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.
Cell Microbiol ; 19(6)2017 06.
Article in English | MEDLINE | ID: mdl-28382773

ABSTRACT

CD4+ CD25+ FoxP3+ regulatory T cells (Tregs) are key players for maintaining immune tolerance and for reducing the inflammation-mediated tissue damage following infection. However, Tregs also suppress protective immune responses to pathogens (including virus, bacteria, parasites, and fungi) and vaccines and enhance pathogen persistence by inhibiting the activation and functions of both innate and adaptive immune cells such as dendritic cells, macrophages, and T and B lymphocytes and by promoting immunosuppressive environment. Therefore, equilibrium in the Treg number and function is important to ensure pathogen clearance and protection from infection-associated immunopathologies. Recent advances in understanding of Treg influence on the outcome of infection opened new avenues to target them. Various small molecules, pharmacological inhibitors, monoclonal antibodies that target Tregs provided proof of concept in experimental models. The field also benefits from advances in other subjects, particularly oncology and autoimmunity, where Treg-targeted therapies are exploited in the clinic to a greater extent. The future research should aim at translating this preclinical success to human application.


Subject(s)
Bacterial Infections/immunology , Immune Tolerance/immunology , Mycoses/immunology , Parasitic Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Virus Diseases/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Humans , Macrophages/immunology
2.
Helicobacter ; 22(3)2017 Jun.
Article in English | MEDLINE | ID: mdl-28124467

ABSTRACT

BACKGROUND: Helicobacter (H.) suis is mainly associated with pigs, but is also the most prevalent gastric non-H. pylori Helicobacter species found in humans. Both H. pylori and H. suis may cause persistent infection of the stomach. Several immune evasion mechanisms have been proposed for H. pylori, which focus to a great extent on its major virulence factors, which are absent in H. suis. The aim of this study was to gain more knowledge on immune evasion by H. suis. MATERIALS AND METHODS: Cytokine expression kinetics were monitored in the stomach of BALB/c mice experimentally infected with H. suis. The cytokine expression profile in the stomach of naturally H. suis-infected pigs was also determined. Subsequently, the effect of H. suis on murine and porcine dendritic cell (DC) maturation and their ability to elicit T-cell effector responses was analyzed. RESULTS: Despite a Th17/Th2 response in the murine stomach, the inflammatory cell influx was unable to clear H. suis infection. H. suis-stimulated murine bone marrow-derived dendritic cells induced IL-17 secretion by CD4+ cells in vitro. Natural H. suis infection in pigs evoked increased expression levels of IL-17 mRNA in the antrum and IL-10 mRNA in the fundus. In contrast to mice, H. suis-stimulated porcine monocyte-derived dendritic cells were unable to express MHCII molecules on their cell surface. These semimature DCs induced proliferation of T-cells, which showed an increased expression of TGF-ß and FoxP3 mRNA levels. CONCLUSIONS: Helicobacter suis might evade host immune responses by skewing toward a Treg-biased response.


Subject(s)
Helicobacter Infections/veterinary , Helicobacter heilmannii/immunology , Host-Pathogen Interactions , Immune Evasion , Animals , Cytokines/metabolism , Female , Gene Expression Profiling , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Mice, Inbred BALB C , Stomach/immunology , Stomach/microbiology , Swine , T-Lymphocytes/immunology
3.
Methods Mol Biol ; 1494: 107-125, 2017.
Article in English | MEDLINE | ID: mdl-27718189

ABSTRACT

Adjuvants are substances that boost the protective immune response to vaccine antigens. The majority of known adjuvants have been identified through the use of empirical approaches. Our aim was to identify novel adjuvants with well-defined cellular and molecular mechanisms by combining a knowledge of immunoregulatory mechanisms with an in silico approach. CD4+CD25+FoxP3+ regulatory T cells (Tregs) inhibit the protective immune responses to vaccines by suppressing the activation of antigen presenting cells such as dendritic cells (DCs). In this chapter, we describe the identification and functional validation of small molecule antagonists to CCR4, a chemokine receptor expressed on Tregs. The CCR4 binds the chemokines CCL22 and CCL17 that are produced in large amounts by activated innate cells including DCs. In silico identified small molecule CCR4 antagonists inhibited the migration of Tregs both in vitro and in vivo and when combined with vaccine antigens, significantly enhanced protective immune responses in experimental models.


Subject(s)
Adjuvants, Immunologic , Computer Simulation , Drug Design , Models, Immunological , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Chemokine CCL17/immunology , Chemokine CCL22/immunology , Female , Humans , Mice , Receptors, CCR4/immunology
4.
Helicobacter ; 22(2)2017 Apr.
Article in English | MEDLINE | ID: mdl-27558281

ABSTRACT

BACKGROUND: Helicobacter suis (H. suis) is the most prevalent gastric non-H. pylori Helicobacter species in humans. This bacterium mainly colonizes the stomach of pigs, but it has also been detected in the stomach of nonhuman primates. The aim of this study was to obtain better insights into potential differences between pig- and primate-associated H. suis strains in virulence and pathogenesis. MATERIALS AND METHODS: In vitro-isolated H. suis strains obtained from pigs, cynomolgus monkeys (Macaca fascicularis), and rhesus monkeys (Macaca mulatta) were used for intragastric inoculation of BALB/c mice and Mongolian gerbils. Nine weeks and six months later, samples of the stomach of inoculated and control animals were taken for PCR analysis and histopathological examination. RESULTS: The cynomolgus monkey-associated H. suis strain only colonized the stomach of mice, but not of Mongolian gerbils. All other H. suis strains colonized the stomach in both rodent models. In all colonized animals, severe gastric inflammation was induced. Gastric lymphoid follicles and destruction of the antral epithelium were observed in infected gerbils, but not in mice. Infection with both pig- and primate-associated H. suis strains evoked a similar marked Th17 response in mice and gerbils, accompanied by increased CXCL-13 expression levels. CONCLUSIONS: Apart from the cynomolgus monkey-associated strain which was unable of colonizing the stomach of Mongolian gerbils, no substantial differences in virulence were found in rodent models between in vitro-cultured pig-associated, cynomolgus monkey-associated and rhesus monkey-associated H. suis strains. The experimental host determines the outcome of the immune response against H. suis infection, rather than the original host.


Subject(s)
Gastritis/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter heilmannii/isolation & purification , Helicobacter heilmannii/pathogenicity , Animals , Chemokine CXCL13/analysis , Disease Models, Animal , Gastric Mucosa/pathology , Gene Expression Profiling , Gerbillinae , Histocytochemistry , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Mice, Inbred BALB C , Polymerase Chain Reaction , Swine/microbiology , Th17 Cells/immunology , Virulence
5.
PLoS One ; 10(6): e0131364, 2015.
Article in English | MEDLINE | ID: mdl-26115373

ABSTRACT

Helicobacter suis (H. suis) is a widespread porcine gastric pathogen, which is also of zoonotic importance. The first goal of this study was to investigate the efficacy of several vaccine adjuvants (CpG-DNA, Curdlan, Freund's Complete and Incomplete, Cholera toxin), administered either subcutaneously or intranasally along with H. suis whole-cell lysate, to protect against subsequent H. suis challenge in a BALB/c infection model. Subcutaneous immunization with Freund's complete (FC)/lysate and intranasal immunization with Cholera toxin (CT)/lysate were shown to be the best options for vaccination against H. suis, as determined by the amount of colonizing H. suis bacteria in the stomach, although adverse effects such as post-immunization gastritis/pseudo-pyloric metaplasia and increased mortality were observed, respectively. Therefore, we decided to test alternative strategies, including sublingual vaccine administration, to reduce the unwanted side-effects. A CCR4 antagonist that transiently inhibits the migration of regulatory T cells was also included as a new adjuvant in this second study. Results confirmed that immunization with CT (intranasally or sublingually) is among the most effective vaccination protocols, but increased mortality was still observed. In the groups immunized subcutaneously with FC/lysate and CCR4 antagonist/lysate, a significant protection was observed. Compared to the FC/lysate immunized group, gastric pseudo-pyloric metaplasia was less severe or even absent in the CCR4 antagonist/lysate immunized group. In general, an inverse correlation was observed between IFN-γ, IL-4, IL-17, KC, MIP-2 and LIX mRNA expression and H. suis colonization density, whereas lower IL-10 expression levels were observed in partially protected animals.


Subject(s)
Adjuvants, Immunologic , Bacterial Vaccines , Helicobacter Infections , Helicobacter heilmannii/immunology , Receptors, CCR4/antagonists & inhibitors , Vaccination , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/pharmacology , Animals , Bacterial Vaccines/adverse effects , Bacterial Vaccines/immunology , Bacterial Vaccines/pharmacology , Cell Movement/drug effects , Cell Movement/immunology , Cytokines/immunology , Female , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter Infections/prevention & control , Mice , Mice, Inbred BALB C , Receptors, CCR4/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
6.
Vet Res ; 46: 31, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25889172

ABSTRACT

Helicobacter (H.) suis can colonize the stomach of pigs as well as humans, causing chronic gastritis and other gastric pathological changes including gastric ulceration and mucosa-associated lymphoid tissue (MALT) lymphoma. Recently, a virulence factor of H. suis, γ-glutamyl transpeptidase (GGT), has been demonstrated to play an important role in the induction of human gastric epithelial cell death and modulation of lymphocyte proliferation depending on glutamine and glutathione catabolism. In the present study, the relevance of GGT in the pathogenesis of H. suis infection was studied in mouse and Mongolian gerbil models. In addition, the relative importance of H. suis GGT was compared with that of the H. pylori GGT. A significant and different contribution of the GGT of H. suis and H. pylori was seen in terms of bacterial colonization, inflammation and the evoked immune response. In contrast to H. pyloriΔggt strains, H. suisΔggt strains were capable of colonizing the stomach at levels comparable to WT strains, although they induced significantly less overall gastric inflammation in mice. This was characterized by lower numbers of T and B cells, and a lower level of epithelial cell proliferation. In general, compared to WT strain infection, ggt mutant strains of H. suis triggered lower levels of Th1 and Th17 signature cytokine expression. A pronounced upregulation of B-lymphocyte chemoattractant CXCL13 was observed, both in animals infected with WT and ggt mutant strains of H. suis. Interestingly, H. suis GGT was shown to affect the glutamine metabolism of gastric epithelium through downregulation of the glutamine transporter ASCT2.


Subject(s)
Bacterial Proteins/genetics , Helicobacter Infections/veterinary , Helicobacter heilmannii/physiology , Helicobacter pylori/physiology , Virulence Factors/genetics , gamma-Glutamyltransferase/genetics , Animals , Bacterial Proteins/metabolism , Female , Gerbillinae , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Mice , Mice, Inbred BALB C , Swine , Virulence Factors/metabolism , gamma-Glutamyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...