Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 13(5): e0196950, 2018.
Article in English | MEDLINE | ID: mdl-29734365

ABSTRACT

Safe and efficacious orally-delivered mucosal vaccine platforms are desperately needed to combat the plethora of mucosally transmitted pathogens. Lactobacillus spp. have emerged as attractive candidates to meet this need and are known to activate the host innate immune response in a species- and strain-specific manner. For selected bacterial isolates and mutants, we investigated the role of key innate immune pathways required for induction of innate and subsequent adaptive immune responses. Co-culture of murine macrophages with L. gasseri (strain NCK1785), L. acidophilus (strain NCFM), or NCFM-derived mutants-NCK2025 and NCK2031-elicited an M2b-like phenotype associated with TH2 skewing and immune regulatory function. For NCFM, this M2b phenotype was dependent on expression of lipoteichoic acid and S layer proteins. Through the use of macrophage genetic knockouts, we identified Toll-like receptor 2 (TLR2), the cytosolic nucleotide-binding oligomerization domain containing 2 (NOD2) receptor, and the inflammasome-associated caspase-1 as contributors to macrophage activation, with NOD2 cooperating with caspase-1 to induce inflammasome derived interleukin (IL)-1ß in a pyroptosis-independent fashion. Finally, utilizing an NCFM-based mucosal vaccine platform with surface expression of human immunodeficiency virus type 1 (HIV-1) Gag or membrane proximal external region (MPER), we demonstrated that NOD2 signaling is required for antigen-specific mucosal and systemic humoral responses. We show that lactobacilli differentially utilize innate immune pathways and highlight NOD2 as a key mediator of macrophage function and antigen-specific humoral responses to a Lactobacillus acidophilus mucosal vaccine platform.


Subject(s)
Immunity, Humoral/genetics , Macrophages/immunology , Nod2 Signaling Adaptor Protein/genetics , Vaccines/administration & dosage , Administration, Oral , Animals , Antigens/administration & dosage , Caspase 1/genetics , Caspase 1/immunology , Genes, gag/genetics , HIV-1/immunology , HIV-1/pathogenicity , Humans , Immunity, Humoral/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lactobacillus acidophilus/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Macrophages/microbiology , Mice , Nod2 Signaling Adaptor Protein/immunology , Teichoic Acids/immunology , Teichoic Acids/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Vaccines/immunology
2.
Appl Environ Microbiol ; 77(18): 6587-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21784918

ABSTRACT

Display of heterologous antigens on the cell surface is considered a useful technique for vaccine delivery by recombinant lactobacilli. In this study, two recombinant Lactobacillus acidophilus derivatives displaying Salmonella flagellin (FliC) were constructed using different anchor motifs. In one instance, the FliC protein was fused to the C-terminal region of a cell envelope proteinase (PrtP) and was bound to the cell wall by electrostatic bonds. In the other case, the same antigen was conjugated to the anchor region of mucus binding protein (Mub) and was covalently associated with the cell wall by an LPXTG motif. These two recombinant L. acidophilus cell surface displays resulted in dissimilar maturation and cytokine production by human myeloid dendritic cells. The surface-associated antigen was highly sensitive to simulated gastric and small intestinal juices. By supplementation with bicarbonate buffer and soybean trypsin inhibitor, the cell surface antigen was protected from proteolytic enzymes during gastric challenge in vitro. The protective reagents also increased the viability of the L. acidophilus cells upon challenge with simulated digestive juices. These results demonstrate the importance of protecting cells and their surface-associated antigens during oral immunization.


Subject(s)
Antigens, Bacterial/metabolism , Dendritic Cells/immunology , Flagellin/metabolism , Lactobacillus acidophilus/metabolism , Salmonella Vaccines/immunology , Acids/metabolism , Antigens, Bacterial/genetics , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/microbiology , Drug Carriers , Drug Stability , Flagellin/genetics , Genetic Vectors , Humans , Lactobacillus acidophilus/genetics , Peptide Hydrolases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salmonella Vaccines/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
3.
Methods Mol Biol ; 302: 167-78, 2005.
Article in English | MEDLINE | ID: mdl-15937351

ABSTRACT

The enzyme-linked immunospot (ELISPOT) assay is a sensitive and relatively simple assay for detecting secreted cellular products such as cytokines and has become an invaluable immunological tool. The ELISPOT has been used extensively in human and murine research but has only recently been used to assess the feline immune system. For researchers studying feline disease or using the cat as a model of human disease, the quantification of cytokine-producing cells by ELISPOT is an invaluable technique for investigations of disease immunopathogenesis and vaccine efficacy. For example, use of the interferon (IFN)-gamma ELISPOT to measure the frequency of antigen-specific T-cells during feline immunodeficiency virus (FIV) infection or after immunization with candidate FIV vaccines is of particular interest. This application of the ELISPOT may serve to expand the utility of FIV as a model for human immunodeficiency virus. Broader applications of the ELISPOT should further our understanding of feline diseases and be useful in the rational development of more efficacious vaccines and therapeutic modalities for the enhancement of feline health. This chapter discusses important parameters of ELISPOT design that will enable researchers to develop and analyze the feline-specific assays within their own laboratory.


Subject(s)
Enzyme-Linked Immunosorbent Assay/veterinary , Animals , Antibodies , Antigens, Viral/administration & dosage , Cats , Cytokines/analysis , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay/methods , Feline Acquired Immunodeficiency Syndrome/immunology , Immunodeficiency Virus, Feline/immunology , T-Lymphocytes/immunology , Viral Vaccines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...