribotype 027 lineage, displayed a unique behavior compared to the other strains by forming numerous aggregates. By immunochemistry analyses, we showed that bacteria were localized inside and outside the mucus layer, irrespective of the strains tested. Most bacteria were entrapped in 3-D structures overlaying the mucus layer. For the R20291 strain, the cell-wall associated polysaccharide PS-II was detected in large amounts in the 3-D structure. As this component has been detected in the extrapolymeric matrix of in vitro C. difficile biofilms, our data suggest strongly that at least the R20291 strain is organized in the mono-associated mouse model in glycan-rich biofilm architecture, which sustainably maintains bacteria outside the mucus layer.

7.
Environ Microbiol ; 19(5): 1933-1958, 2017 05.
Article in English | MEDLINE | ID: mdl-28198085

ABSTRACT

Clostridium difficile is a major cause of diarrhoea associated with antibiotherapy. Exposed to stresses in the gut, C. difficile can survive by inducing protection, detoxification and repair systems. In several firmicutes, most of these systems are controlled by the general stress response involving σB . In this work, we studied the role of σB in the physiopathology of C. difficile. We showed that the survival of the sigB mutant during the stationary phase was reduced. Using a transcriptome analysis, we showed that σB controls the expression of ∼25% of genes including genes involved in sporulation, metabolism, cell surface biogenesis and the management of stresses. By contrast, σB does not control toxin gene expression. In agreement with the up-regulation of sporulation genes, the sporulation efficiency is higher in the sigB mutant than in the wild-type strain. sigB inactivation also led to increased sensitivity to acidification, cationic antimicrobial peptides, nitric oxide and ROS. In addition, we showed for the first time that σB also plays a crucial role in oxygen tolerance in this strict anaerobe. Finally, we demonstrated that the fitness of colonisation by the sigB mutant is greatly affected in a dixenic mouse model of colonisation when compared to the wild-type strain.


Subject(s)
Bacterial Proteins/genetics , Clostridioides difficile/genetics , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Bacterial/genetics , Sigma Factor/genetics , Animals , Bacterial Proteins/metabolism , Clostridioides difficile/pathogenicity , DNA Repair/genetics , Diarrhea/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Female , Gene Expression Profiling , Germ-Free Life , Mice , Mice, Inbred C3H , Oxidative Stress/genetics , Sigma Factor/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/growth & development , Up-Regulation , Virulence Factors/genetics
8.
PLoS One ; 11(6): e0158204, 2016.
Article in English | MEDLINE | ID: mdl-27351947

ABSTRACT

Clostridium difficile is responsible for a wide spectrum of infection from asymptomatic carriage to severe, relapsing colitis. Since 2003, C. difficile infections have increased with a higher morbidity and mortality due to the emergence of epidemic and hypervirulent C. difficile strains such as those of the epidemic lineage 027/BI/NAP1. To decipher the hypervirulence and epidemicity of 027 strains, we analyzed gene expression profiles of the R20291 027 strain using a monoxenic mouse model during the first 38h of infection. A total of 741 genes were differentially expressed during the course of infection. They are mainly distributed in functional categories involved in host adaptation. Several genes of PTS and ABC transporters were significantly regulated during the infection, underlying the ability of strain R20291 to adapt its metabolism according to nutrient availability in the digestive tract. In this animal model, despite the early sporulation process, sporulation efficiency seems to indicate that growth of R20291 vegetative cells versus spores were favored during infection. The bacterial mechanisms associated to adaptability and flexibility within the gut environment, in addition to the virulence factor expression and antibiotic resistance, should contribute to the epidemicity and hypervirulence of the C. difficile 027 strains.


Subject(s)
Adaptation, Physiological , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/microbiology , Transcriptome , Animals , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Genes, Bacterial , Mice , Virulence/genetics
9.
PLoS One ; 9(5): e96876, 2014.
Article in English | MEDLINE | ID: mdl-24841151

ABSTRACT

Clostridium difficile is the main agent responsible for hospital acquired antibiotic associated diarrhoea. In recent years, epidemic strains have emerged causing more severe infections. Whilst C. difficile has two major virulence factors, toxins TcdA and TcdB, it is generally accepted that other virulence components of the bacterium contribute to disease. Previously, it has been suggested that flagella expression from pathogenic bacteria might be implicated in virulence. In a recent study, we observed an increased mortality in a gnotobiotic mouse model when animals were colonized with an isogenic fliC mutant constructed in the PCR-ribotype 027 (B1/NAP1) strain R20291, while animals survived when colonized by the parental strain or after colonization by other high-toxin-producing C. difficile strains. To understand the reasons for this increased virulence, we compared the global gene expression profiles between the fliC-R20291 mutant and its parental strain using an in vitro and in vivo transcriptomic approach. The latter made use of the gnotobiotic mouse model. Interestingly, in the fliC mutant, we observed considerable up-regulation of genes involved in mobility, membrane transport systems (PTS, ABC transporters), carbon metabolism, known virulence factors and sporulation. A smaller but significant up-regulation of genes involved in cell growth, fermentation, metabolism, stress and antibiotic resistance was also apparent. All of these genes may be associated with the increased virulence of the fliC-R20921 mutant. We confirmed that the fliC mutation is solely responsible for the observed changes in gene expression in the mutant strain since expression profiles were restored to that of the wild-type strain in the fliC-complemented strain. Thus, the absence of FliC is directly or indirectly involved in the high mortality observed in the fliC mutant infected animals. Therefore, we provide the first evidence that when the major structural component of the flagellum is neutralized, deregulation of gene expression can occur during infection.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Flagellin/metabolism , Animals , Bacterial Proteins/genetics , Clostridioides difficile/genetics , Enterocolitis, Pseudomembranous/microbiology , Flagellin/genetics , Gene Expression Regulation, Bacterial , Genetic Pleiotropy , Male , Mice , Virulence/genetics , Virulence/physiology , Virulence Factors/genetics , Virulence Factors/metabolism
10.
PLoS One ; 8(11): e81112, 2013.
Article in English | MEDLINE | ID: mdl-24303034

ABSTRACT

Clostridium difficile is a pathogen which is responsible for diarrhea and colitis, particularly after treatment with antibiotics. Clinical signs are mainly due to two toxins, TcdA and TcdB. However, the first step of pathogenesis is the colonization process. We evaluated C. difficile surface proteins as vaccine antigens in the hamster model to prevent intestinal colonization. This vaccination induced a partial protection of hamsters against death after a C. difficile challenge. A proteomic analysis of animal sera allowed us to identify proteins which could be responsible for the protection observed. Among these proteins, we identified the GroEL heat shock protein. To confirm the role of the specific GroEL antibodies in the delayed C. difficile colonization of hamsters, we performed an immunization assay in a mouse model. After intranasal immunization with the recombinant protein GroEL, we observed a lower C. difficile intestinal colonization in the immunized group as compared to the control group.


Subject(s)
Bacterial Proteins/immunology , Chaperonin 60/immunology , Clostridioides difficile/immunology , Intestines/immunology , Intestines/microbiology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibody Specificity/immunology , Bacterial Proteins/metabolism , Bacterial Vaccines/immunology , Cell Wall/immunology , Cell Wall/metabolism , Chaperonin 60/metabolism , Clostridioides difficile/metabolism , Cricetinae , Disease Models, Animal , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/mortality , Female , Immunization , Mice , Proteomics/methods , Recombinant Proteins/immunology
11.
Infect Immun ; 81(10): 3757-69, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897605

ABSTRACT

Clostridium difficile is currently the major cause of nosocomial intestinal diseases associated with antibiotic therapy in adults. In order to improve our knowledge of C. difficile-host interactions, we analyzed the genome-wide temporal expression of C. difficile 630 genes during the first 38 h of mouse colonization to identify genes whose expression is modulated in vivo, suggesting that they may play a role in facilitating the colonization process. In the ceca of the C. difficile-monoassociated mice, 549 genes of the C. difficile genome were differentially expressed compared to their expression during in vitro growth, and they were distributed in several functional categories. Overall, our results emphasize the roles of genes involved in host adaptation. Colonization results in a metabolic shift, with genes responsible for the fermentation as well as several other metabolic pathways being regulated inversely to those involved in carbon metabolism. In addition, several genes involved in stress responses, such as ferrous iron uptake or the response to oxidative stress, were regulated in vivo. Interestingly, many genes encoding conserved hypothetical proteins (CHP) were highly and specifically upregulated in vivo. Moreover, genes for all stages of sporulation were quickly induced in vivo, highlighting the observation that sporulation is central to the persistence of C. difficile in the gut and to its ability to spread in the environment. Finally, we inactivated two genes that were differentially expressed in vivo and evaluated the relative colonization fitness of the wild-type and mutant strains in coinfection experiments. We identified a CHP as a putative colonization factor, supporting the suggestion that the in vivo transcriptomic approach can unravel new C. difficile virulence genes.


Subject(s)
Adaptation, Physiological/genetics , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Gene Expression Regulation, Bacterial/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cecum/microbiology , Clostridioides difficile/physiology , Clostridium Infections/microbiology , Genome, Bacterial , Mice , Mutation , Peptidoglycan/genetics , Peptidoglycan/metabolism , Stress, Physiological , Up-Regulation , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
12.
Eur J Pharm Biopharm ; 79(3): 566-73, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21664462

ABSTRACT

We have designed an oral vaccine against Clostridium difficile infection. The virulent factor Cwp84, that is a cystein protease highly immunogenic in patients with C. difficile-associated disease, was entrapped within pectin beads. Beads encapsulating Cwp84 were shown to be stable in the simulated intestinal medium and to release the cystein protease once in the simulated colonic medium. Three groups of hamsters were immunized, the first receiving pectin beads encapsulating Cwp84, the second unloaded beads and the third one free Cwp84. After three immunizations by the intragastric route, all groups received clindamycine. Post-challenge survival with a strain of C. difficile showed that 2 days after infection, all hamsters treated with unloaded beads and all hamsters treated with free Cwp84 have deceased after 7 days, whereas about 40% of hamsters administered with Cwp84-loaded beads survived 10 days after challenge, proving that oral vaccination provides partial protection. These first data obtained with an oral vaccine against C. difficile appear promising for preventing this infection.


Subject(s)
Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Cysteine Endopeptidases/immunology , Drug Carriers/chemistry , Pectins/chemistry , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Chemistry, Pharmaceutical , Clostridioides difficile/enzymology , Clostridium Infections/immunology , Cricetinae , Cysteine Endopeptidases/administration & dosage , Cysteine Endopeptidases/chemistry , Disease Models, Animal , Drug Compounding , Drug Stability , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/blood , Molecular Weight , Survival Analysis
13.
FEMS Immunol Med Microbiol ; 63(1): 73-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21707776

ABSTRACT

Clostridium difficile is a pathogen responsible for diarrhoea and colitis, particularly after antibiotic treatment. We evaluated the C. difficile protease Cwp84, found to be associated with the S-layer proteins, as a vaccine antigen to limit the C. difficile intestinal colonization and therefore the development of the infection in a clindamycin-treated hamster model. First, we evaluated the immune response and the animal protection against death induced by several immunization routes: rectal, intragastric and subcutaneous. Antibody production was variable according to the immunization routes. In addition, serum Cwp84 antibody titres did not always correlate with animal protection after challenge with a toxigenic C. difficile strain. The best survival rate was observed with the rectal route of immunization. Then, in a second assay, we selected this immunization route to perform a larger immunization assay including a Cwp84 immunized group and a control group. Clostridium difficile intestinal colonization and survival rate, as well as the immune response were examined. Clostridium difficile hamster challenge resulted in a 26% weaker and slower C. difficile intestinal colonization in the immunized group. Furthermore, hamster survival in the Cwp84 immunized group was 33% greater than that of the control group, with a significant statistical difference.


Subject(s)
Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Cysteine Endopeptidases/immunology , Immunization/methods , Administration, Rectal , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Clostridioides difficile/enzymology , Clostridium Infections/immunology , Cricetinae , Disease Models, Animal , Gastric Lavage , Gastrointestinal Tract/microbiology , Injections, Subcutaneous , Male , Survival Analysis
14.
J Med Microbiol ; 60(Pt 8): 1155-1161, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21349990

ABSTRACT

Clostridium difficile is a frequent cause of severe, recurrent, post-antibiotic diarrhoea and pseudomembranous colitis. Its pathogenicity is mediated mainly by two toxins, TcdA and TcdB. However, different adhesins have also been described as important colonization factors which are implicated in the first step of the intestinal infection. In this study, we focused our interest on one of these adhesins, fibronectin-binding protein A (FbpA), and on its role in the intestinal colonization process. A mutant of FbpA (CDΔFbpA) was constructed in C. difficile strain 630Δerm by using ClosTron technology. This mutant was characterized in vitro and in vivo and compared to the isogenic wild-type strain. Adhesion of the CDΔFbpA mutant to the human colonic epithelial cell line Caco-2 and to mucus-secreting HT29-MTX cells was examined. Surprisingly, the CDΔFbpA mutant adhered more than the wild-type parental strain. The CDΔFbpA mutant was also analysed in three different mouse models by following the intestinal implantation kinetics (faecal shedding) and caecal colonization (7 days post-challenge). We showed that in monoxenic mice, CDΔFbpA shed C. difficile in faeces at the same rate as that of the isogenic wild-type strain but its colonization of the caecal wall was significantly reduced. In dixenic mice, the shedding rate was slower for the CDΔFbpA mutant than for the isogenic wild-type strain during the first days of infection, but no significant difference was observed in caecal colonization. Similar rates of intestinal implantation and caecal colonization were observed for both strains in assays performed in human microbiota-associated mice. Taken together, our data suggest that FbpA plays a role in intestinal colonization by C. difficile.


Subject(s)
Adhesins, Bacterial/metabolism , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial/physiology , Adhesins, Bacterial/genetics , Animals , Bacterial Adhesion , Caco-2 Cells , Clostridioides difficile/genetics , Clostridioides difficile/physiology , Feces/microbiology , HT29 Cells , Humans , Mice , Mutation
15.
Vaccine ; 25(20): 3946-54, 2007 May 16.
Article in English | MEDLINE | ID: mdl-17433506

ABSTRACT

Clostridium difficile pathogenesis is mainly due to toxins A and B. However, the first step of pathogenesis is the colonization process. We evaluated C. difficile surface proteins as vaccine antigens to diminish intestinal colonization in a human flora-associated mouse model. First, we used the flagellar cap protein FliD of C. difficile, in order to test several immunization routes: intranasal, rectal, and intragastric. The rectal route, which is the most efficient, was used to vaccine groups of mice with different antigen combinations. After immunizations, the mice were challenged with the toxigenic C. difficile and a significant statistical difference between the control group and the immunized groups was observed in the colonization levels of C. difficile.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/administration & dosage , Clostridioides difficile/growth & development , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/microbiology , Intestines/microbiology , Administration, Rectal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Cholera Toxin/immunology , Cholera Toxin/pharmacology , Clostridioides difficile/immunology , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/immunology , Female , Humans , Immunity, Mucosal/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Intestines/immunology , Mice , Mice, Inbred C3H
SELECTION OF CITATIONS
SEARCH DETAIL
...