Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Infect Immun ; 89(11): e0043821, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34424751

ABSTRACT

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.


Subject(s)
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
2.
Infect Immun ; 89(10): e0027421, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34097471

ABSTRACT

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.


Subject(s)
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
3.
Front Immunol ; 12: 818734, 2021.
Article in English | MEDLINE | ID: mdl-35095921

ABSTRACT

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.


Subject(s)
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
4.
Front Immunol ; 11: 565648, 2020.
Article in English | MEDLINE | ID: mdl-33042146

ABSTRACT

Bacterial enteric pathogens individually and collectively represent a serious global health burden. Humoral immune responses following natural or experimentally-induced infections are broadly appreciated to contribute to pathogen clearance and prevention of disease recurrence. Herein, we have compared observations on humoral immune mechanisms following infection with Citrobacter rodentium, the model for enteropathogenic Escherichia coli, Vibrio cholerae, Shigella species, Salmonella enterica species, and Clostridioides difficile. A comparison of what is known about the humoral immune responses to these pathogens reveals considerable variance in specific features of humoral immunity including establishment of high affinity, IgG class-switched memory B cell and long-lived plasma cell compartments. This article suggests that such variance could be contributory to persistent and recurrent disease.


Subject(s)
Bacterial Infections/immunology , Intestinal Diseases/immunology , Animals , Bacteria , Humans , Immunity, Humoral
5.
Infect Immun ; 88(3)2020 02 20.
Article in English | MEDLINE | ID: mdl-31871095

ABSTRACT

The intracellularly active bacterial toxin TcdB is a major Clostridioides difficile virulence factor that contributes to inflammation and tissue damage during disease. Immunization with an inactive TcdB fragment prevents C. difficile infection (CDI)-associated pathology. The protective immune response against inactive TcdB involves development of antigen-specific memory B cells and long-lived plasma cells that encode TcdB-neutralizing antibodies. Unlike the response to inactive TcdB, very little is known about the host humoral immune response to C. difficile and TcdB during primary and recurrent infection. Here, we used a murine model of C. difficile disease recurrence to demonstrate that an initial infection induced a serum IgM and mucosal IgA response against the toxin, but a low serum IgG response, which is associated with a lack of protection against disease during reinfection. Infection induced a partial expansion of the T follicular helper cell compartment, essential for B cell memory responses, and, consistent with that, failed to significantly expand the memory B cell compartment. Further, infection failed to stimulate the memory B cell compartment in preimmunized mice, although they were protected against associated disease. These results delineate the key humoral immune events that follow primary and recurrent C. difficile infection and provide a compelling inverse correlation between B cell memory and disease recurrence.


Subject(s)
B-Lymphocytes/immunology , Clostridioides difficile/immunology , Clostridium Infections/immunology , Immunization , Immunoglobulin G/blood , T-Lymphocytes, Helper-Inducer/immunology , Animals , Bacterial Toxins/immunology , Clostridium Infections/microbiology , Immunoglobulin A/metabolism , Mice , Mucous Membrane/metabolism
6.
Immunohorizons ; 3(3): 88-93, 2019 03.
Article in English | MEDLINE | ID: mdl-31342012

ABSTRACT

The CD1d-binding glycolipid α-galactosylceramide (α-GC) is a potent adjuvant that activates NKT cells and in turn enhances T-dependent humoral immunity. Very little is known about how NKT cells and the NKT follicular helper (NKTfh) subset influence the immune response to T-independent polysaccharides. In this study, we used a Cre-Lox approach to generate mice devoid of the Bcl6 master transcription factor in CD4 lineage cells and thus devoid of NKTfh cells but not total NKT cells. It was observed that α-GC-driven IgG1 class switch against a polysaccharide Ag was dependent on the NKTfh subset. However, α-GC was unable to stimulate a polysaccharide-specific Ab recall response. It was observed that NKT-derived IL-21 was able to exert limited influence on the IgG1 response and was therefore likely to work in concert with other factors. This work shows that α-GC-driven NKTfh cells can direct polysaccharide-specific B cell responses by promoting IgG1 class switch but do not provide signals needed for generation of polysaccharide-specific B cell memory.


Subject(s)
Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Immunologic Memory , Natural Killer T-Cells/immunology , Polysaccharides/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Communication , Female , Galactosylceramides/immunology , Immunization , Immunoglobulin Class Switching/genetics , Immunoglobulin G/genetics , Lymphocyte Activation , Male , Mice , Mice, Transgenic , Natural Killer T-Cells/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL