Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 31(12): 2007-2022.e12, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-37967555

RESUMO

Exclusive enteral nutrition (EEN) with fiber-free diets is an effective steroid-sparing treatment to induce clinical remission in children with Crohn's disease (CD). However, the mechanism underlying the beneficial effects of EEN remains obscure. Using a model of microbiota-dependent colitis with the hallmarks of CD, we find that the administration of a fiber-free diet prevents the development of colitis and inhibits intestinal inflammation in colitic animals. Remarkably, fiber-free diet alters the intestinal localization of Mucispirillum schaedleri, a mucus-dwelling pathobiont, which is required for triggering disease. Mechanistically, the absence of dietary fiber reduces nutrient availability and impairs the dissimilatory nitrate reduction to ammonia (DNRA) metabolic pathway of Mucispirillum, leading to its exclusion from the mucus layer and disease remission. Thus, appropriate localization of the specific pathobiont in the mucus layer is critical for disease development, which is disrupted by fiber exclusion. These results suggest strategies to treat CD by targeting the intestinal niche and metabolism of disease-causing microbes.


Assuntos
Colite , Doença de Crohn , Microbiota , Humanos , Criança , Animais , Doença de Crohn/terapia , Dieta , Colite/terapia , Resultado do Tratamento
2.
Cell ; 182(2): 447-462.e14, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32758418

RESUMO

The precise mechanism by which oral infection contributes to the pathogenesis of extra-oral diseases remains unclear. Here, we report that periodontal inflammation exacerbates gut inflammation in vivo. Periodontitis leads to expansion of oral pathobionts, including Klebsiella and Enterobacter species, in the oral cavity. Amassed oral pathobionts are ingested and translocate to the gut, where they activate the inflammasome in colonic mononuclear phagocytes, triggering inflammation. In parallel, periodontitis results in generation of oral pathobiont-reactive Th17 cells in the oral cavity. Oral pathobiont-reactive Th17 cells are imprinted with gut tropism and migrate to the inflamed gut. When in the gut, Th17 cells of oral origin can be activated by translocated oral pathobionts and cause development of colitis, but they are not activated by gut-resident microbes. Thus, oral inflammation, such as periodontitis, exacerbates gut inflammation by supplying the gut with both colitogenic pathobionts and pathogenic T cells.


Assuntos
Colite/patologia , Enterobacter/fisiologia , Microbioma Gastrointestinal , Klebsiella/fisiologia , Boca/microbiologia , Animais , Colite/microbiologia , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Enterobacter/isolamento & purificação , Feminino , Inflamassomos/metabolismo , Interleucina-10/deficiência , Interleucina-10/genética , Interleucina-1beta/metabolismo , Klebsiella/isolamento & purificação , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periodontite/microbiologia , Periodontite/patologia , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo
3.
Nat Med ; 26(4): 608-617, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32066975

RESUMO

The involvement of host immunity in the gut microbiota-mediated colonization resistance to Clostridioides difficile infection (CDI) is incompletely understood. Here, we show that interleukin (IL)-22, induced by colonization of the gut microbiota, is crucial for the prevention of CDI in human microbiota-associated (HMA) mice. IL-22 signaling in HMA mice regulated host glycosylation, which enabled the growth of succinate-consuming bacteria Phascolarctobacterium spp. within the gut microbiome. Phascolarctobacterium reduced the availability of luminal succinate, a crucial metabolite for the growth of C. difficile, and therefore prevented the growth of C. difficile. IL-22-mediated host N-glycosylation is likely impaired in patients with ulcerative colitis (UC) and renders UC-HMA mice more susceptible to CDI. Transplantation of healthy human-derived microbiota or Phascolarctobacterium reduced luminal succinate levels and restored colonization resistance in UC-HMA mice. IL-22-mediated host glycosylation thus fosters the growth of commensal bacteria that compete with C. difficile for the nutritional niche.


Assuntos
Bactérias/crescimento & desenvolvimento , Bactérias/metabolismo , Clostridioides difficile/imunologia , Infecções por Clostridium/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Interleucinas/fisiologia , Animais , Bactérias/efeitos dos fármacos , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/imunologia , Enterocolite Pseudomembranosa/imunologia , Enterocolite Pseudomembranosa/metabolismo , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/prevenção & controle , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Glicosilação/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Interleucinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Veillonellaceae/efeitos dos fármacos , Veillonellaceae/crescimento & desenvolvimento , Veillonellaceae/metabolismo , Interleucina 22
4.
Nat Microbiol ; 5(1): 116-125, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31686025

RESUMO

Metabolic reprogramming is associated with the adaptation of host cells to the disease environment, such as inflammation and cancer. However, little is known about microbial metabolic reprogramming or the role it plays in regulating the fitness of commensal and pathogenic bacteria in the gut. Here, we report that intestinal inflammation reprograms the metabolic pathways of Enterobacteriaceae, such as Escherichia coli LF82, in the gut to adapt to the inflammatory environment. We found that E. coli LF82 shifts its metabolism to catabolize L-serine in the inflamed gut in order to maximize its growth potential. However, L-serine catabolism has a minimal effect on its fitness in the healthy gut. In fact, the absence of genes involved in L-serine utilization reduces the competitive fitness of E. coli LF82 and Citrobacter rodentium only during inflammation. The concentration of luminal L-serine is largely dependent on dietary intake. Accordingly, withholding amino acids from the diet markedly reduces their availability in the gut lumen. Hence, inflammation-induced blooms of E. coli LF82 are significantly blunted when amino acids-particularly L-serine-are removed from the diet. Thus, the ability to catabolize L-serine increases bacterial fitness and provides Enterobacteriaceae with a growth advantage against competitors in the inflamed gut.


Assuntos
Dieta , Enterobacteriaceae/fisiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Serina/metabolismo , Animais , Citrobacter rodentium/genética , Citrobacter rodentium/crescimento & desenvolvimento , Citrobacter rodentium/metabolismo , Citrobacter rodentium/fisiologia , Colite/microbiologia , Colite/patologia , Dieta/efeitos adversos , Enterobacteriaceae/genética , Enterobacteriaceae/crescimento & desenvolvimento , Enterobacteriaceae/metabolismo , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Escherichia coli/fisiologia , Regulação Bacteriana da Expressão Gênica , Mucosa Intestinal/metabolismo , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Interações Microbianas , Serina/deficiência , Organismos Livres de Patógenos Específicos
5.
Sci Rep ; 9(1): 1223, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718924

RESUMO

Macrophages (Mϕs) are known to be major producers of the anti-inflammatory cytokine interleukin-10 (IL-10) in the intestine, thus playing an important role in maintaining gastrointestinal homeostasis. Mϕs that reside in the small intestine (SI) have been previously shown to be regulated by dietary antigens, while colonic Mϕs are regulated by the microbiota. However, the role which resident Mϕs play in SI homeostasis has not yet been fully elucidated. Here, we show that SI Mϕs regulate the integrity of the epithelial barrier via secretion of IL-10. We used an animal model of non-steroidal anti-inflammatory drug (NSAID)-induced SI epithelial injury to show that IL-10 is mainly produced by MHCII+ CD64+ Ly6Clow Mϕs early in injury and that it is involved in the restoration of the epithelial barrier. We found that a lack of IL-10, particularly its secretion by Mϕs, compromised the recovery of SI epithelial barrier. IL-10 production by MHCII+ CD64+ Ly6Clow Mϕs in the SI is not regulated by the gut microbiota, hence depletion of the microbiota did not influence epithelial regeneration in the SI. Collectively, these results highlight the critical role IL-10-producing Mϕs play in recovery from intestinal epithelial injury induced by NSAID.


Assuntos
Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Macrófagos/imunologia , Úlcera Péptica/imunologia , Regeneração/imunologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/toxicidade , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/imunologia , Humanos , Indometacina/administração & dosagem , Indometacina/toxicidade , Injeções Subcutâneas , Interleucina-10/genética , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Úlcera Péptica/induzido quimicamente , Úlcera Péptica/patologia , Permeabilidade , Organismos Livres de Patógenos Específicos
6.
Mucosal Immunol ; 12(3): 632-643, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30742042

RESUMO

Intestinal fibrosis is a severe complication in patients with Crohn's disease (CD). Unfortunately, the trigger leading to the development of intestinal fibrosis in the context of CD remains elusive. Here, we show that colonization by a CD-associated pathobiont adherent-invasive Escherichia coli (AIEC) promotes the development of intestinal fibrosis. Exogenously inoculated AIEC strain LF82 and commensal E. coli HS were gradually eradicated from the intestine in healthy mice. In Salmonella- or dextran sodium sulfate-induced colitis models, AIEC exploited inflammation and stably colonize the gut. Consequently, persistent colonization by AIEC LF82 led to substantial fibrosis. In contrast, commensal E. coli HS was unable to derive a growth advantage from inflammation, thereby failing to colonize the inflamed intestine or promote intestinal fibrosis. AIEC colonization potentiated the expression of the IL-33 receptor ST2 in the intestinal epithelium, which is crucial for the development of intestinal fibrosis. The induction of ST2 by AIEC LF82 was mediated by flagellin, as the ΔfliC mutant failed to induce ST2. These observations provide novel insights into pathobiont-driven intestinal fibrosis and can lead to the development of novel therapeutic approaches for the treatment of intestinal fibrosis in the context of CD that target AIEC and/or its downstream IL-33-ST2 signaling.


Assuntos
Colite/imunologia , Doença de Crohn/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/fisiologia , Flagelina/metabolismo , Mucosa Intestinal/imunologia , Infecções por Salmonella/imunologia , Salmonella/fisiologia , Animais , Células Cultivadas , Colite/induzido quimicamente , Sulfato de Dextrana , Modelos Animais de Doenças , Fibrose , Flagelina/genética , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Transdução de Sinais
7.
Cell Mol Gastroenterol Hepatol ; 2(4): 468-481, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27795980

RESUMO

BACKGROUND & AIMS: Gut dysbiosis is closely involved in the pathogenesis of inflammatory bowel disease (IBD). However, it remains unclear whether IBD-associated gut dysbiosis contributes to disease pathogenesis or is merely secondary to intestinal inflammation. We established a humanized gnotobiotic (hGB) mouse system to assess the functional role of gut dysbiosis associated with 2 types of IBD: Crohn's disease (CD) and ulcerative colitis (UC). METHODS: Germ-free mice were colonized by the gut microbiota isolated from patients with CD and UC, and healthy controls. Microbiome analysis, bacterial functional gene analysis, luminal metabolome analysis, and host gene expression analysis were performed in hGB mice. Moreover, the colitogenic capacity of IBD-associated microbiota was evaluated by colonizing germ-free colitis-prone interleukin 10-deficient mice with dysbiotic patients' microbiota. RESULTS: Although the microbial composition seen in donor patients' microbiota was not completely reproduced in hGB mice, some dysbiotic features of the CD and UC microbiota (eg, decreased diversity, alteration of bacterial metabolic functions) were recapitulated in hGB mice, suggesting that microbial community alterations, characteristic for IBD, can be reproduced in hGB mice. In addition, colonization by the IBD-associated microbiota induced a proinflammatory gene expression profile in the gut that resembles the immunologic signatures found in CD patients. Furthermore, CD microbiota triggered more severe colitis than healthy control microbiota when colonized in germ-free interleukin 10-deficient mice. CONCLUSIONS: Dysbiosis potentially contributes to the pathogenesis of IBD by augmenting host proinflammatory immune responses. Transcript profiling: GSE73882.

8.
Sci Rep ; 6: 27634, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27302484

RESUMO

Intestinal resident macrophages (Mϕs) regulate gastrointestinal homeostasis via production of an anti-inflammatory cytokine interleukin (IL)-10. Although a constant replenishment by circulating monocytes is required to maintain the pool of resident Mϕs in the colonic mucosa, the homeostatic regulation of Mϕ in the small intestine (SI) remains unclear. Here, we demonstrate that direct stimulation by dietary amino acids regulates the homeostasis of intestinal Mϕs in the SI. Mice that received total parenteral nutrition (TPN), which deprives the animals of enteral nutrients, displayed a significant decrease of IL-10-producing Mϕs in the SI, whereas the IL-10-producing CD4(+) T cells remained intact. Likewise, enteral nutrient deprivation selectively affected the monocyte-derived F4/80(+) Mϕ population, but not non-monocytic precursor-derived CD103(+) dendritic cells. Notably, in contrast to colonic Mϕs, the replenishment of SI Mϕs and their IL-10 production were not regulated by the gut microbiota. Rather, SI Mϕs were directly regulated by dietary amino acids. Collectively, our study highlights the diet-dependent, microbiota-independent regulation of IL-10-producing resident Mϕs in the SI.


Assuntos
Interleucina-10/metabolismo , Intestino Delgado/metabolismo , Macrófagos/metabolismo , Mucosa/metabolismo , Aminoácidos/metabolismo , Ração Animal , Animais , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/citologia , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Microbioma Gastrointestinal , Homeostase , Cadeias alfa de Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/microbiologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Mucosa/microbiologia , Receptores CCR2/metabolismo
9.
Intest Res ; 14(2): 127-38, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27175113

RESUMO

The gastrointestinal (GI) tract is colonized by a dense community of commensal microorganisms referred to as the gut microbiota. The gut microbiota and the host have co-evolved, and they engage in a myriad of immunogenic and metabolic interactions. The gut microbiota contributes to the maintenance of host health. However, when healthy microbial structure is perturbed, a condition termed dysbiosis, the altered gut microbiota can trigger the development of various GI diseases including inflammatory bowel disease, colon cancer, celiac disease, and irritable bowel syndrome. There is a growing body of evidence suggesting that multiple intrinsic and extrinsic factors, such as genetic variations, diet, stress, and medication, can dramatically affect the balance of the gut microbiota. Therefore, these factors regulate the development and progression of GI diseases by inducing dysbiosis. Herein, we will review the recent advances in the field, focusing on the mechanisms through which intrinsic and extrinsic factors induce dysbiosis and the role a dysbiotic microbiota plays in the pathogenesis of GI diseases.

10.
J Gastroenterol ; 51(3): 195-205, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26553054

RESUMO

Colonization resistance by the commensal microbiota is a key defense against infectious pathogens in the gastrointestinal tract. The microbiota directly competes with incoming pathogens by occupying the colonization niche, depleting nutrients in the gut lumen as well as indirectly inhibiting the growth of pathogens through activation of host immunity. Enteric pathogens have evolved strategies to cope with microbiota-mediated colonization resistance. Pathogens utilize a wide array of virulence factors to outcompete their commensal rivals in the gut. However, since the expression of virulence factors is costly to maintain and reduces bacterial fitness, pathogens need to regulate their virulence properly in order to maximize their fitness. To this end, most pathogens use environmental cues to regulate their virulence gene expression. Thus, a dynamic regulation of virulence factor expression is a key invasion strategy utilized by enteric pathogens. On the other hand, host immunity selectively targets virulent pathogens in order to counter infection in the gut. The host immune system is generally tolerant of harmless microorganisms, such as the commensal microbiota. Moreover, the host relies on its commensal microbiota to contribute, in concert with its immune system, to the elimination of pathogens. Collectively, regulation of virulence determines the fate of enteric pathogens, from the establishment of infection to the eventual elimination. Here, we will review the dynamics of virulence and its role in infection.


Assuntos
Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Virulência/fisiologia , Meio Ambiente , Trato Gastrointestinal/imunologia , Regulação da Expressão Gênica/fisiologia , Humanos , Imunidade Inata , Mecanotransdução Celular/fisiologia , Interações Microbianas/fisiologia , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
11.
Nat Commun ; 6: 8010, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26269452

RESUMO

Monocytes play a crucial role in antimicrobial host defence, but the mechanisms by which they protect the host during intestinal infection remains poorly understood. Here we show that depletion of CCR2(+) monocytes results in impaired clearance of the intestinal pathogen Citrobacter rodentium. After infection, the de novo recruited CCR2(+) monocytes give rise to CD11c(+)CD11b(+)F4/80(+)CD103(-) intestinal macrophages (MPs) within the lamina propria. Unlike resident intestinal MPs, de novo differentiated MPs are phenotypically pro-inflammatory and produce robust amounts of IL-1ß (interleukin-1ß) through the non-canonical caspase-11 inflammasome. Intestinal MPs from infected mice elicit the activation of RORγt(+) group 3 innate lymphoid cells (ILC3) in an IL-1ß-dependent manner. Deletion of IL-1ß in blood monocytes blunts the production of IL-22 by ILC3 and increases the susceptibility to infection. Collectively, these studies highlight a critical role of de novo differentiated monocyte-derived intestinal MPs in ILC3-mediated host defence against intestinal infection.


Assuntos
Citrobacter rodentium/fisiologia , Infecções por Enterobacteriaceae/microbiologia , Linfócitos/fisiologia , Macrófagos/fisiologia , Monócitos/classificação , Receptores CCR2 , Animais , Infecções por Enterobacteriaceae/imunologia , Imunidade Celular , Imunidade Inata , Camundongos , Camundongos Endogâmicos , Monócitos/metabolismo
12.
Immunity ; 38(6): 1142-53, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23809161

RESUMO

The NLRP3 inflammasome is an important component of the innate immune system. However, its mechanism of activation remains largely unknown. We show that NLRP3 activators including bacterial pore-forming toxins, nigericin, ATP, and particulate matter caused mitochondrial perturbation or the opening of a large membrane pore, but this was not required for NLRP3 activation. Furthermore, reactive oxygen species generation or a change in cell volume was not necessary for NLRP3 activation. Instead, the only common activity induced by all NLRP3 agonists was the permeation of the cell membrane to K⁺ and Na⁺. Notably, reduction of the intracellular K⁺ concentration was sufficient to activate NLRP3, whereas an increase in intracellular Na⁺ modulated but was not strictly required for inflammasome activation. These results provide a unifying model for the activation of the NLRP3 inflammasome in which a drop in cytosolic K⁺ is the common step that is necessary and sufficient for caspase-1 activation.


Assuntos
Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Macrófagos/imunologia , Mitocôndrias/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Caspase 1/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Imunidade Inata , Inflamassomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nigericina/farmacologia , Material Particulado/farmacologia , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
13.
Nat Immunol ; 13(5): 449-56, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22484733

RESUMO

Intestinal phagocytes transport oral antigens and promote immune tolerance, but their role in innate immune responses remains unclear. Here we found that intestinal phagocytes were anergic to ligands for Toll-like receptors (TLRs) or commensals but constitutively expressed the precursor to interleukin 1ß (pro-IL-1ß). After infection with pathogenic Salmonella or Pseudomonas, intestinal phagocytes produced mature IL-1ß through the NLRC4 inflammasome but did not produce tumor necrosis factor (TNF) or IL-6. BALB/c mice deficient in NLRC4 or the IL-1 receptor were highly susceptible to orogastric but not intraperitoneal infection with Salmonella. That enhanced lethality was preceded by impaired expression of endothelial adhesion molecules, lower neutrophil recruitment and poor intestinal pathogen clearance. Thus, NLRC4-dependent production of IL-1ß by intestinal phagocytes represents a specific response that discriminates pathogenic bacteria from commensal bacteria and contributes to host defense in the intestine.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/imunologia , Anergia Clonal , Interações Hospedeiro-Patógeno/imunologia , Interleucina-1beta/metabolismo , Intestinos/imunologia , Intestinos/microbiologia , Fagócitos/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Caspase 1/metabolismo , Flagelina/imunologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interleucina-6/biossíntese , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/patologia , Fagócitos/microbiologia , Pseudomonas/imunologia , Infecções por Pseudomonas/imunologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Salmonella/genética , Salmonella/imunologia , Infecções por Salmonella/genética , Infecções por Salmonella/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
14.
Proc Natl Acad Sci U S A ; 104(1): 139-44, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17101977

RESUMO

Angiotensin II (Ang II) is a peptide hormone that, like many cytokines, acts as a proinflammatory agent and growth factor. After injury to the liver, the hormone assists in tissue repair by stimulating hepatocytes and hepatic stellate cells to synthesize extracellular matrix proteins and secrete secondary cytokines and by stimulating myofibroblasts to proliferate. However, under conditions of chronic liver injury, all of these effects conspire to promote pathologic liver fibrosis. Much of this effect of Ang II results from activation of the proinflammatory NF-kappaB transcription factor in response to stimulation of the type 1 Ang II receptor, a G protein-coupled receptor. Here, we characterize a previously undescribed signaling pathway mediating Ang II-dependent activation of NF-kappaB, which is composed of three principal proteins, CARMA3, Bcl10, and MALT1. Blocking the function of any of these proteins, through the use of either dominant-negative mutants, RNAi, or gene targeting, effectively abolishes Ang II-dependent NF-kappaB activation in hepatocytes. In addition, Bcl10(-/-) mice show defective hepatic cytokine production after Ang II treatment. Evidence also is presented that this pathway activates NF-kappaB through ubiquitination of IKKgamma, the regulatory subunit of the IkappaB kinase complex. These results elucidate a concrete series of molecular events that link ligand activation of the type 1 Ang II receptor to stimulation of the NF-kappaB transcription factor. These findings also uncover a function of the CARMA, Bcl10, and MALT1 proteins in cells outside the immune system.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Angiotensina II/farmacologia , Proteínas Reguladoras de Apoptose/fisiologia , Proteínas Adaptadoras de Sinalização CARD/fisiologia , Caspases/fisiologia , Inflamação/etiologia , NF-kappa B/metabolismo , Proteínas de Neoplasias/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteína 10 de Linfoma CCL de Células B , Hepatócitos/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Camundongos , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Receptor Tipo 1 de Angiotensina/fisiologia , Ubiquitina/metabolismo
15.
Nat Struct Mol Biol ; 11(12): 1192-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15543157

RESUMO

MEK1 and MEK2 are closely related, dual-specificity tyrosine/threonine protein kinases found in the Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway. Approximately 30% of all human cancers have a constitutively activated MAPK pathway, and constitutive activation of MEK1 results in cellular transformation. Here we present the X-ray structures of human MEK1 and MEK2, each determined as a ternary complex with MgATP and an inhibitor to a resolution of 2.4 A and 3.2 A, respectively. The structures reveal that MEK1 and MEK2 each have a unique inhibitor-binding pocket adjacent to the MgATP-binding site. The presence of the potent inhibitor induces several conformational changes in the unphosphorylated MEK1 and MEK2 enzymes that lock them into a closed but catalytically inactive species. Thus, the structures reported here reveal a novel, noncompetitive mechanism for protein kinase inhibition.


Assuntos
Inibidores Enzimáticos/farmacologia , MAP Quinase Quinase 1/química , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/química , MAP Quinase Quinase 2/metabolismo , Sítios de Ligação , Sequência Conservada , Dimerização , Inibidores Enzimáticos/química , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Modelos Moleculares , Estrutura Molecular , Estrutura Quaternária de Proteína , Homologia Estrutural de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...