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.
J Immunol ; 191(4): 1927-34, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851695

ABSTRACT

Helminthic infections protect mice from colitis in murine models of inflammatory bowel disease and also may protect people. Helminths like Heligmosomoides polygyrus bakeri can induce regulatory T cells (Treg). Experiments explored whether H. polygyrus bakeri infection could protect mice from colitis through activation of colonic Treg and examined mechanisms of action. We showed that H. polygyrus bakeri infection increased the number of T cells expressing Foxp3 in the colon. More importantly, Foxp3(+)/IL-10(-) and Foxp3(+)/IL-10(+) T cell subsets isolated from the colon of H. polygyrus bakeri-infected mice prevented colitis when adoptively transferred into a murine model of inflammatory bowel disease, whereas Treg from uninfected mice could not provide protection. Only the transferred colonic Foxp3(+)/IL-10(-) T cells from H. polygyrus bakeri-infected mice readily accumulated in the colon and mesenteric lymph nodes of recipient mice, and they reconstituted the Foxp3(+)/IL-10(-) and Foxp3(+)/IL-10(+) T cell subsets. However, transferred Foxp3(+)/IL-10(+) T cells disappeared. IL-10 expression by Foxp3(+) T cells was necessary for colitis prevention. Thus, H. polygyrus bakeri infection activates colonic Foxp3(+) T cells, making them highly regulatory. The Foxp3(+) T cells that fail to express IL-10 may be critical for populating the colon with the Foxp3(+)/IL-10(+) T cells, which are required to control colitis.


Subject(s)
Colitis/prevention & control , Colon/immunology , Intestinal Diseases, Parasitic/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Therapy with Helminths , Animals , Colitis/immunology , Colitis/parasitology , Colon/parasitology , Cytokines/biosynthesis , Cytokines/metabolism , DNA-Binding Proteins/deficiency , Disease Models, Animal , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/deficiency , Genes, Reporter , Graft Survival , Helminthiasis, Animal/immunology , Immunotherapy, Adoptive , Inflammatory Bowel Diseases/therapy , Interleukin-10/analysis , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/chemistry , T-Lymphocytes, Regulatory/transplantation
2.
J Immunol ; 189(5): 2512-20, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844110

ABSTRACT

Immunological diseases such as inflammatory bowel disease (IBD) are infrequent in less developed countries, possibly because helminths provide protection by modulating host immunity. In IBD murine models, the helminth Heligmosomoides polygyrus bakeri prevents colitis. It was determined whether H. polygyrus bakeri mediated IBD protection by altering dendritic cell (DC) function. We used a Rag IBD model where animals were reconstituted with IL10⁻/⁻ T cells, making them susceptible to IBD and with OVA Ag-responsive OT2 T cells, allowing study of a gut antigenic response. Intestinal DC from H. polygyrus bakeri-infected Rag mice added to lamina propria mononuclear cells (LPMC) isolated from colitic animals blocked OVA IFN-γ/IL-17 responses in vitro through direct contact with the inflammatory LPMC. DC from uninfected Rag mice displayed no regulatory activity. Transfer of DC from H. polygyrus bakeri-infected mice into Rag mice reconstituted with IL10⁻/⁻ T cells protected animals from IBD, and LPMC from these mice lost OVA responsiveness. After DC transfer, OT2 T cells populated the intestines normally. However, the OT2 T cells were rendered Ag nonresponsive through regulatory action of LPMC non-T cells. The process of regulation appeared to be regulatory T cell independent. Thus, H. polygyrus bakeri modulates intestinal DC function, rendering them tolerogenic. This appears to be an important mechanism through which H. polygyrus bakeri suppresses colitis. IFN-γ and IL-17 are colitogenic. The capacity of these DC to block a gut Ag-specific IFN-γ/IL-17 T cell response also is significant.


Subject(s)
Colitis/immunology , Dendritic Cells/immunology , Immune Tolerance , Inflammatory Bowel Diseases/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Cells, Cultured , Colitis/parasitology , Colitis/prevention & control , Dendritic Cells/parasitology , Dendritic Cells/pathology , Disease Models, Animal , Enterocolitis/immunology , Enterocolitis/parasitology , Enterocolitis/prevention & control , Epitopes, T-Lymphocyte/immunology , Inflammatory Bowel Diseases/parasitology , Inflammatory Bowel Diseases/prevention & control , Interleukin-10/administration & dosage , Interleukin-10/deficiency , Interleukin-10/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nematospiroides dubius/immunology , Strongylida Infections/pathology , Strongylida Infections/prevention & control , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/pathology
3.
Inflamm Bowel Dis ; 18(8): 1447-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22223533

ABSTRACT

BACKGROUND: Developing countries have a low incidence of inflammatory bowel disease (IBD), perhaps prevented by the high prevalence of helminth infections and other alterations in intestinal flora and fauna. Helminth infections prevent colitis in various murine models of IBD. IBD may be driven by an aberrant immune response to luminal antigen(s). METHODS: We developed a murine model of IBD in which gut injury was induced by a specific antigen to better simulate the IBD disease process and to determine if helminth infections could abolish gut injury induced by an orally administered antigen. The model features pan-enterocolitis triggered by feeding ovalbumin (OVA). RESULTS: The intestinal inflammation is antigen-specific and generates interleukin (IL)-17 and interferon-gamma (IFN-γ), but not IL-4. Full expression of the disease required T cells with defective capacity to make IL-10 and treatment with a noninjurious, low dose of a nonsteroidal antiinflammatory drug. Exposure to Heligmosomoides polygyrus abrogated this antigen-induced gut injury. H. polygyrus colonization induced Foxp3(+) T regulatory cells (Tregs) and mucosal production of IL-10 from non-T cells. Lamina propria mononuclear cells from H. polygyrus-infected mice released less IL-17 and IFN-γ constitutively and when stimulated with OVA or anti-CD3/CD28 monoclonal antibodies. CONCLUSIONS: We developed a murine IBD model featuring antigen-specific enterocolitis and demonstrate for the first time that gut inflammation induced by an antigen could be abrogated by H. polygyrus infection. Protection was associated with suppressed IL-17 and IFN-γ production, induction of Foxp3(+) Tregs, and elevated secretion of non-T-cell-derived IL-10, all of which could be part of the protective processes.


Subject(s)
Disease Models, Animal , Enterocolitis/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/injuries , Inflammation/immunology , Nematospiroides dubius , Strongylida Infections/immunology , Animals , Cells, Cultured , Enterocolitis/parasitology , Enterocolitis/prevention & control , Enzyme-Linked Immunosorbent Assay , Forkhead Transcription Factors/metabolism , Gastrointestinal Tract/parasitology , Homeodomain Proteins/physiology , Inflammation/parasitology , Inflammation/prevention & control , Interferon-gamma/metabolism , Interleukin-10/physiology , Interleukin-17/metabolism , Interleukin-4/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/adverse effects , Strongylida Infections/pathology , Strongylida Infections/prevention & control , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/parasitology , Thy-1 Antigens/physiology
4.
J Immunol ; 185(6): 3184-9, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20702728

ABSTRACT

Less developed countries have a low incidence of immunological diseases like inflammatory bowel disease (IBD), perhaps prevented by the high prevalence of helminth infections in their populations. In the Rag IL-10(-/-) T cell transfer model of colitis, Heligmosomoides polygyrus, an intestinal helminth, prevents and reverses intestinal inflammation. This model of colitis was used to explore the importance of innate immunity in H. polygyrus protection from IBD. Rag mice briefly exposed to H. polygyrus before reconstitution with IL-10(-/-) colitogenic T cells are protected from colitis. Exposure to H. polygyrus before introduction of IL-10(-/-) and OT2 T cells reduced the capacity of the intestinal mucosa to make IFN-gamma and IL-17 after either anti-CD3 mAb or OVA stimulation. This depressed cytokine response was evident even in the absence of colitis, suggesting that the downmodulation in proinflammatory cytokine secretion was not just secondary to improvement in intestinal inflammation. Following H. polygyrus infection, dendritic cells (DCs) from the lamina propria of Rag mice displayed decreased expression of CD80 and CD86, and heightened expression of plasmacytoid dendritic cell Ag-1 and CD40. They were also less responsive to lamina proprias, producing less IL-12p40 and IL-10. Also diminished was their capacity to present OVA to OT2 T cells. These experiments infer that H. polygyrus does not require direct interactions with T or B cells to render animals resistant to colitis. DCs have an important role in driving both murine and human IBD. Data suggest that phenotypic alternations in mucosal DC function are part of the regulatory process.


Subject(s)
Colitis/immunology , Colitis/prevention & control , Immunity, Innate , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Animals , Cells, Cultured , Colitis/parasitology , Dendritic Cells/immunology , Dendritic Cells/parasitology , Disease Models, Animal , Immunity, Innate/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/parasitology , Inflammatory Bowel Diseases/prevention & control , Interleukin-10/deficiency , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucous Membrane/immunology , Mucous Membrane/parasitology , Mucous Membrane/pathology , Ovalbumin/immunology , Strongylida Infections/genetics , Strongylida Infections/parasitology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/transplantation
5.
Proc Natl Acad Sci U S A ; 107(9): 4293-8, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20160079

ABSTRACT

Substance P (SP) is a proinflammatory mediator implicated in inflammatory bowel disease (IBD) and other inflammatory states. SP acts by stimulating the neurokinin-1 receptor (NK-1R) on T lymphocytes and other cell types, and regulates these cells in a complex interplay with multiple cytokines. The mechanisms of interaction among these inflammatory mediators are not yet fully understood. Here, we demonstrate that function of the NK-1R, a member of the G protein-coupled receptor (GPCR) superfamily, is modulated by TGF-beta. The latter acts not on a GPCR but via serine-threonine kinase-class receptors. By flow confocal image analysis, we demonstrate that TGF-beta delays SP-induced NK-1R internalization on mucosal T cells isolated from a mouse model of IBD and on granuloma T cells in murine schistosomiasis. Furthermore, luciferase reporter-gene assays revealed that NK-1R stimulation activates the nuclear factor of activated T cell- and activator protein-1-dependent signaling pathways, which are known triggers of effector T-cell cytokine production. TGF-beta markedly increases SP-induced activation of these signaling cascades, suggesting that delayed NK-1R internalization results in enhanced signaling. Providing a link to amplified immune function, SP and TGF-beta, when applied in combination, trigger a strong release of the proinflammatory cytokines IFN-gamma and IL17 from intestinal inflammatory T cells, whereas either agonist alone shows no effect. These observations establish precedent that members of two distinct receptor superfamilies can interact via a previously unrecognized mechanism, and reveal a paradigm of GPCR transregulation that is relevant to IBD and possibly other disease processes.


Subject(s)
Endocytosis , Receptors, Neurokinin-1/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/physiology , Animals , Cell Line , Flow Cytometry , Humans , Inflammatory Bowel Diseases/immunology , Interleukin-10/genetics , Interleukin-10/physiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Receptors, Neurokinin-1/metabolism , Signal Transduction , Substance P
6.
Infect Immun ; 76(8): 3651-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18505813

ABSTRACT

Substance P is a tachykinin that enhances pathways of inflammation. Leukocytes at sites of intestinal inflammation make substance P. This study explored the role of interleukin-12 (IL-12), IL-23, and the regulatory cytokines IL-10 and transforming growth factor beta (TGF-beta) in controlling leukocyte substance P production. In murine schistosomiasis, it was found that IL-12 and IL-23 drive substance P gene expression and peptide synthesis in murine splenic T cells and macrophages, respectively. Cytokine induction of substance P synthesis both in T cells and in macrophages depends on intracellular NF-kappaB activation and is Stat4 independent. IL-10 inhibits T-cell substance P production, while TGF-beta blocks macrophage substance P expression. Intestinal macrophages also produce substance P, subject mostly to IL-23 and TGF-beta regulation. Hemokinin is another tachykinin with homology to substance P. Macrophages and T cells make hemokinin, but hemokinin production is not subject to IL-12 or IL-23 regulation.


Subject(s)
Interleukin-10/metabolism , Interleukin-12/immunology , Interleukin-23/immunology , Macrophages/immunology , Substance P/biosynthesis , T-Lymphocytes/immunology , Transforming Growth Factor beta/metabolism , Animals , Gene Expression Profiling , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/metabolism , Schistosomiasis/immunology , Spleen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...