Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nature ; 504(7478): 158-62, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24196717

ABSTRACT

Newborn infants are highly susceptible to infection. This defect in host defence has generally been ascribed to the immaturity of neonatal immune cells; however, the degree of hyporesponsiveness is highly variable and depends on the stimulation conditions. These discordant responses illustrate the need for a more unified explanation for why immunity is compromised in neonates. Here we show that physiologically enriched CD71(+) erythroid cells in neonatal mice and human cord blood have distinctive immunosuppressive properties. The production of innate immune protective cytokines by adult cells is diminished after transfer to neonatal mice or after co-culture with neonatal splenocytes. Neonatal CD71(+) cells express the enzyme arginase-2, and arginase activity is essential for the immunosuppressive properties of these cells because molecular inhibition of this enzyme or supplementation with L-arginine overrides immunosuppression. In addition, the ablation of CD71(+) cells in neonatal mice, or the decline in number of these cells as postnatal development progresses parallels the loss of suppression, and restored resistance to the perinatal pathogens Listeria monocytogenes and Escherichia coli. However, CD71(+) cell-mediated susceptibility to infection is counterbalanced by CD71(+) cell-mediated protection against aberrant immune cell activation in the intestine, where colonization with commensal microorganisms occurs swiftly after parturition. Conversely, circumventing such colonization by using antimicrobials or gnotobiotic germ-free mice overrides these protective benefits. Thus, CD71(+) cells quench the excessive inflammation induced by abrupt colonization with commensal microorganisms after parturition. This finding challenges the idea that the susceptibility of neonates to infection reflects immune-cell-intrinsic defects and instead highlights processes that are developmentally more essential and inadvertently mitigate innate immune protection. We anticipate that these results will spark renewed investigation into the need for immunosuppression in neonates, as well as improved strategies for augmenting host defence in this vulnerable population.


Subject(s)
Antigens, CD/metabolism , Erythroid Cells/immunology , Escherichia coli Infections/immunology , Immune Tolerance/immunology , Listeriosis/immunology , Receptors, Transferrin/metabolism , Animals , Animals, Newborn , Arginase/genetics , Arginase/metabolism , Disease Susceptibility/immunology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Erythroid Cells/enzymology , Escherichia coli/immunology , Female , Fetal Blood/cytology , Humans , Immune Tolerance/drug effects , Immune Tolerance/genetics , Listeria monocytogenes/immunology , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
2.
Proc Natl Acad Sci U S A ; 111(29): 10672-7, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002484

ABSTRACT

The costimulatory B7-1 (CD80)/B7-2 (CD86) molecules, along with T-cell receptor stimulation, together facilitate T-cell activation. This explains why in vivo B7 costimulation neutralization efficiently silences a variety of human autoimmune disorders. Paradoxically, however, B7 blockade also potently moderates accumulation of immune-suppressive regulatory T cells (Tregs) essential for protection against multiorgan systemic autoimmunity. Here we show that B7 deprivation in mice overrides the necessity for Tregs in averting systemic autoimmunity and inflammation in extraintestinal tissues, whereas peripherally induced Tregs retained in the absence of B7 selectively mitigate intestinal inflammation caused by Th17 effector CD4(+) T cells. The need for additional immune suppression in the intestine reflects commensal microbe-driven T-cell activation through the accessory costimulation molecules ICOSL and OX40L. Eradication of commensal enteric bacteria mitigates intestinal inflammation and IL-17 production triggered by Treg depletion in B7-deficient mice, whereas re-establishing intestinal colonization with Candida albicans primes expansion of Th17 cells with commensal specificity. Thus, neutralizing B7 costimulation uncovers an essential role for Tregs in selectively averting intestinal inflammation by Th17 CD4(+) T cells with commensal microbe specificity.


Subject(s)
B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inflammation/immunology , Interleukin-17/biosynthesis , Intestines/pathology , OX40 Ligand/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CTLA-4 Antigen/metabolism , Candida albicans/physiology , Cell Differentiation/immunology , Cell Proliferation , Humans , Inflammation/microbiology , Inflammation/pathology , Intestines/immunology , Intestines/microbiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Phenotype , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
3.
Histochem Cell Biol ; 146(4): 445-55, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27246004

ABSTRACT

Guanylin (GN) and uroguanylin (UGN), through activation of guanylyl cyclase C (GCC), serve to control intestinal fluid homeostasis. Both peptides are produced in the intestinal epithelium, but their cellular origin has not been fully charted. Using quantitative PCR and an improved in situ hybridization technique (RNAscope), we have assessed the expression of GN (Guca2a), UGN (Guca2b), and GCC (Gucy2c) in mouse intestine. In the crypts of Lieberkühn, expression of Guca2a and Guca2b was restricted to cells of secretory lineage, at the crypt's base, and to a region above, previously identified as a common origin of cellular differentiation. In this compartment, comparatively uniform levels of Guca2a and Guca2b expression were observed throughout the length of the gut. In contrast, Guca2a and Guca2b expression in the villus-surface region was more variable, and reflected the distinct, but overlapping expression pattern observed previously. Accordingly, in jejunum and ileum, Guca2a and Guca2b were abundantly expressed by enterocytes, whereas in colon only Guca2a transcript was found in the surface region. In duodenum, only low levels of Guca2b transcript were observed in columnar cells, and Guca2a expression was restricted entirely to cells of the secretory lineage. Gucy2c was shown to be expressed relatively uniformly along the rostrocaudal and crypt-villus axes and was also found in the duodenal glands. Our study reveals novel aspects of the cellular localization of the GCC signaling axis that, apart from its role in the regulation of fluid balance, link it to pH regulation, cell cycle control, and host defense.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gastrointestinal Hormones/biosynthesis , Intestines/cytology , Natriuretic Peptides/biosynthesis , Animals , Gastrointestinal Hormones/analysis , Gastrointestinal Hormones/genetics , Intestinal Mucosa/metabolism , Mice , Mice, Inbred Strains , Natriuretic Peptides/analysis , Natriuretic Peptides/genetics , Signal Transduction
4.
Blood ; 123(12): 1938-47, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24501220

ABSTRACT

Fanconi anemia (FA) is a genetic disorder associated with bone marrow (BM) failure and leukemia. Recent studies demonstrate variable immune defects in FA. However, the cause for FA immunodeficiency is unknown. Here we report that deletion of Fanca or Fancd2 dysregulates the suppressive activity of regulatory T cells (Tregs), shown functionally as exacerbation of graft-vs-host disease (GVHD) in mice. Recipient mice of Fanca(-/-) or Fancd2(-/-) BM chimeras exhibited severe acute GVHD after allogeneic BM transplantation (BMT). T cells from Fanca(-/-) or Fancd2(-/-) mice induced higher GVHD lethality than those from wild-type (WT) littermates. FA Tregs possessed lower proliferative suppression potential compared with WT Tregs, as demonstrated by in vitro proliferation assay and BMT. Analysis of CD25(+)Foxp3(+) Tregs indicated that loss of Fanca or Fancd2 dysregulated Foxp3 target gene expression. Additionally, CD25(+)Foxp3(+) Tregs of Fanca(-/-) or Fancd2(-/-) mice were less efficient in suppressing the production of GVHD-associated inflammatory cytokines. Consistently, aberrant NF-κB activity was observed in infiltrated T cells from FA GVHD mice. Conditional deletion of p65 in FA Tregs decreased GVHD mortality. Our study uncovers an essential role for FA proteins in maintaining Treg homeostasis, possibly explaining, at least in part, the immune deficiency reported in some FA patients.


Subject(s)
Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/immunology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , Cytokines/biosynthesis , Fanconi Anemia/genetics , Fanconi Anemia/immunology , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Forkhead Transcription Factors/metabolism , Gene Expression , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism , Transplantation Chimera
5.
Hepatology ; 59(5): 1830-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24115079

ABSTRACT

UNLABELLED: Inflammation plays a central pathogenic role in the pernicious metabolic and end-organ sequelae of obesity. Among these sequelae, nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease in the developed world. The twinned observations that obesity is associated with increased activation of the interleukin (IL)-17 axis and that this axis can regulate liver damage in diverse contexts prompted us to address the role of IL-17RA signaling in the progression of NAFLD. We further examined whether microbe-driven IL-17A regulated NAFLD development and progression. We show here that IL-17RA(-/-) mice respond to high-fat diet stress with significantly greater weight gain, visceral adiposity, and hepatic steatosis than wild-type controls. However, obesity-driven lipid accumulation was uncoupled from its end-organ consequences in IL-17RA(-/-) mice, which exhibited decreased steatohepatitis, nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase enzyme expression, and hepatocellular damage. Neutralization of IL-17A significantly reduced obesity-driven hepatocellular damage in wild-type mice. Further, colonization of mice with segmented filamentous bacteria (SFB), a commensal that induces IL-17A production, exacerbated obesity-induced hepatocellular damage. In contrast, SFB depletion protected from obesity-induced hepatocellular damage. CONCLUSION: These data indicate that obesity-driven activation of the IL-17 axis is central to the development and progression of NAFLD to steatohepatitis and identify the IL-17 pathway as a novel therapeutic target in this condition.


Subject(s)
Fatty Liver/etiology , Interleukin-17/physiology , Signal Transduction/physiology , Animals , Bacterial Infections/complications , Diet, High-Fat , Disease Progression , Fatty Liver/microbiology , Inflammation/etiology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Obesity/complications , Reactive Oxygen Species/metabolism , Receptors, Interleukin-17/physiology
6.
J Immunol ; 191(5): 2806-17, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23926327

ABSTRACT

Hematopoietic stem cells (HSCs) can either self-renew or differentiate into various types of cells of the blood lineage. Signaling pathways that regulate this choice of self-renewal versus differentiation are currently under extensive investigation. In this study, we report that deregulation of Notch signaling skews HSC differentiation in mouse models of Fanconi anemia (FA), a genetic disorder associated with bone marrow failure and progression to leukemia and other cancers. In mice expressing a transgenic Notch reporter, deletion of the Fanca or Fancc gene enhances Notch signaling in multipotential progenitors (MPPs), which is correlated with decreased phenotypic long-term HSCs and increased formation of MPP1 progenitors. Furthermore, we found an inverse correlation between Notch signaling and self-renewal capacity in FA hematopoietic stem and progenitor cells. Significantly, FA deficiency in MPPs deregulates a complex network of genes in the Notch and canonical NF-κB pathways. Genetic ablation or pharmacologic inhibition of NF-κB reduces Notch signaling in FA MPPs to near wild type level, and blocking either NF-κB or Notch signaling partially restores FA HSC quiescence and self-renewal capacity. These results suggest a functional crosstalk between Notch signaling and NF-κB pathway in regulation of HSC differentiation.


Subject(s)
Cell Differentiation/physiology , Fanconi Anemia/metabolism , Hematopoietic Stem Cells/metabolism , NF-kappa B/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Disease Models, Animal , Flow Cytometry , Immunoblotting , Inflammation/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Receptor Cross-Talk/physiology
7.
J Immunol ; 190(9): 4773-85, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23562811

ABSTRACT

In inflammatory bowel diseases (IBDs), particularly ulcerative colitis, intestinal macrophages (MΦs), eosinophils, and the eosinophil-selective chemokine CCL11, have been associated with disease pathogenesis. MΦs, a source of CCL11, have been reported to be of a mixed classical (NF-κB-mediated) and alternatively activated (STAT-6-mediated) phenotype. The importance of NF-κB and STAT-6 pathways to the intestinal MΦ/CCL11 response and eosinophilic inflammation in the histopathology of experimental colitis is not yet understood. Our gene array analyses demonstrated elevated STAT-6- and NF-κB-dependent genes in pediatric ulcerative colitis colonic biopsies. Dextran sodium sulfate (DSS) exposure induced STAT-6 and NF-κB activation in mouse intestinal F4/80(+)CD11b(+)Ly6C(hi) (inflammatory) MΦs. DSS-induced CCL11 expression, eosinophilic inflammation, and histopathology were attenuated in RelA/p65(Δmye) mice, but not in the absence of STAT-6. Deletion of p65 in myeloid cells did not affect inflammatory MΦ recruitment or alter apoptosis, but did attenuate LPS-induced cytokine production (IL-6) and Ccl11 expression in purified F4/80(+)CD11b(+)Ly6C(hi) inflammatory MΦs. Molecular and cellular analyses revealed a link between expression of calprotectin (S100a8/S100a9), Ccl11 expression, and eosinophil numbers in the DSS-treated colon. In vitro studies of bone marrow-derived MΦs showed calprotectin-induced CCL11 production via a p65-dependent mechanism. Our results indicate that myeloid cell-specific NF-κB-dependent pathways play an unexpected role in CCL11 expression and maintenance of eosinophilic inflammation in experimental colitis. These data indicate that targeting myeloid cells and NF-κB-dependent pathways may be of therapeutic benefit for the treatment of eosinophilic inflammation and histopathology in IBD.


Subject(s)
Chemokine CCL11/metabolism , Eosinophils/metabolism , Inflammation/metabolism , Intestinal Mucosa/metabolism , Myeloid Cells/metabolism , Transcription Factor RelA/metabolism , Animals , Apoptosis/genetics , Chemokine CCL11/genetics , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/pathology , Eosinophils/pathology , Female , Gene Expression , Humans , Inflammation/genetics , Inflammation/pathology , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Intestines/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Transcription Factor RelA/genetics
8.
Curr Opin Gastroenterol ; 30(1): 1-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24304979

ABSTRACT

PURPOSE OF REVIEW: Guanylate cyclase C (GC-C) is a transmembrane receptor that is expressed primarily on intestinal epithelial cells. Activation of this receptor by its endogenous peptide ligands initiates cyclic guanosine monophosphate-dependent (cGMP) salt and water movement in the intestine. GC-C is targeted by the enterotoxigenic Escherichia coli heat-stable enterotoxin STa, which deregulates this pathway and causes secretory diarrhea. This review discusses current work on the physiological function of GC-C in the intestine. RECENT FINDINGS: Familial GC-C mutations demonstrate that epithelial cGMP signaling is critical to electrolyte and fluid balance in the neonatal intestine. Chronic deregulation of GC-C activity in early life increases susceptibility to a number of disorders, including obstruction and inflammatory bowel disease. Murine models indicate that GC-C regulates the composition of intestinal commensal microflora and that it suppresses bacterial infection and modulates colonic injury and inflammation. Therapeutic GC-C ligands are used to successfully treat constipation-predominant irritable bowel syndrome and recent studies show that extracellular cGMP is an important mechanism of reducing abdominal pain associated with this disorder. SUMMARY: Originally identified as a target of E. coli enterotoxin STa, GC-C is an important regulator of physiological salt and water homeostasis and may directly impact a wide range of intestinal disorders.


Subject(s)
Inflammatory Bowel Diseases/enzymology , Irritable Bowel Syndrome/enzymology , Receptors, Atrial Natriuretic Factor/physiology , Adhesins, Bacterial/physiology , Homeostasis/physiology , Humans , Intestinal Absorption/physiology , Intestines/microbiology , Irritable Bowel Syndrome/drug therapy , Receptors, Atrial Natriuretic Factor/agonists , Signal Transduction/physiology , Water-Electrolyte Balance/physiology
9.
BMC Gastroenterol ; 13: 135, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-24004613

ABSTRACT

BACKGROUND: Guanylate Cyclase C (GC-C) is an apically-oriented transmembrane receptor that is expressed on epithelial cells of the intestine. Activation of GC-C by the endogenous ligands guanylin or uroguanylin elevates intracellular cGMP and is implicated in intestinal ion secretion, cell proliferation, apoptosis, intestinal barrier function, as well as the susceptibility of the intestine to inflammation. Our aim was to determine if GC-C is required for host defense during infection by the murine enteric pathogen Citrobacter rodentium of the family Enterobacteriacea. METHODS: GC-C+/+ control mice or those having GC-C genetically ablated (GC-C-/-) were administered C. rodentium by orogastric gavage and analyzed at multiple time points up to post-infection day 20. Commensal bacteria were characterized in uninfected GC-C+/+ and GC-C-/- mice using 16S rRNA PCR analysis. RESULTS: GC-C-/- mice had an increase in C. rodentium bacterial load in stool relative to GC-C+/+. C. rodentium infection strongly decreased guanylin expression in GC-C+/+ mice and, to an even greater degree, in GC-C-/- animals. Fluorescent tracer studies indicated that mice lacking GC-C, unlike GC-C+/+ animals, had a substantial loss of intestinal barrier function early in the course of infection. Epithelial cell apoptosis was significantly increased in GC-C-/- mice following 10 days of infection and this was associated with increased frequency and numbers of C. rodentium translocation out of the intestine. Infection led to significant liver histopathology in GC-C-/- mice as well as lymphocyte infiltration and elevated cytokine and chemokine expression. Relative to naïve GC-C+/+ mice, the commensal microflora load in uninfected GC-C-/- mice was decreased and bacterial composition was imbalanced and included outgrowth of the Enterobacteriacea family. CONCLUSIONS: This work demonstrates the novel finding that GC-C signaling is an essential component of host defense during murine enteric infection by reducing bacterial load and preventing systemic dissemination of attaching/effacing-lesion forming bacterial pathogens such as C. rodentium.


Subject(s)
Colon/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/immunology , Receptors, Guanylate Cyclase-Coupled/immunology , Receptors, Peptide/immunology , Animals , Apoptosis/immunology , Bacterial Load , Bacterial Translocation/physiology , Citrobacter rodentium/physiology , Colon/pathology , Enterobacteriaceae Infections/genetics , Intestinal Mucosa/pathology , Liver/pathology , Mice , Mice, Knockout , Permeability , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Peptide/genetics , Signal Transduction/immunology
10.
J Immunol ; 186(10): 5993-6003, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21498668

ABSTRACT

Recent genome-wide association studies of pediatric inflammatory bowel disease have implicated the 17q12 loci, which contains the eosinophil-specific chemokine gene CCL11, with early-onset inflammatory bowel disease susceptibility. In the current study, we employed a murine model of experimental colitis to define the molecular pathways that regulate CCL11 expression in the chronic intestinal inflammation and pathophysiology of experimental colitis. Bone marrow chimera experiments showed that hematopoietic cell-derived CCL11 is sufficient for CCL11-mediated colonic eosinophilic inflammation. We show that dextran sodium sulfate (DSS) treatment promotes the recruitment of F4/80(+)CD11b(+)CCR2(+)Ly6C(high) inflammatory monocytes into the colon. F4/80(+)CD11b(+)CCR2(+)Ly6C(high) monocytes express CCL11, and their recruitment positively correlated with colonic eosinophilic inflammation. Phenotypic analysis of purified Ly6C(high) intestinal inflammatory macrophages revealed that these cells express both M1- and M2-associated genes, including Il6, Ccl4, Cxcl2, Arg1, Chi3l3, Ccl11, and Il10, respectively. Attenuation of DSS-induced F4/80(+)CD11b(+)CCR2(+)Ly6C(high) monocyte recruitment to the colon in CCR2(-/-) mice was associated with decreased colonic CCL11 expression, eosinophilic inflammation, and DSS-induced histopathology. These studies identify a mechanism for DSS-induced colonic eosinophilia mediated by Ly6C(high)CCR2(+) inflammatory monocyte/macrophage-derived CCL11.


Subject(s)
Chemokine CCL11/immunology , Colitis/immunology , Eosinophilia/immunology , Inflammatory Bowel Diseases/immunology , Animals , Antigens, Differentiation/genetics , Antigens, Ly/analysis , Antigens, Ly/immunology , Bone Marrow Cells , CD11b Antigen/immunology , Chemokine CCL11/genetics , Chemokine CCL11/metabolism , Colitis/chemically induced , Colitis/metabolism , Colon/immunology , Dextran Sulfate/pharmacology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation , Inflammatory Bowel Diseases/metabolism , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Monocytes/drug effects , Monocytes/immunology , Polymerase Chain Reaction , Receptors, CCR2/analysis , Receptors, CCR2/immunology
11.
J Immunol ; 186(12): 7205-14, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21555532

ABSTRACT

Guanylate cyclase C (GUCY2C or GC-C) and its ligands, guanylin (GUCA2A or Gn) and uroguanylin (GUCA2B or Ugn), are expressed in intestinal epithelial cells and regulate ion secretion, intestinal barrier function, and epithelial monolayer homeostasis via cGMP-dependent signaling pathways. The aim of this study was to determine whether GC-C and its ligands direct the course of intestinal inflammation. In this article, we show that dextran sodium sulfate (DSS)-induced clinical disease and histological damage to the colonic mucosa were significantly less severe in GC-C(-/-) mice and moderately reduced in Gn(-/-) animals. Relative to wild-type controls, GC-C(-/-) and Gn(-/-) mice had reduced apoptosis and increased proliferation of intestinal epithelial cells during DSS colitis. Basal and DSS-induced production of resistin-like molecule ß (RELMß) was substantially diminished in GC-C(-/-) mice. RELMß is thought to stimulate cytokine production in macrophages in this disease model and, consistent with this, TNF-α and IFN-γ production was minimal in GC-C(-/-) animals. RELMß and cytokine levels were similar to wild-type in Gn(-/-) mice, however. Colonic instillation of recombinant RELMß by enema into GC-C(-/-) mice restores sensitivity to DSS-mediated mucosal injury. These findings demonstrate a novel role for GC-C signaling in facilitating mucosal wounding and inflammation, and further suggest that this may be mediated, in part, through control of RELMß production.


Subject(s)
Guanylate Cyclase/physiology , Animals , Colonic Diseases/etiology , Colonic Diseases/pathology , Gastrointestinal Hormones/physiology , Hormones, Ectopic/biosynthesis , Hormones, Ectopic/physiology , Inflammation/etiology , Intercellular Signaling Peptides and Proteins , Interferon-gamma/biosynthesis , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Natriuretic Peptides/physiology , Tumor Necrosis Factor-alpha/biosynthesis
12.
J Biol Chem ; 286(15): 13357-69, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21303908

ABSTRACT

Interleukin-13 (IL-13) has been linked to the pathogenesis of inflammatory diseases of the gastrointestinal tract. It is postulated that IL-13 drives inflammatory lesions through the modulation of both hematopoietic and nonhematopoietic cell function in the intestine. To delineate the relevant contribution of elevated levels of intestinal IL-13 to intestinal structure and function, we generated an intestinal IL-13 transgenic mouse (iIL-13Tg). We show that constitutive overexpression of IL-13 in the small bowel induces modification of intestinal epithelial architecture (villus blunting, goblet cell hyperplasia, and increased epithelial proliferation) and epithelial function (altered basolateral → apical Cl(-) ion conductance). Pharmacological analyses in vitro and in vivo determined that elevated Cl(-) conductance is mediated by altered cystic fibrosis transmembrane conductance regulator expression and activity. Generation of iIL-13Tg/Il13rα1(-/-), iIL-13Tg/Il13rα2(-/-), and iIL-13Tg/Stat6(-/-) mice revealed that IL-13-mediated dysregulation of epithelial architecture and Cl(-) conductance is dependent on IL-13Rα1 and STAT-6. These observations demonstrate a central role for the IL-13/IL-13Rα1 pathway in the regulation of intestinal epithelial cell Cl(-) secretion via up-regulation of cystic fibrosis transmembrane conductance regulator, suggesting an important role for this pathway in secretory diarrhea.


Subject(s)
Chlorides/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Interleukin-13 Receptor alpha1 Subunit/metabolism , Interleukin-13/metabolism , Intestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Animals , Caco-2 Cells , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cysts/genetics , Cysts/metabolism , Cysts/pathology , Diarrhea/genetics , Diarrhea/metabolism , Diarrhea/pathology , Fibrosis , Humans , Interleukin-13/genetics , Interleukin-13 Receptor alpha1 Subunit/genetics , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Intestinal Mucosa/pathology , Ion Transport/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
13.
J Immunol ; 184(7): 3743-54, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20190135

ABSTRACT

Homeostatic control of the immune system involves mechanisms that ensure the self-tolerance, survival and quiescence of hematopoietic-derived cells. In this study, we demonstrate that the GTPase of immunity associated protein (Gimap)5 regulates these processes in lymphocytes and hematopoietic progenitor cells. As a consequence of a recessive N-ethyl-N-nitrosourea-induced germline mutation in the P-loop of Gimap5, lymphopenia, hepatic extramedullary hematopoiesis, weight loss, and intestinal inflammation occur in homozygous mutant mice. Irradiated fetal liver chimeric mice reconstituted with Gimap5-deficient cells lose weight and become lymphopenic, demonstrating a hematopoietic cell-intrinsic function for Gimap5. Although Gimap5-deficient CD4(+) T cells and B cells appear to undergo normal development, they fail to proliferate upon Ag-receptor stimulation although NF-kappaB, MAP kinase and Akt activation occur normally. In addition, in Gimap5-deficient mice, CD4(+) T cells adopt a CD44(high)CD62L(low)CD69(low) phenotype and show reduced IL-7ralpha expression, and T-dependent and T-independent B cell responses are abrogated. Thus, Gimap5-deficiency affects a noncanonical signaling pathway required for Ag-receptor-induced proliferation and lymphocyte quiescence. Antibiotic-treatment or the adoptive transfer of Rag-sufficient splenocytes ameliorates intestinal inflammation and weight loss, suggesting that immune responses triggered by microbial flora causes the morbidity in Gimap5-deficient mice. These data establish Gimap5 as a key regulator of hematopoietic integrity and lymphocyte homeostasis.


Subject(s)
B-Lymphocytes/immunology , Colitis/immunology , GTP Phosphohydrolases/immunology , T-Lymphocytes/immunology , Wasting Syndrome/immunology , Animals , B-Lymphocyte Subsets/immunology , Colitis/genetics , Female , GTP Phosphohydrolases/genetics , GTP-Binding Proteins , Hematopoiesis/genetics , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Homeostasis/genetics , Homeostasis/immunology , Immunoblotting , Inflammation/genetics , Inflammation/immunology , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Liver Diseases/genetics , Liver Diseases/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Self Tolerance/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Wasting Syndrome/genetics
14.
Gastroenterology ; 139(2): 530-41, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20398663

ABSTRACT

BACKGROUND & AIMS: Innate and adaptive immune responses are regulated by cross talk between activation and inhibitory signals. Dysregulation of the inhibitory signal can lead to aberrant chronic inflammatory diseases such as the inflammatory bowel diseases (IBD). Little is known about negative regulation of innate intestinal immune activation. We examined the role of the inhibitory receptor paired immunoglobulin-like receptor B (PIR-B) in the regulation of macrophage function in innate intestinal immunity. METHODS: We examined the susceptibility of Pirb-/- and wild-type (WT) mice to dextran sodium sulfate (DSS)-induced colitis. We assessed proinflammatory cytokine release and mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB) activation in Pirb-/- and WT macrophages following Escherichia coli stimulation. Macrophage transfer experiments were performed to define the role of PIR-B in the negative regulation of macrophage function in DSS-induced colitis. We also assessed expression of PIR-B human homologues (immunoglobulin-like transcript [ILT]-2 and ILT-3) in colon biopsy samples from healthy individuals (controls) and patients with IBD. RESULTS: Pirb-/- mice had increased susceptibility to DSS-induced colitis. In vitro analysis showed increased production of proinflammatory cytokines (interleukin-6, interleukin-1beta, and tumor necrosis factor alpha) and activation of MAPK and NF-kappaB in Pirb-/- macrophages following bacterial activation. Adoptive transfer of bone marrow-derived Pirb-/- macrophages into WT mice was sufficient to increase disease susceptibility. ILT-2 and ILT-3 were expressed on CD68+ and CD68- mononuclear cells and intestinal epithelium in colon biopsy samples from patients and controls. CONCLUSIONS: PIR-B negatively regulates macrophage functions in response to pathogenic bacteria and chronic intestinal inflammatory responses. Inhibitory receptors such as PIR-B might be used as therapeutic targets for treatment of patients with IBD.


Subject(s)
Colitis/immunology , Colon/immunology , Immunity, Innate , Macrophage Activation , Macrophages/immunology , Receptors, Immunologic/metabolism , Adolescent , Adoptive Transfer , Animals , Antigens, CD/analysis , Biopsy , Case-Control Studies , Child , Child, Preschool , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Escherichia coli/pathogenicity , Female , Humans , Inflammation Mediators/metabolism , Leukocyte Immunoglobulin-like Receptor B1 , Macrophages/microbiology , Macrophages/transplantation , Male , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Cell Surface/analysis , Receptors, Immunologic/analysis , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Time Factors
15.
Curr Opin Gastroenterol ; 27(2): 139-45, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21102322

ABSTRACT

PURPOSE OF REVIEW: Production of cyclic guanosine monophosphate (cGMP) by guanylate cyclase is of critical importance to gastrointestinal physiology. Tight regulation of cGMP concentration is necessary for proper intestinal secretion and intestinal epithelial cell proliferative and apoptotic homeostasis. This review focuses on recent work detailing the role of a subset of transmembrane guanylate cyclases in the pathophysiology of intestinal secretory and motility disorders and intestinal epithelial cell transformation. Also considered is the potential for therapeutic manipulation of intestinal guanylate cyclase/cGMP signaling for the correction of chronic constipation and gastrointestinal cancer. RECENT FINDINGS: Recent work in mice and humans suggests a role for transmembrane guanylate cyclases in intestinal fluid secretion as well as hormonal enteric-renal signaling which mediates postprandial natriuresis. Transmembrane guanylate cyclases are also important in gastrointestinal transit rate and motility. Ongoing clinical trials have found that guanylate cyclase activating peptides are safe and effective in the treatment of constipation-predominant irritable bowel syndrome and chronic constipation. In addition, accumulating evidence indicates that membrane-associated guanylate cyclase receptors regulate intestinal epithelial cell homeostatic proliferation and apoptosis as well as gastrointestinal malignancy. The anticancer activity of cGMP signaling in animal studies suggests additional therapeutic applications for guanylate cyclase agonists. SUMMARY: Progress toward understanding gastrointestinal transmembrane guanylate cyclase/cGMP physiology has recently accelerated due to definitive in-vitro studies and work using gene-targeted animal models and has facilitated the development of safe and effective drugs designed to regulate cGMP production in the intestine. Current work should be directed toward a detailed understanding of cGMP effector pathways and the manner in which subcellular concentrations of cGMP regulate them to influence intestinal health and disease.


Subject(s)
Cyclic GMP/metabolism , Gastrointestinal Neoplasms/enzymology , Guanylate Cyclase/physiology , Intestines/physiology , Receptors, Guanylate Cyclase-Coupled/physiology , Animals , Apoptosis , Cell Transformation, Neoplastic , Cyclic GMP/physiology , Gastrointestinal Neoplasms/drug therapy , Guanylate Cyclase/therapeutic use , Humans , Mice
16.
J Thromb Haemost ; 18(1): 91-103, 2020 01.
Article in English | MEDLINE | ID: mdl-31539206

ABSTRACT

BACKGROUND: Protease-activated receptor-1 (PAR-1) plays a major role in multiple disease processes, including colitis. Understanding the mechanisms coupling PAR-1 to disease pathogenesis is complicated by the fact that PAR-1 is broadly expressed across multiple cell types. OBJECTIVE: Determine the specific contributions of PAR-1 expressed by macrophages and colonic enterocytes to infectious colitis. METHODS: Mice carrying a conditional PAR-1 allele were generated and bred to mice expressing Cre recombinase in a myeloid- (PAR-1ΔM ) or enterocyte-specific (PAR-1ΔEPI ) fashion. Citrobacter rodentium colitis pathogenesis was analyzed in mice with global PAR-1 deletion (PAR-1-/- ) and cell type-specific deletions. RESULTS: Constitutive deletion of PAR-1 had no significant impact on weight loss, crypt hypertrophy, crypt abscess formation, or leukocyte infiltration in Citrobacter colitis. However, colonic shortening was significantly blunted in infected PAR-1-/- mice, and these animals exhibited decreased local levels of IL-1ß, IL-22, IL-6, and IL-17A. In contrast, infected PAR-1ΔM mice lost less weight and had fewer crypt abscesses relative to controls. PAR-1ΔM mice had diminished CD3+ T cell infiltration into colonic tissue, but macrophage and CD4+ T cell infiltration were similar to controls. Also contrasting results in global knockouts, PAR-1ΔM mice exhibited lower levels of IL-1ß, but not Th17-related cytokines (ie, IL-22, IL-6, IL-17A). Infected PAR-1ΔEPI mice exhibited increased crypt hypertrophy and crypt abscess formation, but local cytokine elaboration was similar to controls. CONCLUSIONS: These studies reveal complex, cell type-specific roles for PAR-1 in modulating the immune response to Citrobacter colitis that are not readily apparent in analyses limited to mice with global PAR-1 deficiency.


Subject(s)
Colitis , Receptor, PAR-1 , Animals , Citrobacter rodentium , Colitis/genetics , Colitis/microbiology , Enterobacteriaceae Infections , Mice , Mice, Inbred C57BL , Receptor, PAR-1/genetics , Th17 Cells
17.
Cell Signal ; 19(6): 1221-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17317103

ABSTRACT

The extracellular signal regulated kinase (ERK1/2) signaling cascade has been implicated as both a pro-apoptotic and anti-apoptotic pathway depending on cell type and context. In the T84 intestinal epithelial cell line, cAMP activates ERK1/2 resulting in the inhibition of apoptosis. Cyclic-AMP signaling relies on the binding and activation of a cAMP binding protein. In most cell types, the majority of this signaling occurs through an isoform of protein kinase A (PKAI or PKAII). Despite evidence to the contrary, we hypothesized that ERK1/2 activation is through a PKA isoform. Pharmacological activators and inhibitors of PKA as well as siRNA were used to further interrogate this potential signaling pathway. Our results demonstrate that at doses sufficient to increase PKA activity, PKAII specific cAMP analogs activate ERK1/2 while PKAI analogs do not. Pharmacological inhibition of the PKAII regulatory subunit and catalytic subunit as well as siRNA knockdown of the catalytic subunit blocks ERK1/2 activation. We conclude that in the T84 cell line, cAMP binding to the PKAII regulatory subunit leads to the subsequent phosphorylation of ERK1/2 and provides insight into the mechanism of cAMP mediated survival signaling in the intestinal epithelium. These results directly implicate PKAII as a mediator of cell survival in T84 cells and provide evidence for an additional means by which cAMP can influence intestinal cell turnover.


Subject(s)
Cyclic AMP/pharmacology , Intestines/cytology , Intestines/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Catalytic Domain , Cell Line , Cyclic AMP/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isoquinolines/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Models, Biological , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology
18.
Mol Cell Biol ; 25(19): 8444-55, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16166627

ABSTRACT

Loss of glycogen synthase kinase 3beta (GSK-3beta) in mice results in embryonic lethality via hepatocyte apoptosis. Consistent with this result, cells from these mice have diminished nuclear factor kappaB (NF-kappaB) activity, implying a functional role for GSK-3beta in regulating NF-kappaB. Here, we have explored mechanisms by which GSK-3beta may control NF-kappaB function. We show that cytokine-induced IkappaB kinase activity and subsequent phosphorylation of IkappaBalpha, p105, and p65 are not affected by the absence of GSK-3beta activity. Furthermore, nuclear accumulation of p65 following tumor necrosis factor treatment is unaffected by the loss of GSK-3beta. However, NF-kappaB DNA binding activity is reduced in GSK-3beta null cells and in cells treated with a pharmacological inhibitor of GSK-3. Expression of certain NF-kappaB-regulated genes, such as IkappaBalpha and macrophage inflammatory protein 2, is minimally affected by the absence of GSK-3beta. Conversely, we have identified a subset of NF-kappaB-regulated genes, including those for interleukin-6 and monocyte chemoattractant protein 1, that require GSK-3beta for efficient expression. We show that efficient localization of p65 to the promoter regions of the interleukin-6 and monocyte chemoattractant protein 1 genes following tumor necrosis factor alpha treatment requires GSK-3beta. Therefore, GSK-3beta has profound effects on transcription in a gene-specific manner through a mechanism involving control of promoter-specific recruitment of NF-kappaB.


Subject(s)
Glycogen Synthase Kinase 3/physiology , NF-kappa B/metabolism , Transcription, Genetic , Animals , Apoptosis , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Chemokine CCL2/metabolism , Chemokine CXCL2 , Chemokines/metabolism , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA/chemistry , DNA/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta , Immunoprecipitation , Interleukin-6/metabolism , Intestines/cytology , Mice , Promoter Regions, Genetic , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism
19.
Sci Rep ; 8(1): 1521, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367634

ABSTRACT

The guanylate cyclase C (GC-C) receptor regulates electrolyte and water secretion into the gut following activation by the E. coli enterotoxin STa, or by weaker endogenous agonists guanylin and uroguanylin. Our previous work has demonstrated that GC-C plays an important role in controlling initial infection as well as carrying load of non-invasive bacterial pathogens in the gut. Here, we use Salmonella enterica serovar Typhimurium to determine whether GC-C signaling is important in host defense against pathogens that actively invade enterocytes. In vitro studies indicated that GC-C signaling significantly reduces Salmonella invasion into Caco2-BBE monolayers. Relative to controls, GC-C knockout mice develop severe systemic illness following oral Salmonella infection, characterized by disrupted intestinal mucus layer, elevated cytokines and organ CFUs, and reduced animal survival. In Salmonella-infected wildtype mice, oral gavage of GC-C agonist peptide reduced host/pathogen physical interaction and diminished bacterial translocation to mesenteric lymph nodes. These studies suggest that early life susceptibility to STa-secreting enterotoxigenic E. coli may be counter-balanced by a critical role of GC-C in protecting the mucosa from non-STa producing, invasive bacterial pathogens.


Subject(s)
Endocytosis , Enterocytes/enzymology , Enterocytes/microbiology , Receptors, Enterotoxin/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/immunology , Animal Structures/microbiology , Animals , Bacterial Load , Caco-2 Cells , Cytokines/metabolism , Disease Models, Animal , Humans , Mice, Knockout , Salmonella Infections/microbiology , Survival Analysis
20.
Oncoimmunology ; 7(6): e1435250, 2018.
Article in English | MEDLINE | ID: mdl-29872577

ABSTRACT

Activation of the inflammatory transcription factor NF-κB in tumor-associated macrophages (TAMs) is assumed to contribute to tumor promotion. However, whether and how NF-κB drives the antitumor macrophages to become pro-tumorigenic have not been determined in any cancer type yet. Similarly, how TAMs repress CD8+ cytotoxic T lymphocytes (CTLs) remains largely unknown, although their importance in regulatory T (Treg) cell regulation and tumor promotion has been well appreciated. Here, using an endogenous lung cancer model we uncover a direct crosstalk between TAMs and CTLs. TAMs suppress CTLs through the T-cell inhibitory molecule B7x (B7-H4/B7S1) in a cell-cell contact manner, whereas CTLs kill TAMs in a tumor antigen-specific manner. Remarkably, TAMs secrete the known T-cell suppressive cytokine interleukin-10 (IL-10) to activate, but not to repress, CTLs. Notably, one major role of cell-intrinsic NF-κB RelA is to drive TAMs to suppress CTLs for tumor promotion. It induces B7x expression in TAMs directly, and restricts IL-10 expression indirectly by repressing expression of the NF-κB cofactor Bcl3 and subsequent Bcl3/NF-κB1-mediated transcription of IL-10. It also renders TAMs resistant to CTLs by up-regulating anti-apoptotic genes. These studies help understand how immunity is shaped in lung tumorigenesis, and suggest a RelA-targeted immunotherapy for this deadliest cancer.

SELECTION OF CITATIONS
SEARCH DETAIL