Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










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.
Eur J Immunol ; 39(7): 1870-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19544487

ABSTRACT

Colonization with helminthic parasites induces mucosal regulatory cytokines, like IL-10 or TGF-beta, that are important in suppressing colitis. Helminths induce mucosal T cell IL-10 secretion and regulate lamina propria mononuclear cell (LPMC) Th1 cytokine generation in an IL-10-dependent manner in WT mice. Helminths also stimulate mucosal TGF-beta release. As TGF-beta exerts major regulatory effects on T lymphocytes, we investigated the role of T lymphocyte TGF-beta signaling in helminthic modulation of intestinal immunity. T cell TGF-beta signaling is interrupted in TGF-beta receptor II dominant negative (TGF-betaRII DN) mice by T-cell-specific over-expression of a TGF-betaRII DN. We studied LPMC responses in WT and TGF-betaRII DN mice that were uninfected or colonized with the nematode, Heligmosomoides polygyrus. Our results indicate an essential role of T cell TGF-beta signaling in limiting mucosal Th1 and Th2 responses. Furthermore, we demonstrate that helminthic induction of intestinal T cell IL-10 secretion requires intact T cell TGF-beta-signaling pathway. Helminths fail to curtail robust, dysregulated intestinal Th1 cytokine production and chronic colitis in TGF-betaRII DN mice. Thus, T cell TGF-beta signaling is essential for helminthic stimulation of mucosal IL-10 production, helminthic modulation of intestinal IFN-gamma generation and H. polygyrus-mediated suppression of chronic colitis.


Subject(s)
Cytokines/metabolism , Nematospiroides dubius/physiology , Signal Transduction/physiology , Strongylida Infections/metabolism , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Colitis/immunology , Colitis/metabolism , Colitis/parasitology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Host-Parasite Interactions , Interferon-gamma/metabolism , Interleukin-10/metabolism , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism , Intestinal Diseases, Parasitic/parasitology , Intestine, Small/cytology , Intestine, Small/metabolism , Intestine, Small/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/genetics , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Lymphocytes/cytology , Transforming Growth Factor beta/genetics
7.
Infect Immun ; 76(11): 5164-72, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18710859

ABSTRACT

Infection with the trematode helminth Schistosoma mansoni results in a parasite egg-induced, CD4 T-cell-mediated, hepatointestinal granulomatous and fibrosing inflammation that varies greatly in severity, with a higher frequency of milder forms typically occurring in regions where the disease is endemic. One possible explanation for this is that in these regions the degree of inflammation is lessened by widespread concurrent infection with gastrointestinal nematodes. We tested this hypothesis by establishing a murine coinfection model in which mice were infected with the intestinal nematode parasite Heligmosomoides polygyrus prior to infection with S. mansoni. In CBA mice that naturally display a severe form of schistosomiasis, preinfection with H. polygyrus resulted in a marked reduction in schistosome egg-induced hepatic immunopathology, which was associated with significant decreases in the levels of interleukin-17 (IL-17), gamma interferon, tumor necrosis factor alpha, IL-23, IL-6, and IL-1beta and with increases in the levels of IL-4, IL-5, IL-10, and transforming growth factor beta in mesenteric lymph node cells, purified CD4 T cells, and isolated liver granuloma cells. There also were increases in liver Ym1 and forkhead box P3 transcription factor expression. In another model of high-pathology schistosomiasis induced in C57BL/6 mice by immunization with schistosome egg antigens in complete Freund's adjuvant, coinfection with the nematodes also resulted in a marked inhibition of hepatic immunopathology accompanied by similar shifts in cytokine production. These findings demonstrate that intestinal nematodes prevent Th1- and Th17-cell-mediated inflammation by promoting a strong Th2-polarized environment associated with increases in the levels of alternatively activated macrophages and T regulatory cells, which result in significant amelioration of schistosome-induced immunopathology.


Subject(s)
Cytokines/biosynthesis , Liver Diseases, Parasitic/immunology , Liver/parasitology , Schistosomiasis mansoni/complications , Strongylida Infections/complications , Animals , Cytokines/immunology , Disease Models, Animal , Female , Liver/immunology , Liver/pathology , Liver Diseases, Parasitic/parasitology , Liver Diseases, Parasitic/pathology , Macrophages/immunology , Mice , Nematospiroides dubius/immunology , Ovum , Reverse Transcriptase Polymerase Chain Reaction , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/pathology , Strongylida Infections/immunology , Strongylida Infections/pathology , T-Lymphocytes, Regulatory/immunology
8.
J Immunol ; 181(4): 2414-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684931

ABSTRACT

Helminth exposure appears to protect hosts from inappropriate inflammatory responses, such as those causing inflammatory bowel disease. A recently identified, strongly proinflammatory limb of the immune response is characterized by T cell IL-17 production. Many autoimmune type inflammatory diseases are associated with IL-17 release. Because helminths protect from these diseases, we examined IL-17 production in helminth-colonized mice. We colonized mice with Heligmosomoides polygyrus, an intestinal helminth, and analyzed IL-17 production by lamina propria mononuclear cells (LPMC) and mesenteric lymph node (MLN) cells. Colonization with H. polygyrus reduces IL-17A mRNA by MLN cells and inhibits IL-17 production by cultured LPMC and MLN cells. Helminth exposure augments IL-4 and IL-10 production. Blocking both IL-4 and IL-10, but not IL-10 alone, restores IL-17 production in vitro. Colonization of colitic IL-10-deficient mice with H. polygyrus suppresses LPMC IL-17 production and improves colitis. Ab-mediated blockade of IL-17 improves colitis in IL-10-deficient mice. Thus, helminth-associated inhibition of IL-17 production is most likely an important mechanism mediating protection from inappropriate intestinal inflammation.


Subject(s)
Immune Tolerance , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Nematospiroides dubius/growth & development , Nematospiroides dubius/immunology , Animals , Cells, Cultured , Colitis/immunology , Colitis/metabolism , Colitis/parasitology , Interleukin-17/metabolism , Interleukin-4/physiology , Intestinal Mucosa/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/parasitology , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
9.
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
10.
Infect Immun ; 75(9): 4655-63, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606601

ABSTRACT

Helminths down-regulate inflammation and may prevent development of several autoimmune illnesses, such as inflammatory bowel disease. We determined if exposure to the duodenal helminth Heligmosomoides polygyrus establishes cytokine pathways in the distal intestine that may protect from intestinal inflammation. Mice received 200 H. polygyrus larvae and were studied 2 weeks later. Lamina propria mononuclear cells (LPMC) were isolated from the terminal ileum for analysis and in vitro experiments. Mice with H. polygyrus were resistant to trinitrobenzenesulfonic acid (TNBS)-induced colitis, a Th1 cytokine-dependent inflammation. Heligmosomoides polygyrus did not change the normal microscopic appearance of the terminal ileum and colon and minimally affected LPMC composition. However, colonization altered LPMC cytokine profiles, blocking gamma interferon (IFN-gamma) and interleukin 12 (IL-12) p40 release but promoting IL-4, IL-5, IL-13, and IL-10 secretion. IL-10 blockade in vitro with anti-IL-10 receptor (IL-10R) monoclonal antibody restored LPMC IFN-gamma and IL-12 p40 secretion. IL-10 blockade in vivo worsened TNBS colitis in H. polygyrus-colonized mice. Lamina propria CD4(+) T cells isolated from colonized mice inhibited IFN-gamma production by splenic T cells from worm-free mice. This inhibition did not require cell contact and was dependent on IL-10. Heligmosomoides polygyrus colonization inhibits Th1 and promotes Th2 and regulatory cytokine production in distant intestinal regions without changing histology or LPMC composition. IL-10 is particularly important for limiting the Th1 response. The T-cell origin of these cytokines demonstrates mucosal regulatory T-cell induction.


Subject(s)
Cytokines/biosynthesis , Intestine, Large/parasitology , Nematospiroides dubius/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Intestine, Large/immunology , Mice , Mice, Inbred C57BL , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology , T-Lymphocytes, Regulatory/parasitology
11.
Am J Physiol Gastrointest Liver Physiol ; 291(2): G253-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16825660

ABSTRACT

This study determined whether Heligmosomoides polygyrus induces intestinal regulatory T cells. Splenic T cells proliferate strongly when cultured with anti-CD3 and antigen-presenting cells (APC). Lamina propria T cells from mice with H. polygyrus mixed with normal splenic T cells from uninfected mice inhibited proliferation over 90%. Lamina propria T cells from mice without H. polygyrus only modestly affected T cell proliferation. The worm-induced regulatory T cell was CD8+ and required splenic T cell contact to inhibit proliferation. The regulation also was IL-10 independent, but TAP-dependent, suggesting that it requires major histocompatibility complex (MHC) class I interaction. Additional studies employed mice with transgenic T cells that did not express functional TGF-beta receptors. The lamina propria T regulator inhibited proliferation of these transgenic T cells nearly 100%, suggesting that TGF-beta signaling via the T cell was not required. CD8+ T cells were needed for worms to reverse piroxicam-induced colitis in Rag mice (T and B cell deficient) reconstituted with IL-10-/- T cells. Thus H. polygyrus induces a regulatory CD8+ lamina propria T cell that inhibits T cell proliferation and that appears to have a role in control of colitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Intestines/immunology , Lymphocyte Activation/immunology , Nematospiroides dubius , Strongylida Infections/immunology , Strongylida Infections/pathology , Animals , CD8-Positive T-Lymphocytes/pathology , Colitis/parasitology , Intestines/parasitology , Mice , Mice, Inbred C57BL
12.
J Immunol ; 176(2): 726-9, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16393954

ABSTRACT

Helminths are immune modulators that down-regulate colitis in inflammatory bowel disease. In animal models, intestinal bacteria drive colitis and in humans certain alleles of the LPS receptor protein TLR4 increase inflammatory bowel disease susceptibility. To understand helminthic immune modulation in the gut, we studied the influence of intestinal Heligmosomoides polygyrus colonization on LPS-induced lamina propria mononuclear cell (LPMC) cytokine responses in mice. LPS did not stimulate TGFbeta production from LPMC of uninfected mice. LPS strongly induced LPMC from worm-infected animals to secrete TGFbeta, but not TNF-alpha or IL-12. The TGFbeta derived from mucosal T cells. Helminth infection up-regulated TLR4 expression only in lamina propria T cells. LPMC from worm-infected TLR4 mutant animals did not respond to LPS, suggesting that LPS required TLR4 to stimulate TGFbeta secretion. Thus, during helminth infection, LPS challenge induces mucosal T cells to make TGFbeta through a TLR4-dependent process without promoting synthesis of proinflammatory cytokines.


Subject(s)
Nematospiroides dubius/pathogenicity , Strongylida Infections/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 4/biosynthesis , Transforming Growth Factor beta/biosynthesis , Animals , Cells, Cultured , Gene Expression , Immunity, Mucosal , In Vitro Techniques , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nematospiroides dubius/immunology , Strongylida Infections/genetics , T-Lymphocytes/drug effects , Toll-Like Receptor 4/genetics
13.
Eur J Immunol ; 34(10): 2690-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15368285

ABSTRACT

Inflammatory bowel disease (IBD) is prevalent in industrialized countries, but rare in less-developed countries. Helminths, common in less-developed countries, may induce immunoregulatory circuits protective against IBD. IL-10(-/-) mice given piroxicam develop severe and persistent colitis. Lamina propria mononuclear cells from colitic IL-10(-/-) mice released IFN-gamma and IL-12. The ongoing piroxicam-induced colitis could be partially blocked with anti-IL-12 monoclonal antibody suggesting that the inflammation was at least partly IL-12 dependent. Colonization of piroxicam-treated colitic IL-10(-/-) mice with Heligmosomoides polygyrus (an intestinal helminth) suppressed established inflammation and inhibited mucosal IL-12 and IFN-gamma production. H. polygyrus augmented mucosal IL-13, but not IL-4 or IL-5 production. Transfer of mesenteric lymph node (MLN) T cells from IL-10(-/-) animals harboring H. polygyrus into colitic IL-10(-/-) recipients inhibited colitis. MLN T cells from worm-free mice did not. Foxp3 (scurfin) drives regulatory T cell function. H. polygyrus enhanced Foxp3 mRNA expression in MLN T cells that had regulatory activity. This suggests that H. polygyrus inhibits ongoing IL-10(-/-) colitis in part through blocking mucosal Th1 cytokine production. Resolution of inflammation is associated with increased IL-13 production and can be adoptively transferred by MLN T cells.


Subject(s)
Colitis/immunology , Colitis/microbiology , Interleukin-10/deficiency , Strongylida Infections/immunology , Th1 Cells/immunology , Adoptive Transfer , Animals , Colitis/chemically induced , Colitis/pathology , Cyclooxygenase Inhibitors/pharmacology , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-12/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Nematospiroides dubius/immunology , Piroxicam/pharmacology , RNA, Messenger/analysis , Th1 Cells/microbiology
14.
J Immunol ; 172(11): 6528-32, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15153465

ABSTRACT

Substance P (SP) belongs to the tachykinin family of molecules. SP, cleaved from preprotachykinin A, is a neuropeptide and a proinflammatory leukocyte product. SP engages neurokinin 1 receptor (NK-1R) to stimulate cells. Hemokinin (HK) is another tachykinin that binds NK-1R. HK comes from preprotachykinin C, which is distinct from preprotachykinin A. We determined whether HK functions like SP at inflammatory sites. Preprotachykinin C mRNA was in murine schistosome granulomas and intestinal lamina propria mononuclear cells. Granuloma T cells and macrophages expressed preprotachykinin C mRNA. HK bound granuloma T cell NK-1R with high affinity. SP and HK stimulated IFN-gamma production with equal potency. NK-1R antagonist blocked the effect of SP and HK on IFN-gamma secretion. Thus, both HK and SP are expressed at sites of chronic inflammation and share cell origin, receptor, and immunoregulatory function. Two distinct but functionally overlapping tachykinins govern inflammation through NK-1R at sites of chronic inflammation.


Subject(s)
Inflammation/metabolism , Protein Precursors/physiology , Substance P/physiology , Tachykinins/physiology , Animals , Cell Line , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Protein Precursors/genetics , Receptors, Neurokinin-1/metabolism , Tachykinins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...