Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nature ; 504(7478): 158-62, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24196717

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Células Eritroides/imunologia , Infecções por Escherichia coli/imunologia , Tolerância Imunológica/imunologia , Listeriose/imunologia , Receptores da Transferrina/metabolismo , Animais , Animais Recém-Nascidos , Arginase/genética , Arginase/metabolismo , Suscetibilidade a Doenças/imunologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Eritroides/enzimologia , Escherichia coli/imunologia , Feminino , Sangue Fetal/citologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/genética , Listeria monocytogenes/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(29): 10672-7, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002484

RESUMO

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.


Assuntos
Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfócitos T CD4-Positivos/imunologia , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Inflamação/imunologia , Interleucina-17/biossíntese , Intestinos/patologia , Ligante OX40/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Antígeno CTLA-4/metabolismo , Candida albicans/fisiologia , Diferenciação Celular/imunologia , Proliferação de Células , Humanos , Inflamação/microbiologia , Inflamação/patologia , Intestinos/imunologia , Intestinos/microbiologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
3.
Histochem Cell Biol ; 146(4): 445-55, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27246004

RESUMO

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.


Assuntos
Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Hormônios Gastrointestinais/biossíntese , Intestinos/citologia , Peptídeos Natriuréticos/biossíntese , Animais , Hormônios Gastrointestinais/análise , Hormônios Gastrointestinais/genética , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos , Peptídeos Natriuréticos/análise , Peptídeos Natriuréticos/genética , Transdução de Sinais
4.
Blood ; 123(12): 1938-47, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24501220

RESUMO

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.


Assuntos
Proteína do Grupo de Complementação A da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação A da Anemia de Fanconi/imunologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/deficiência , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/imunologia , Linfócitos T Reguladores/imunologia , Animais , Transplante de Medula Óssea , Citocinas/biossíntese , Anemia de Fanconi/genética , Anemia de Fanconi/imunologia , Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/metabolismo , Quimeras de Transplante
5.
Hepatology ; 59(5): 1830-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24115079

RESUMO

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.


Assuntos
Fígado Gorduroso/etiologia , Interleucina-17/fisiologia , Transdução de Sinais/fisiologia , Animais , Infecções Bacterianas/complicações , Dieta Hiperlipídica , Progressão da Doença , Fígado Gorduroso/microbiologia , Inflamação/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica , Obesidade/complicações , Espécies Reativas de Oxigênio/metabolismo , Receptores de Interleucina-17/fisiologia
6.
J Immunol ; 190(9): 4773-85, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23562811

RESUMO

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.


Assuntos
Quimiocina CCL11/metabolismo , Eosinófilos/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Células Mieloides/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Apoptose/genética , Quimiocina CCL11/genética , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Colite Ulcerativa/patologia , Colo/metabolismo , Colo/patologia , Eosinófilos/patologia , Feminino , Expressão Gênica , Humanos , Inflamação/genética , Inflamação/patologia , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Intestinos/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Fator de Transcrição RelA/genética
7.
Curr Opin Gastroenterol ; 30(1): 1-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24304979

RESUMO

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.


Assuntos
Doenças Inflamatórias Intestinais/enzimologia , Síndrome do Intestino Irritável/enzimologia , Receptores do Fator Natriurético Atrial/fisiologia , Adesinas Bacterianas/fisiologia , Homeostase/fisiologia , Humanos , Absorção Intestinal/fisiologia , Intestinos/microbiologia , Síndrome do Intestino Irritável/tratamento farmacológico , Receptores do Fator Natriurético Atrial/agonistas , Transdução de Sinais/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia
8.
BMC Gastroenterol ; 13: 135, 2013 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-24004613

RESUMO

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.


Assuntos
Colo/imunologia , Infecções por Enterobacteriaceae/imunologia , Mucosa Intestinal/imunologia , Receptores Acoplados a Guanilato Ciclase/imunologia , Receptores de Peptídeos/imunologia , Animais , Apoptose/imunologia , Carga Bacteriana , Translocação Bacteriana/fisiologia , Citrobacter rodentium/fisiologia , Colo/patologia , Infecções por Enterobacteriaceae/genética , Mucosa Intestinal/patologia , Fígado/patologia , Camundongos , Camundongos Knockout , Permeabilidade , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase/genética , Receptores de Peptídeos/genética , Transdução de Sinais/imunologia
9.
J Immunol ; 186(12): 7205-14, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21555532

RESUMO

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.


Assuntos
Guanilato Ciclase/fisiologia , Animais , Doenças do Colo/etiologia , Doenças do Colo/patologia , Hormônios Gastrointestinais/fisiologia , Hormônios Ectópicos/biossíntese , Hormônios Ectópicos/fisiologia , Inflamação/etiologia , Peptídeos e Proteínas de Sinalização Intercelular , Interferon gama/biossíntese , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Peptídeos Natriuréticos/fisiologia , Fator de Necrose Tumoral alfa/biossíntese
10.
J Biol Chem ; 286(15): 13357-69, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21303908

RESUMO

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.


Assuntos
Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Subunidade alfa1 de Receptor de Interleucina-13/metabolismo , Interleucina-13/metabolismo , Enteropatias/metabolismo , Mucosa Intestinal/metabolismo , Animais , Células CACO-2 , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cistos/genética , Cistos/metabolismo , Cistos/patologia , Diarreia/genética , Diarreia/metabolismo , Diarreia/patologia , Fibrose , Humanos , Interleucina-13/genética , Subunidade alfa1 de Receptor de Interleucina-13/genética , Enteropatias/genética , Enteropatias/patologia , Mucosa Intestinal/patologia , Transporte de Íons/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo
11.
Curr Opin Gastroenterol ; 27(2): 139-45, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21102322

RESUMO

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.


Assuntos
GMP Cíclico/metabolismo , Neoplasias Gastrointestinais/enzimologia , Guanilato Ciclase/fisiologia , Intestinos/fisiologia , Receptores Acoplados a Guanilato Ciclase/fisiologia , Animais , Apoptose , Transformação Celular Neoplásica , GMP Cíclico/fisiologia , Neoplasias Gastrointestinais/tratamento farmacológico , Guanilato Ciclase/uso terapêutico , Humanos , Camundongos
12.
J Thromb Haemost ; 18(1): 91-103, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31539206

RESUMO

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.


Assuntos
Colite , Receptor PAR-1 , Animais , Citrobacter rodentium , Colite/genética , Colite/microbiologia , Infecções por Enterobacteriaceae , Camundongos , Camundongos Endogâmicos C57BL , Receptor PAR-1/genética , Células Th17
13.
Cell Signal ; 19(6): 1221-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17317103

RESUMO

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.


Assuntos
AMP Cíclico/farmacologia , Intestinos/citologia , Intestinos/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Domínio Catalítico , Linhagem Celular , AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Isoquinolinas/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Modelos Biológicos , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Sulfonamidas/farmacologia
14.
Mol Cell Biol ; 25(19): 8444-55, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16166627

RESUMO

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.


Assuntos
Quinase 3 da Glicogênio Sintase/fisiologia , NF-kappa B/metabolismo , Transcrição Gênica , Animais , Apoptose , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CXCL2 , Quimiocinas/metabolismo , Cromatina/metabolismo , Imunoprecipitação da Cromatina , DNA/química , DNA/metabolismo , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta , Imunoprecipitação , Interleucina-6/metabolismo , Intestinos/citologia , Camundongos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
15.
Sci Rep ; 8(1): 1521, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367634

RESUMO

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.


Assuntos
Endocitose , Enterócitos/enzimologia , Enterócitos/microbiologia , Receptores de Enterotoxina/metabolismo , Infecções por Salmonella/patologia , Salmonella typhimurium/imunologia , Estruturas Animais/microbiologia , Animais , Carga Bacteriana , Células CACO-2 , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos Knockout , Infecções por Salmonella/microbiologia , Análise de Sobrevida
16.
Oncoimmunology ; 7(6): e1435250, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29872577

RESUMO

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.

17.
PLoS One ; 13(6): e0198434, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29856838

RESUMO

BACKGROUND: Polymeric immunoglobulin receptor (pIgR) transport of secretory immunoglobulin A (SIgA) to mucosal surfaces is thought to promote gut integrity and immunity to Salmonella enterica serovar Typhimurium (S. Typhimurium), an invasive pathogen in mice. To elucidate potential mechanisms, we assessed intestinal barrier function and both oral and systemic S. Typhimurium virulence in pIgR knockout (KO) and wildtype (WT) mice. METHODS: In uninfected animals, we harvested jejunal segments for Ussing chamber analyses of transepithelial resistance (TER); mesenteric lymph nodes (mLN) for bacterial culture; and serum and stool for IgA. Separately, we infected mice either orally or intravenously (IV) with S. Typhimurium to compare colonization, tissue dynamics, and inflammation between KOs and WTs. RESULTS: Uninfected KOs displayed decreased TER and dramatically increased serum IgA and decreased fecal IgA vs. WT; however, KO mLNs yielded fewer bacterial counts. Remarkably, WTs challenged orally with S. Typhimurium exhibited increased splenomegaly, tissue colonization, and pro-inflammatory cytokines vs. pIgR KOs, which showed increased survival following either oral or IV infection. CONCLUSIONS: Absence of pIgR compromises gut integrity but does not exacerbate bacterial translocation nor S. Typhimurium infection. These findings raise the possibility that immune adaptation to increased gut permeability and elevated serum IgA in the setting of SIgA deficiency provides compensatory protection against invasive gut pathogens.


Assuntos
Receptores de Imunoglobulina Polimérica/genética , Salmonelose Animal/patologia , Salmonella enterica/patogenicidade , Administração Oral , Animais , Citocinas/sangue , Fezes/química , Imunoglobulina A/análise , Imunoglobulina A/sangue , Injeções Intravenosas , Intestinos/patologia , Linfonodos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Imunoglobulina Polimérica/deficiência , Salmonelose Animal/microbiologia , Salmonelose Animal/mortalidade , Salmonella enterica/fisiologia , Esplenomegalia/etiologia , Taxa de Sobrevida
18.
Innate Immun ; 23(2): 175-187, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27932520

RESUMO

Sepsis is a life-threatening event predominantly caused by Gram-negative bacteria. Bacterial infection causes a pronounced macrophage (MΦ) and dendritic cell activation that leads to excessive pro-inflammatory cytokine IL-1ß, IL-6 and TNF-α production (cytokine storm), resulting in endotoxic shock. Previous experimental studies have revealed that inhibiting NF-κB signaling ameliorates disease symptoms; however, the contribution of myeloid p65 in endotoxic shock remains elusive. In this study, we demonstrate increased mortality in mice lacking p65 in the myeloid lineage (p65Δmye) compared with wild type mice upon ultra-pure LPS challenge. We show that increased susceptibility to LPS-induced shock was associated with elevated serum level of IL-1ß and IL-6. Mechanistic analyses revealed that LPS-induced pro-inflammatory cytokine production was ameliorated in p65-deficient bone marrow-derived MΦs; however, p65-deficient 'activated' peritoneal MΦs exhibited elevated IL-1ß and IL-6. We show that the elevated pro-inflammatory cytokine secretion was due, in part, to increased accumulation of IL-1ß mRNA and protein in activated inflammatory MΦs. The increased IL-1ß was linked with heightened binding of PU.1 and CCAAT/enhancer binding protein-ß to Il1b and Il6 promoters in activated inflammatory MΦs. Our data provide insight into a role for NF-κB in the negative regulation of pro-inflammatory cytokines in myeloid cells.


Assuntos
Macrófagos Peritoneais/imunologia , Macrófagos/imunologia , Sepse/imunologia , Fator de Transcrição RelA/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células Cultivadas , Feminino , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/metabolismo , Sepse/induzido quimicamente , Transativadores/metabolismo , Fator de Transcrição RelA/genética , Ativação Transcricional
19.
Physiol Rep ; 5(7)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28373409

RESUMO

The etiology and mechanisms for inflammatory bowel disease (IBD) are incompletely known. Determination of new, clinically important mechanisms for intestinal inflammation is imperative for developing effective therapies to treat IBD We sought to define a widespread mechanism for colon mucosal inflammation via the activation of TGF-ß activated Kinase 1 (TAK1), a central regulator of cellular inflammatory actions. Activation of TAK1 and the downstream inflammatory signaling mediators was determined in pediatric patients with ulcerative colitis (UC) or Crohn's disease (CD) as well as in DSS-induced and spontaneous IBD in mice. The role of TAK1 in facilitating intestinal inflammation in murine models of IBD was investigated by using (5Z)-7-Oxozeaenol, a highly selective pharmacological inhibitor of TAK1. We found hyper-activation of TAK1 in patients with UC or CD and in murine models of IBD Pharmacological inhibition of TAK1 prevented loss in body weight, disease activity, microscopic histopathology, infiltration of inflammatory cells in the colon mucosa, and elevated proinflammatory cytokine production in two murine models of IBD We demonstrated that at the early phase of the disease activation of TAK1 is restricted in the epithelial cells. However, at a more advanced stage of the disease, TAK1 activation predominantly occurs in nonepithelial cells, especially in macrophages. These findings elucidate the activation of TAK1 as crucial in promoting intestinal inflammation. Thus, the TAK1 activation pathway may represent a suitable target to design new therapies for treating IBD in humans.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Colo/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Adolescente , Animais , Criança , Pré-Escolar , Colo/patologia , Feminino , Humanos , Doenças Inflamatórias Intestinais/patologia , Interleucina-10/genética , Interleucina-10/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout
20.
JCI Insight ; 2(19)2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28978796

RESUMO

Cystic fibrosis (CF) is a genetic disorder in which epithelium-generated fluid flow from the lung, intestine, and pancreas is impaired due to mutations disrupting CF transmembrane conductance regulator (CFTR) channel function. CF manifestations of the pancreas and lung are present in the vast majority of CF patients, and 15% of CF infants are born with obstructed gut or meconium ileus. However, constipation is a significantly underreported outcome of CF disease, affecting 47% of the CF patients, and management becomes critical in the wake of increasing life span of CF patients. In this study, we unraveled a potentially novel molecular role of a membrane-bound cyclic guanosine monophosphate-synthesizing (cGMP-synthesizing) intestinal enzyme, guanylate cyclase 2C (GCC) that could be targeted to ameliorate CF-associated intestinal fluid deficit. We demonstrated that GCC agonism results in functional rescue of murine F508del/F508del and R117H/R117H Cftr and CFTR mutants in CF patient-derived intestinal spheres. GCC coexpression and activation facilitated processing and ER exit of F508del CFTR and presented a potentially novel rescue modality in the intestine, similar to the CF corrector VX-809. Our findings identify GCC as a biological CFTR corrector and potentiator in the intestine.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Receptores de Enterotoxina/fisiologia , Animais , Fibrose Cística/enzimologia , Fibrose Cística/genética , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Camundongos Mutantes , Mutação , Organoides/metabolismo , Organoides/patologia , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA