Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(3): 374, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30705416

ABSTRACT

In the version of this article initially published, a word ("neutraling") in sentence 2 of paragraph 5 is incorrect. The correct phrase is "...neutralizing properties...". The error has been corrected in the HTML and PDF version of the article.

3.
Nat Immunol ; 18(5): 541-551, 2017 05.
Article in English | MEDLINE | ID: mdl-28288099

ABSTRACT

Inflammatory bowel diseases involve the dynamic interaction of host genetics, the microbiome and inflammatory responses. Here we found lower expression of NLRP12 (which encodes a negative regulator of innate immunity) in human ulcerative colitis, by comparing monozygotic twins and other patient cohorts. In parallel, Nlrp12 deficiency in mice caused increased basal colonic inflammation, which led to a less-diverse microbiome and loss of protective gut commensal strains (of the family Lachnospiraceae) and a greater abundance of colitogenic strains (of the family Erysipelotrichaceae). Dysbiosis and susceptibility to colitis associated with Nlrp12 deficency were reversed equally by treatment with antibodies targeting inflammatory cytokines and by the administration of beneficial commensal Lachnospiraceae isolates. Fecal transplants from mice reared in specific-pathogen-free conditions into germ-free Nlrp12-deficient mice showed that NLRP12 and the microbiome each contributed to immunological signaling that culminated in colon inflammation. These findings reveal a feed-forward loop in which NLRP12 promotes specific commensals that can reverse gut inflammation, while cytokine blockade during NLRP12 deficiency can reverse dysbiosis.


Subject(s)
Clostridiales/physiology , Colitis, Ulcerative/immunology , Colon/physiology , Firmicutes/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Microbiota , RNA, Ribosomal, 16S/analysis , Animals , Biodiversity , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colon/microbiology , Dextran Sulfate , Feces/microbiology , Gene-Environment Interaction , Humans , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/genetics , Microbiota/immunology , Symbiosis , Twins, Monozygotic
6.
Proc Natl Acad Sci U S A ; 116(52): 26157-26166, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31811024

ABSTRACT

The mammalian immune system is tolerized to trillions of microbes residing on bodily surfaces and can discriminate between symbionts and pathogens despite their having related microbial structures. Mechanisms of innate immune activation and the subsequent signaling pathways used by symbionts to communicate with the adaptive immune system are poorly understood. Polysaccharide A (PSA) of Bacteroides fragilis is the model symbiotic immunomodulatory molecule. Here we demonstrate that PSA-dependent immunomodulation requires the Toll-like receptor (TLR) 2/1 heterodimer in cooperation with Dectin-1 to initiate signaling by the downstream phosphoinositide 3-kinase (PI3K) pathway, with consequent CREB-dependent transcription of antiinflammatory genes, including antigen presentation and cosignaling molecules. High-resolution LC-MS/MS analysis of PSA identified a previously unknown small molecular-weight, covalently attached bacterial outer membrane-associated lipid that is required for activation of antigen-presenting cells. This archetypical commensal microbial molecule initiates a complex collaborative integration of Toll-like receptor and C-type lectin-like receptor signaling mechanisms culminating in the activation of the antiinflammatory arm of the PI3K pathway that serves to educate CD4+ Tregs to produce the immunomodulatory cytokine IL-10. Immunomodulation is a key function of the microbiome and is a focal point for developing new therapeutic agents.

7.
J Immunol ; 192(8): 3958-68, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24634494

ABSTRACT

The p110δ subunit of class IA PI3K modulates signaling in innate immune cells. We previously demonstrated that mice harboring a kinase-dead p110δ subunit (p110δ(KD)) develop spontaneous colitis. Macrophages contributed to the Th1/Th17 cytokine bias in p110δ(KD) mice through increased IL-12 and IL-23 expression. In this study, we show that the enteric microbiota is required for colitis development in germfree p110δ(KD) mice. Colonic tissue and macrophages from p110δ(KD) mice produce significantly less IL-10 compared with wild-type mice. p110δ(KD) APCs cocultured with naive CD4+ Ag-specific T cells also produce significantly less IL-10 and induce more IFN-γ- and IL-17A-producing CD4+ T cells compared with wild-type APCs. Illustrating the importance of APC-T cell interactions in colitis pathogenesis in vivo, Rag1(-/-)/p110δ(KD) mice develop mild colonic inflammation and produced more colonic IL-12p40 compared with Rag1(-/-) mice. However, CD4+ CD45RB(high/low) T cell Rag1(-/-)/p110δ(KD) recipient mice develop severe colitis with increased percentages of IFN-γ- and IL-17A-producing lamina propria CD3+D4+ T cells compared with Rag1(-/-) recipient mice. Intestinal tissue samples from patients with Crohn's disease reveal significantly lower expression of PIK3CD compared with intestinal samples from non-inflammatory bowel disease control subjects (p < 0.05). PIK3CD expression inversely correlates with the ratio of IL12B:IL10 expression. In conclusion, the PI3K subunit p110δ controls homeostatic APC-T cell interactions by altering the balance between IL-10 and IL-12/23. Defects in p110δ expression and/or function may underlie the pathogenesis of human inflammatory bowel disease and lead to new therapeutic strategies.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Colitis/immunology , Colitis/metabolism , Immunity, Innate , Th1 Cells/metabolism , Th17 Cells/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Cytokines/biosynthesis , Disease Models, Animal , Gene Expression Regulation , Immunity, Innate/genetics , Interleukin-10/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Microbiota , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/immunology , Th17 Cells/immunology
8.
J Immunol ; 192(4): 1918-27, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442434

ABSTRACT

NFIL3 is a transcription factor that regulates multiple immunologic functions. In myeloid cells, NFIL3 is IL-10 inducible and has a key role as a repressor of IL-12p40 transcription. NFIL3 is a susceptibility gene for the human inflammatory bowel diseases. In this article, we describe spontaneous colitis in Nfil3(-/-) mice. Mice lacking both Nfil3 and Il10 had severe early-onset colitis, suggesting that NFIL3 and IL-10 independently regulate mucosal homeostasis. Lymphocytes were necessary for colitis, because Nfil3/Rag1 double-knockout mice were protected from disease. However, Nfil3/Rag1 double-knockout mice adoptively transferred with wild-type CD4(+) T cells developed severe colitis compared with Rag1(-/-) recipients, suggesting that colitis was linked to defects in innate immune cells. Colitis was abrogated in Nfil3/Il12b double-deficient mice, identifying Il12b dysregulation as a central pathogenic event. Finally, germ-free Nfil3(-/-) mice do not develop colonic inflammation. Thus, NFIL3 is a microbiota-dependent, IL-10-independent regulator of mucosal homeostasis via IL-12p40.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Interleukin-10/genetics , Interleukin-12 Subunit p40/metabolism , Interleukin-23 Subunit p19/metabolism , Microbiota/immunology , Adoptive Transfer , Animals , Arabidopsis Proteins/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Colon/immunology , Colon/pathology , Genetic Predisposition to Disease , Interleukin-12 Subunit p40/genetics , Interleukin-23 Subunit p19/genetics , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/immunology , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/genetics
9.
Microbiol Immunol ; 59(8): 452-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26146866

ABSTRACT

Induction of mammalian heme oxygenase (HO)-1 and exposure of animals to carbon monoxide (CO) ameliorates experimental colitis. When enteric bacteria, including Escherichia coli, are exposed to low iron conditions, they express an HO-like enzyme, chuS, and metabolize heme into iron, biliverdin and CO. Given the abundance of enteric bacteria residing in the intestinal lumen, our postulate was that commensal intestinal bacteria may be a significant source of CO and those that express chuS and other Ho-like molecules suppress inflammatory immune responses through release of CO. According to real-time PCR, exposure of mice to CO results in changes in enteric bacterial composition and increases E. coli 16S and chuS DNA. Moreover, the severity of experimental colitis correlates positively with E. coli chuS expression in IL-10 deficient mice. To explore functional roles, E. coli were genetically modified to overexpress chuS or the chuS gene was deleted. Co-culture of chuS-overexpressing E. coli with bone marrow-derived macrophages resulted in less IL-12p40 and greater IL-10 secretion than in wild-type or chuS-deficient E. coli. Mice infected with chuS-overexpressing E. coli have more hepatic CO and less serum IL-12 p40 than mice infected with chuS-deficient E. coli. Thus, CO alters the composition of the commensal intestinal microbiota and expands populations of E. coli that harbor the chuS gene. These bacteria are capable of attenuating innate immune responses through expression of chuS. Bacterial HO-like molecules and bacteria-derived CO may represent novel targets for therapeutic intervention in inflammatory conditions.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/immunology , Heme Oxygenase (Decyclizing)/immunology , Heme Oxygenase (Decyclizing)/metabolism , Immune Evasion , Immunity, Innate , Animals , Carbon Monoxide/metabolism , Cells, Cultured , Coculture Techniques , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Escherichia coli/metabolism , Gene Deletion , Gene Expression , Heme Oxygenase (Decyclizing)/genetics , Interleukin-10/metabolism , Interleukin-12 Subunit p40/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics
10.
J Immunol ; 191(5): 2780-2795, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918988

ABSTRACT

The spleen regulatory B cell subset with the functional capacity to express IL-10 (B10 cells) modulates both immune responses and autoimmune disease severity. However, the peritoneal cavity also contains relatively high frequencies of functionally defined IL-10-competent B10 cells. In this study, peritoneal cavity B10 cells shared similar cell surface phenotypes with their spleen counterparts. However, peritoneal cavity B10 cells were 10-fold more frequent among B cells than occurred within the spleen, intestinal tract, or mesenteric lymph nodes and were present at higher proportions among the phenotypically defined peritoneal B1a > B1b > B2 cell subpopulations. The development or localization of B10 cells within the peritoneal cavity was not dependent on the presence of commensal microbiota, T cells, IL-10 or B10 cell IL-10 production, or differences between their fetal liver or adult bone marrow progenitor cell origins. The BCR repertoire of peritoneal cavity B10 cells was diverse, as occurs in the spleen, and predominantly included germline-encoded VH and VL regions commonly found in either the conventional or B1 B cell compartments. Thereby, the capacity to produce IL-10 appears to be an intrinsic functional property acquired by clonally diverse B cells. Importantly, IL-10 production by peritoneal cavity B cells significantly reduced disease severity in spontaneous and induced models of colitis by regulating neutrophil infiltration, colitogenic CD4(+) T cell activation, and proinflammatory cytokine production during colitis onset. Thus, the numerically small B10 cell subset within the peritoneal cavity has regulatory function and is important for maintaining homeostasis within gastrointestinal tissues and the immune system.


Subject(s)
B-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Peritoneal Cavity/cytology , Adoptive Transfer , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Gastroenterology ; 144(4): 789-98, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23266559

ABSTRACT

BACKGROUND & AIMS: Heme oxygenase-1 (HO-1) and its metabolic by-product, carbon monoxide (CO), protect against intestinal inflammation in experimental models of colitis, but little is known about their intestinal immune mechanisms. We investigated the interactions among CO, HO-1, and the enteric microbiota in mice and zebrafish. METHODS: Germ-free, wild-type, and interleukin (Il)10(-/-) mice and germ-free zebrafish embryos were colonized with specific pathogen-free (SPF) microbiota. Germ-free or SPF-raised wild-type and Il10(-/-) mice were given intraperitoneal injections of cobalt(III) protoporphyrin IX chloride (CoPP), which up-regulates HO-1, the CO-releasing molecule Alfama-186, or saline (control). Colitis was induced in wild-type mice housed in SPF conditions by infection with Salmonella typhimurium. RESULTS: In colons of germ-free, wild-type mice, SPF microbiota induced production of HO-1 via activation of nuclear factor erythroid 2-related factor 2-, IL-10-, and Toll-like receptor-dependent pathways; similar observations were made in zebrafish. SPF microbiota did not induce HO-1 in colons of germ-free Il10(-/-) mice. Administration of CoPP to Il10(-/-) mice before transition from germ-free to SPF conditions reduced their development of colitis. In Il10(-/-) mice, CO and CoPP reduced levels of enteric bacterial genomic DNA in mesenteric lymph nodes. In mice with S typhimurium-induced enterocolitis, CoPP reduced the numbers of live S typhimurium recovered from the lamina propria, mesenteric lymph nodes, spleen, and liver. Knockdown of HO-1 in mouse macrophages impaired their bactericidal activity against E coli, E faecalis, and S typhimurium, whereas exposure to CO or overexpression of HO-1 increased their bactericidal activity. HO-1 induction and CO increased acidification of phagolysosomes. CONCLUSIONS: Colonic HO-1 prevents colonic inflammation in mice. HO-1 is induced by the enteric microbiota and its homeostatic function is mediated, in part, by promoting bactericidal activities of macrophages.


Subject(s)
Bacterial Translocation/physiology , Carbon Monoxide/pharmacology , Colitis/prevention & control , Heme Oxygenase-1/metabolism , Salmonella typhimurium/physiology , Animals , Bacterial Translocation/drug effects , Blotting, Western , Colitis/drug therapy , Colitis/microbiology , Disease Models, Animal , Escherichia coli/pathogenicity , Gentamicins/pharmacology , Heme Oxygenase-1/biosynthesis , Macrophages/cytology , Macrophages/physiology , Metagenome , Mice , Mice, Inbred C57BL , Random Allocation , Real-Time Polymerase Chain Reaction
12.
J Immunol ; 188(10): 4897-905, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22508928

ABSTRACT

Graft-versus-host disease (GVHD) is a severe and frequent complication of allogeneic bone marrow transplantation (BMT) that involves the gastrointestinal (GI) tract and lungs. The pathobiology of GVHD is complex and involves immune cell recognition of host Ags as foreign. We hypothesize a central role for the collectin surfactant protein A (SP-A) in regulating the development of GVHD after allogeneic BMT. C57BL/6 (H2b; WT) and SP-A-deficient mice on a C57BL/6 background (H2b; SP-A(-/-)) mice underwent allogeneic or syngeneic BMT with cells from either C3HeB/FeJ (H2k; SP-A-deficient recipient mice that have undergone an allogeneic BMT [SP-A(-/-)alloBMT] or SP-A-sufficient recipient mice that have undergone an allogeneic BMT) or C57BL/6 (H2b; SP-A-deficient recipient mice that have undergone a syngeneic BMT or SP-A-sufficient recipient mice that have undergone a syngeneic BMT) mice. Five weeks post-BMT, mice were necropsied, and lung and GI tissue were analyzed. SP-A(-/-) alloBMT or SP-A-sufficient recipient mice that have undergone an allogeneic BMT had no significant differences in lung pathology; however, SP-A(-/-)alloBMT mice developed marked features of GI GVHD, including decreased body weight, increased tissue inflammation, and lymphocytic infiltration. SP-A(-/-)alloBMT mice also had increased colon expression of IL-1ß, IL-6, TNF-α, and IFN-γ and as well as increased Th17 cells and diminished regulatory T cells. Our results demonstrate the first evidence, to our knowledge, of a critical role for SP-A in modulating GI GVHD. In these studies, we demonstrate that mice deficient in SP-A that have undergone an allogeneic BMT have a greater incidence of GI GVHD that is associated with increased Th17 cells and decreased regulatory T cells. The results of these studies demonstrate that SP-A protects against the development of GI GVHD and establishes a role for SP-A in regulating the immune response in the GI tract.


Subject(s)
Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Pulmonary Surfactant-Associated Protein A/physiology , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Gastrointestinal Diseases/genetics , Graft vs Host Disease/genetics , Lymphocyte Count , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Surfactant-Associated Protein A/deficiency , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology
13.
J Immunol ; 189(4): 1792-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786766

ABSTRACT

To prevent excessive inflammatory responses to commensal microbes, intestinal macrophages, unlike their systemic counterparts, do not produce inflammatory cytokines in response to enteric bacteria. Consequently, loss of macrophage tolerance to the enteric microbiota plays a central role in the pathogenesis of inflammatory bowel diseases. Therefore, we examined whether the hyporesponsive phenotype of intestinal macrophages is programmed by prior exposure to the microbiota. IL-10, but not in vivo exposure to the microbiota, programs intestinal macrophage tolerance, because wild-type (WT) colonic macrophages from germ-free and specific pathogen-free (SPF)-derived mice produce IL-10, but not IL-12 p40, when activated with enteric bacteria. Basal and activated IL-10 expression is mediated through a MyD88-dependent pathway. Conversely, colonic macrophages from germ-free and SPF-derived colitis-prone Il10(-/-) mice demonstrated robust production of IL-12 p40. Next, mechanisms through which IL-10 inhibits Il12b expression were investigated. Although Il12b mRNA was transiently induced in LPS-activated WT bone marrow-derived macrophages (BMDMs), expression persisted in Il10(-/-) BMDMs. There were no differences in nucleosome remodeling, mRNA stability, NF-κB activation, or MAPK signaling to explain prolonged transcription of Il12b in Il10(-/-) BMDMs. However, acetylated histone H4 transiently associated with the Il12b promoter in WT BMDMs, whereas association of these factors was prolonged in Il10(-/-) BMDMs. Experiments using histone deacetylase (HDAC) inhibitors and HDAC3 short hairpin RNA indicate that HDAC3 is involved in histone deacetylation of the Il12b promoter by IL-10. These results suggest that histone deacetylation on the Il12b promoter by HDAC3 mediates homeostatic effects of IL-10 in macrophages.


Subject(s)
Gene Expression Regulation/immunology , Homeostasis/immunology , Interleukin-10/immunology , Interleukin-12 Subunit p40/biosynthesis , Macrophages/immunology , Acetylation , Animals , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , Histones/genetics , Histones/immunology , Histones/metabolism , Immune Tolerance/genetics , Immune Tolerance/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics
14.
J Immunol ; 186(8): 4649-55, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21383239

ABSTRACT

Regulation of innate inflammatory responses against the enteric microbiota is essential for the maintenance of intestinal homeostasis. Key participants in innate defenses are macrophages. In these studies, the basic leucine zipper protein, NFIL3, is identified as a regulatory transcription factor in macrophages, controlling IL-12 p40 production induced by bacterial products and the enteric microbiota. Exposure to commensal bacteria and bacterial products induced NFIL3 in cultured macrophages and in vivo. The Il12b promoter has a putative DNA-binding element for NFIL3. Basal and LPS-activated NFIL3 binding to this site was confirmed by chromatin immunoprecipitation. LPS-induced Il12b promoter activity was inhibited by NFIL3 expression and augmented by NFIL3-short hairpin RNA in an Il12b-bacterial artificial chromosome-GFP reporter macrophage line. Il12b inhibition by NFIL3 does not require IL-10 expression, but a C-terminal minimal repression domain is necessary. Furthermore, colonic CD11b(+) lamina propria mononuclear cells from Nfil3(-/-) mice spontaneously expressed Il12b mRNA. Importantly, lower expression of NFIL3 was observed in CD14(+) lamina propria mononuclear cells from Crohn's disease and ulcerative colitis patients compared with control subjects. Likewise, no induction of Nfil3 was observed in colons of colitis-prone Il10(-/-) mice transitioned from germ-free to a conventional microbiota. In conclusion, these experiments characterize NFIL3 as an Il12b transcriptional inhibitor. Interactions of macrophages with the enteric microbiota induce NFIL3 to limit their inflammatory capacity. Furthermore, altered intestinal NFIL3 expression may have implications for the pathogenesis of experimental and human inflammatory bowel diseases.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Immunity, Mucosal/immunology , Interleukin-12 Subunit p40/immunology , Macrophages/immunology , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Molecular Sequence Data , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Sequence Homology, Nucleic Acid
15.
J Immunol ; 186(9): 5506-13, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21444764

ABSTRACT

Cigarette smoking is a significant environmental factor in the human inflammatory bowel diseases, remarkably, conferring protection in ulcerative colitis. We previously demonstrated that a prominent component of cigarette smoke, CO, suppresses Th17-mediated experimental colitis in IL-10(-/-) mice through a heme oxygenase (HO)-1-dependent pathway. In this study, homeostatic and therapeutic effects of CO and HO-1 were determined in chronic colonic inflammation in TCR-α-deficient ((-/-)) mice, in which colitis is mediated by Th2 cytokines, similar to the cytokine milieu described in human ulcerative colitis. TCRα(-/-) mice exposed to CO or treated with the pharmacologic HO-1 inducer cobalt protoporphyrin demonstrated amelioration of active colitis. CO and cobalt protoporphyrin suppressed colonic IL-1ß, TNF, and IL-4 production, whereas IL-10 protein secretion was increased. CO induced IL-10 expression in macrophages and in vivo through an HO-1-dependent pathway. Bacterial products regulate HO-1 expression in macrophages through MyD88- and IL-10-dependent pathways. CO exposure and pharmacologic HO-1 induction in vivo resulted in increased expression of HO-1 and IL-10 in CD11b(+) lamina propria mononuclear cells. Moreover, induction of the IL-10 family member IL-22 was demonstrated in CD11b(-) lamina propria mononuclear cells. In conclusion, CO and HO-1 induction ameliorated active colitis in TCRα(-/-) mice, and therapeutic effects correlated with induction of IL-10. This study provides further evidence that HO-1 mediates an important homeostatic pathway with pleiotropic anti-inflammatory effects in different experimental models of colitis and that targeting HO-1, therefore, is a potential therapeutic strategy in human inflammatory bowel diseases.


Subject(s)
Carbon Monoxide/pharmacology , Colitis/immunology , Heme Oxygenase-1/immunology , Interleukin-10/immunology , Th2 Cells/immunology , Animals , Blotting, Western , Cell Separation , Colitis/pathology , Cytokines/biosynthesis , Cytokines/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Heme Oxygenase-1/metabolism , Interleukin-10/biosynthesis , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
16.
J Biol Chem ; 286(2): 1174-80, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21097874

ABSTRACT

We have demonstrated previously that IFN-γ plays a protective role in the initiation of chronic intestinal inflammation through attenuation of Toll-like receptor-mediated IL-23 induction in macrophages. Here, an interferon-stimulated response element (ISRE) is identified in a region of conserved nucleotide sequences in the Il23a promoter. This ISRE mediated, in part, Il23a promoter induction by LPS and inhibition of LPS-induced activity by IFN-γ. LPS and IFN-γ recruit interferon regulatory factors (IRFs) to the Il23a ISRE in murine bone marrow-derived macrophages (BMMs). Functionally, IRF-1 is a negative regulator of Il23a in LPS-stimulated BMMs. IRF-1(-/-) BMMs demonstrated enhanced LPS-induced Il23a expression compared with WT BMMs. Moreover, IRF-1 deficiency resulted in prolonged occupancy of RelA on the Il23a promoter. Consequently, IRF-1(-/-) mice were more susceptible to colonic injury by trinitrobenzenesulfonic acid, and IL-10/IRF-1 double-deficient (IL-10/IRF-1(-/-)) mice demonstrated more severe colonic inflammation compared with IL-10(-/-) mice. The severity of colitis in both models correlated with increased colonic IL-23. CD11b(+) lamina propria mononuclear cells, comprising predominantly macrophages, were identified as the major source of IL-23 in colitis-prone mice. Basal and heat-killed Escherichia coli-stimulated levels of Il23a were increased in IL-10/IRF-1(-/-) compared with WT and IL-10(-/-) colonic CD11b(+) lamina propria mononuclear cells. In conclusion, these experiments characterize IRF-ISRE interactions on the Il23a promoter, which have in vivo relevance as a homeostatic checkpoint in chronic intestinal inflammation.


Subject(s)
Colitis/genetics , Colitis/immunology , Interferon-gamma/immunology , Interleukin-23 Subunit p19/genetics , Response Elements/genetics , Animals , Bone Marrow Cells/physiology , Chronic Disease , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-23 Subunit p19/immunology , Intestinal Mucosa/immunology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Response Elements/immunology , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism
17.
Gastroenterology ; 140(6): 1838-46, 2011 May.
Article in English | MEDLINE | ID: mdl-21530750

ABSTRACT

In this review, we speculate about future therapeutic approaches for inflammatory bowel diseases (IBDs), focusing on the need for better preclinical and clinical models and approaches beyond small molecules and systemically administered biologics. We offer ideas to change clinical trial programs and to use immunologic and genetic biomarkers to personalize medicine. We attempt to reconcile past therapeutic successes and failures to improve future approaches. Some of our ideas might be provocative, but we hope that the examples we provide will stimulate discussion about what will advance the field of IBD therapy.


Subject(s)
Gastroenterology/trends , Inflammatory Bowel Diseases/therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Clinical Trials as Topic , Drug Delivery Systems , Genetic Therapy , Humans , Immunity , Intestinal Mucosa/immunology , Precision Medicine/methods , Stem Cell Transplantation , T-Lymphocytes, Regulatory/transplantation
18.
J Immunol ; 184(8): 4069-73, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20228197

ABSTRACT

IL-23 regulation is a central event in the pathogenesis of the inflammatory bowel diseases. We demonstrate that IFN-gamma has anti-inflammatory properties in the initiation phase of IL-23-mediated experimental colitis. IFN-gamma attenuates LPS-mediated IL-23 expression in murine macrophages. Mechanistically, IFN-gamma inhibits Il23a promoter activation through altering NF-kappaB binding and histone modification. Moreover, intestinal inflammation is inhibited by IFN-gamma signaling through attenuation of Il23a gene expression. In germ-free wild-type mice colonized with enteric microbiota, inhibition of colonic Il23a temporally correlates with induction of IFN-gamma. IFN-gammaR1/IL-10 double-deficient mice demonstrate markedly increased colonic inflammation and IL23a expression compared with those of IL-10(-/-) mice. Colonic CD11b(+) cells are the primary source of IL-23 and a target for IFN-gamma. This study describes an important anti-inflammatory role for IFN-gamma through inhibition of IL-23. Converging genetic and functional findings suggest that IL-23 and IFN-gamma are important pathogenic molecules in human inflammatory bowel disease.


Subject(s)
Colitis/immunology , Down-Regulation/immunology , Interferon-gamma/physiology , Interleukin-23 Subunit p19/antagonists & inhibitors , Macrophages/immunology , Macrophages/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Colitis/metabolism , Colitis/pathology , Disease Models, Animal , Gene Expression Regulation/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Interleukin-23 Subunit p19/biosynthesis , Interleukin-23 Subunit p19/physiology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/immunology
19.
J Exp Med ; 202(12): 1703-13, 2005 Dec 19.
Article in English | MEDLINE | ID: mdl-16365149

ABSTRACT

Heme oxygenase (HO)-1 and its metabolic product carbon monoxide (CO) play regulatory roles in acute inflammatory states. In this study, we demonstrate that CO administration is effective as a therapeutic modality in mice with established chronic colitis. CO administration ameliorates chronic intestinal inflammation in a T helper (Th)1-mediated model of murine colitis, interleukin (IL)-10-deficient (IL-10(-/-)) mice. In Th1-mediated inflammation, CO abrogates the synergistic effect of interferon (IFN)-gamma on lipopolysaccharide-induced IL-12 p40 in murine macrophages and alters IFN-gamma signaling by inhibiting a member of the IFN regulatory factor (IRF) family of transcription factors, IRF-8. A specific signaling pathway, not previously identified, is delineated that involves an obligatory role for HO-1 induction in the protection afforded by CO. Moreover, CO antagonizes the inhibitory effect of IFN-gamma on HO-1 expression in macrophages. In macrophages and in Th1-mediated colitis, pharmacologic induction of HO-1 recapitulates the immunosuppressive effects of CO. In conclusion, this study begins to elucidate potential etiologic and therapeutic implications of CO and the HO-1 pathway in chronic inflammatory bowel diseases.


Subject(s)
Carbon Monoxide/therapeutic use , Colitis/drug therapy , Heme Oxygenase-1/metabolism , Signal Transduction/immunology , Administration, Inhalation , Animals , Carbon Monoxide/administration & dosage , Carbon Monoxide/metabolism , Colitis/immunology , DNA Primers , Enzyme Induction/drug effects , Enzyme-Linked Immunosorbent Assay , Heme Oxygenase-1/biosynthesis , Interferon Regulatory Factors/metabolism , Interferon-gamma/antagonists & inhibitors , Interleukin-10/genetics , Mice , Mice, Knockout , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction , Th1 Cells/immunology
20.
Gastroenterology ; 139(5): 1642-53, 1653.e1-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20637203

ABSTRACT

BACKGROUND & AIMS: Innate immune responses are crucial for host defense against pathogens but need to be tightly regulated to prevent chronic inflammation. Initial characterization of mice with a targeted inactivating mutation in the p110δ subunit of phosphoinositide 3-kinase (PI3K p110δ(D910A/D910A)) revealed defects in B- and T-cell signaling and chronic colitis. Here, we further characterize features of inflammatory bowel diseases in these mice and investigate underlying innate immune defects. METHODS: Colons and macrophages from PI3K p110δ(D910A/D910A) mice were evaluated for colonic inflammation and innate immune dysfunction. Colonic p110δ messenger RNA expression was examined in interleukin (IL)-10(-/-) and wild-type germ-free mice during transition to a conventional microbiota. To assess polygenic impact on development of colitis, p110δ(D910A/D910A) mice were backcrossed to IL-10(-/-) mice. RESULTS: A mild spontaneous colitis was shown in PI3K p110δ(D910A/D910A) mice at 8 weeks, with inflammation increasing with age. An inflammatory mucosal and systemic cytokine profile was characterized by expression of IL-12/23. In PI3K p110δ(D910A/D910A) macrophages, augmented toll-like receptor signaling and defective bactericidal activity were observed. Consistent with an important homeostatic role for PI3K p110δ, wild-type mice raised in a germ-free environment markedly up-regulated colonic PI3K p110δ expression with the introduction of the enteric microbiota; however, colitis-prone IL-10(-/-) mice did not. Moreover, PI3K p110δ(D910A/D910A) mice crossed to IL-10(-/-) mice developed severe colitis at an early age. CONCLUSIONS: This study describes a novel model of experimental colitis that highlights the importance of PI3K p110δ in maintaining mucosal homeostasis and could provide insight into the pathogenesis of human inflammatory bowel disease.


Subject(s)
Colitis/pathology , Gene Expression , Immunity, Innate/physiology , Macrophages/metabolism , Phosphatidylinositol 3-Kinase/genetics , RNA/genetics , Animals , Chronic Disease , Colitis/immunology , Colitis/metabolism , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinase/biosynthesis , Phosphatidylinositol 3-Kinase/deficiency , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL