Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Immunol ; 202(8): 2473-2481, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30850474

ABSTRACT

Helminthic infections modulate host immunity and may protect their hosts from developing immunological diseases like inflammatory bowel disease. Induction of regulatory T cells (Tregs) may be an important part of this protective process. Heligmosomoides polygyrus bakeri infection also promotes the production of the regulatory cytokines TGF-ß and IL-10 in the gut. In the intestines, TGF-ß helps induce regulatory T cells. This study used Foxp3/IL-10 double reporter mice to investigate the effect of TGF-ß on the differentiation of colon and mesenteric lymph node-derived murine Foxp3- IL-10- CD4+ T cells into their regulatory phenotypes. Foxp3- IL-10- CD4+ T cells from H. polygyrus bakeri-infected mice, as opposed to T cells from uninfected animals, cultured in vitro with TGF-ß and anti-CD3/CD28 mAb differentiated into Foxp3+ and/or IL-10+ T cells. The IL-10-producing T cells nearly all displayed CD25. Smad7 is a natural inhibitor of TGF-ß signaling. In contrast to gut T cells from uninfected mice, Foxp3- IL10- CD4+ T cells from H. polygyrus bakeri-infected mice displayed reduced Smad7 expression and responded to TGF-ß with Smad2/3 phosphorylation. The TGF-ß-induced Tregs that express IL-10 blocked colitis when transferred into the Rag/CD25- CD4+ T cell transfer model of inflammatory bowel disease. TGF-ß had a greatly diminished capacity to induce Tregs in H. polygyrus bakeri-infected transgenic mice with constitutively high T cell-specific Smad7 expression. Thus, infection with H. polygyrus bakeri causes down-modulation in Smad7 expression in intestinal CD4+ T cells, which allows the TGF-ß produced in response to the infection to induce the Tregs that prevent colitis.


Subject(s)
Colitis/immunology , Interleukin-10/immunology , Nematospiroides dubius/immunology , Smad7 Protein/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Colitis/pathology , Colitis/prevention & control , Interleukin-10/genetics , Mice , Mice, Transgenic , Smad7 Protein/genetics , Strongylida Infections/genetics , Strongylida Infections/pathology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics
2.
J Immunol ; 197(7): 2948-57, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27559049

ABSTRACT

Helminthic infections modulate host immunity and may protect people in less-developed countries from developing immunological diseases. In a murine colitis model, the helminth Heligmosomoides polygyrus bakeri prevents colitis via induction of regulatory dendritic cells (DCs). The mechanism driving the development of these regulatory DCs is unexplored. There is decreased expression of the intracellular signaling pathway spleen tyrosine kinase (Syk) in intestinal DCs from H. polygyrus bakeri-infected mice. To explore the importance of this observation, it was shown that intestinal DCs from DC-specific Syk(-/-) mice were powerful inhibitors of murine colitis, suggesting that loss of Syk was sufficient to convert these cells into their regulatory phenotype. DCs sense gut flora and damaged epithelium via expression of C-type lectin receptors, many of which signal through the Syk signaling pathway. It was observed that gut DCs express mRNA encoding for C-type lectin (CLEC) 7A, CLEC9A, CLEC12A, and CLEC4N. H. polygyrus bakeri infection downmodulated CLEC mRNA expression in these cells. Focusing on CLEC7A, which encodes for the dectin-1 receptor, flow analysis showed that H. polygyrus bakeri decreases dectin-1 expression on the intestinal DC subsets that drive Th1/Th17 development. DCs become unresponsive to the dectin-1 agonist curdlan and fail to phosphorylate Syk after agonist stimulation. Soluble worm products can block CLEC7A and Syk mRNA expression in gut DCs from uninfected mice after a brief in vitro exposure. Thus, downmodulation of Syk expression and phosphorylation in intestinal DCs could be important mechanisms through which helminths induce regulatory DCs that limit colitis.


Subject(s)
Colitis/prevention & control , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intestines/cytology , Signal Transduction , Syk Kinase/metabolism , Animals , Colitis/immunology , Colitis/parasitology , Disease Models, Animal , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Syk Kinase/deficiency , Syk Kinase/genetics
3.
Science ; 351(6279): 1329-33, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26847546

ABSTRACT

The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.


Subject(s)
Chemoreceptor Cells/immunology , Intestinal Diseases, Parasitic/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Microbiota/immunology , TRPM Cation Channels/immunology , Animals , Doublecortin-Like Kinases , Eosinophils/immunology , Goblet Cells/immunology , Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Immunity, Mucosal , Interleukin-13/immunology , Interleukin-17/immunology , Intestinal Diseases, Parasitic/parasitology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Serine-Threonine Kinases/immunology , Protozoan Infections/immunology , Protozoan Infections/parasitology , Signal Transduction , Taste , Transducin/genetics , Transducin/immunology , Tritrichomonas/immunology
4.
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
5.
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
6.
Inflamm Bowel Dis ; 16(11): 1841-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20848461

ABSTRACT

BACKGROUND: In a murine model of inflammatory bowel disease (IBD), treatment of colitis in IL-10 gene-deficient mice with the parasitic helminth Heligmosomoides polygyrus ameliorates colonic inflammation. The cellular and molecular mechanisms driving this therapeutic host response are being studied vigorously. One proposed mechanism is that H. polygyrus infection favors the outgrowth or suppression of certain bacteria, which in turn help modulate host immunity. METHODS: To quantify the effect of H. polygyrus infection on the composition of the gastrointestinal (GI) tract microbiota, we conducted two independent microbial ecology analyses of C57BL/6 mice. We obtained and analyzed 3,353 bacterial 16S rRNA encoding gene sequences from the ileum and cecum of infected and uninfected mice as well as incective H. polygyrus larvae at the outset of the second experiment and adult worms taken directly from the mouse duodenum at the end of the second experiment. RESULTS: We found that a significant shift in the abundance and relative distribution of bacterial species in the ileum of mice is associated with H. polygyrus infection. Members of the bacterial family Lactobacillaceae significantly increased in abundance in the ileum of infected mice reproducibly in two independent experiments despite having different microbiotas present at the outset of each experiment. CONCLUSIONS: These data support the concept that helminth infection shifts the composition of intestinal bacteria. The clinical consequences of these shifts in intestinal flora are yet to be explored.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Nematospiroides dubius/immunology , Animals , Disease Models, Animal , Genes, rRNA , Ileum/immunology , Interleukin-10/genetics , Lactobacillaceae/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
7.
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
8.
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
9.
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
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 ; 174(7): 3906-11, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15778345

ABSTRACT

Substance P (SP), a neuropeptide, interacts with the neurokinin 1 receptor (NK-1R) on immune cells to help control IFN-gamma production. In murine schistosomiasis mansoni, schistosome worms produce ova that incite focal Th2-type granulomatous inflammation within the liver and intestines. Normal gut is characterized by a controlled state of inflammation. IL-10 knockout mice develop chronic Th1-type colitis spontaneously. Both schistosome granulomas and gut mucosa display an SP immune regulatory circuit. However, the origin and regulation of SP production at these sites of inflammation are poorly understood. Macrophages are a potential source of SP. We therefore studied macrophages (F4/80(+)) from these models of inflammation. SP mRNA (preprotachykinin A (PPT A)) was detected within the schistosome granuloma, spleen, and lamina propria macrophages. Compared with those from wild-type mice, granuloma macrophages from STAT6(-/-) mice had 10-fold higher PPT A mRNA expression, whereas in STAT4(-/-) animals, PPT A mRNA expression was nearly abolished. IL-12 signals via STAT4 to induce Th1-type inflammation. It was demonstrated that IL-12, but not IL-18, induces SP mRNA expression in resting splenic macrophages from Schistosoma-infected mice and in wild-type lamina propria mononuclear cells. Thus, macrophages are a source for SP at these sites of chronic inflammation, and IL-12 and STAT4 are regulators of macrophage SP mRNA expression.


Subject(s)
Inflammation/pathology , Interleukin-12/physiology , Macrophages/immunology , RNA, Messenger/analysis , Substance P/genetics , Animals , DNA-Binding Proteins/physiology , Gene Expression Regulation , Granuloma/immunology , Inflammation/immunology , Macrophages/chemistry , Mice , Mice, Inbred Strains , Mice, Knockout , STAT4 Transcription Factor , Schistosomiasis/immunology , Spleen/immunology , Trans-Activators/physiology
13.
Am J Physiol Gastrointest Liver Physiol ; 287(2): G320-5, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15246967

ABSTRACT

Products of arachidonic acid metabolism are important for mucosal homeostasis, because blockade of this pathway with an NSAID triggers rapid onset of severe colitis in the IL-10 knockout (IL-10(-/-)) model of IBD. Rag mice do not make T or B cells. This study determined whether reconstitution of Rag mice with T cells from IL-10(-/-) mice transferred NSAID colitis susceptibility. Rag mice were reconstituted by intraperitoneal injection with splenocytes from wild-type (WT) or IL-10(-/-) animals. Colitis was induced by using piroxicam and was graded histologically. Isolated lamina propria mononuclear cells (LPMC), lamina propria T cells, and LPMC depleted of T cells from reconstituted Rag mice were studied for cytokine production. Only animals reconstituted with IL-10(-/-) CD4(+) T cells and administered piroxicam developed severe colitis. LPMC from these colitic animals made IFN-gamma, whose production was dependent on T cells. Some IL-10 was produced but only from non-T cells. LPMC from the healthy Rag mice that were reconstituted with WT T cells and were piroxicam resistant made much more IL-10. This was mostly T cell dependent. In conclusion, only CD4(+) T cells from IL-10(-/-) animals leave Rag mice susceptible to NSAID-induced, Th1 colitis. Lamina propria T cells normally make large quantities of IL-10, suggesting that IL-10 from T cells may be protective.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , CD4-Positive T-Lymphocytes/physiology , Cell Transplantation , Colitis/chemically induced , Interleukin-10/deficiency , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Disease Susceptibility , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Piroxicam/adverse effects , Spleen/cytology , Spleen/metabolism
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
15.
Am J Physiol Gastrointest Liver Physiol ; 284(2): G197-204, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12388184

ABSTRACT

Substance P (SP) enhances antigen-dependent T cell IFN-gamma production. It was determined if a T cell neurokinin-1 receptor (NK-1R) was critical for IFN-gamma regulation. T cells from schistosome-infected mice were mixed with splenocytes from uninfected NK-1R knockout (KO) animals. Thus only the schistosome egg antigen-specific T cells expressed NK-1R. The cells were cultured 18 h with or without SP. SP enhanced antigen-induced IFN-gamma production fourfold without affecting IL-4 or IL-5 secretion. NK-1R inhibitor blocked this stimulation. Neither purified T cells nor naive KO splenocytes cultured alone responded to antigen. To further define the importance of T cell NK-1R, we developed a T cell-selective NK-1R KO mouse by reconstituting T cell-deficient Rag mice with NK-1R KO T cells. These mice challanged with schistosomiasis developed abnormal liver granulomas. Granuloma size was smaller in T cell-selective NK-1R KO mice compared with granulomas in Rag reconstituted with normal T cells. Splenocytes and granuloma cells from NK-1R KO mice made less IFN-gamma. The mice also made less IgG2a. Thus T cell NK-1R is important for IFN-gamma regulation.


Subject(s)
Antigens/pharmacology , Interferon-gamma/biosynthesis , Receptors, Neurokinin-1/physiology , Substance P/physiology , T-Lymphocytes/physiology , Animals , Cell Separation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Granuloma/metabolism , Granuloma/pathology , Immunoglobulin G/biosynthesis , Interleukin-4/metabolism , Interleukin-5/metabolism , Liver/cytology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , Receptors, Neurokinin-1/biosynthesis , Receptors, Neurokinin-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Schistosomiasis/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
16.
Am J Physiol Gastrointest Liver Physiol ; 283(1): G115-21, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065298

ABSTRACT

In schistosomiasis, eggs induce granulomas that have a vasoactive intestinal peptide (VIP) immunoregulatory circuit. This study explored the regulation of VIP production at sites of inflammation. Splenocytes from uninfected C57BL/6 mice expressed VIP mRNA and protein, which stopped following egg deposition. Eggs induce a Th2 response, suggesting that Th2 cytokines like interleukin (IL)-4 can regulate VIP. To address this issue, splenocytes from uninfected mice were incubated for 4 h with or without recombinant IL-4. IL-4 inhibited VIP mRNA expression. F4/80+ macrophages were the source of constitutively expressed VIP, subject to IL-4 regulation. In IL-4 knockout mice, splenic VIP production did not downmodulate during schistosome infection, suggesting that IL-4 is a critical cytokine regulating VIP production in wild-type mouse spleen. IL-4-producing granulomas in schistosomiasis made VIP. Experiments showed that granuloma VIP derived from F4/80- (nonmacrophage) cell populations, explaining this paradox. Granuloma F4/80+ cells from IL-4 knockout mice expressed VIP. Thus macrophages can make VIP, which is subject to IL-4 regulation. However, in the Th2 granulomas, other cell types produce VIP, which compensates for loss of macrophages as a source of this molecule.


Subject(s)
Interleukin-4/physiology , Macrophages/metabolism , Schistosomiasis mansoni/metabolism , Vasoactive Intestinal Peptide/antagonists & inhibitors , Animals , Cells, Cultured , Granuloma/metabolism , Granuloma/parasitology , Interleukin-4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Protein Precursors/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Reference Values , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/pathology , Spleen/metabolism , Spleen/pathology , Vasoactive Intestinal Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...