Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nature ; 477(7364): 335-9, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21921917

RESUMO

Dysfunction of the intestinal epithelium is believed to result in the excessive translocation of commensal bacteria into the bowel wall that drives chronic mucosal inflammation in Crohn's disease, an incurable inflammatory bowel disease in humans characterized by inflammation of the terminal ileum. In healthy individuals, the intestinal epithelium maintains a physical barrier, established by the tight contact of cells. Moreover, specialized epithelial cells such as Paneth cells and goblet cells provide innate immune defence functions by secreting mucus and antimicrobial peptides, which hamper access and survival of bacteria adjacent to the epithelium. Epithelial cell death is a hallmark of intestinal inflammation and has been discussed as a possible pathogenic mechanism driving Crohn's disease in humans. However, the regulation of epithelial cell death and its role in intestinal homeostasis remain poorly understood. Here we demonstrate a critical role for caspase-8 in regulating necroptosis of intestinal epithelial cells (IECs) and terminal ileitis. Mice with a conditional deletion of caspase-8 in the intestinal epithelium (Casp8(ΔIEC)) spontaneously developed inflammatory lesions in the terminal ileum and were highly susceptible to colitis. Casp8(ΔIEC) mice lacked Paneth cells and showed reduced numbers of goblet cells, indicating dysregulated antimicrobial immune cell functions of the intestinal epithelium. Casp8(ΔIEC) mice showed increased cell death in the Paneth cell area of small intestinal crypts. Epithelial cell death was induced by tumour necrosis factor (TNF)-α, was associated with increased expression of receptor-interacting protein 3 (Rip3; also known as Ripk3) and could be inhibited on blockade of necroptosis. Lastly, we identified high levels of RIP3 in human Paneth cells and increased necroptosis in the terminal ileum of patients with Crohn's disease, suggesting a potential role of necroptosis in the pathogenesis of this disease. Together, our data demonstrate a critical function of caspase-8 in regulating intestinal homeostasis and in protecting IECs from TNF-α-induced necroptotic cell death.


Assuntos
Apoptose , Caspase 8/metabolismo , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Caspase 8/genética , Colite/enzimologia , Colite/imunologia , Colite/metabolismo , Colite/patologia , Doença de Crohn/enzimologia , Doença de Crohn/imunologia , Deleção de Genes , Células Caliciformes/patologia , Humanos , Técnicas In Vitro , Camundongos , Necrose , Celulas de Paneth/enzimologia , Celulas de Paneth/imunologia , Celulas de Paneth/metabolismo , Celulas de Paneth/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(43): E4658-67, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25316791

RESUMO

Tumor progression locus-2 (Tpl2) kinase is a major inflammatory mediator in immune cell types recently found to be genetically associated with inflammatory bowel diseases (IBDs). Here we show that Tpl2 may exert a dominant homeostatic rather than inflammatory function in the intestine mediated specifically by subepithelial intestinal myofibroblasts (IMFs). Mice with complete or IMF-specific Tpl2 ablation are highly susceptible to epithelial injury-induced colitis showing impaired compensatory proliferation in crypts and extensive ulcerations without significant changes in inflammatory responses. Following epithelial injury, IMFs sense innate or inflammatory signals and activate, via Tpl2, the cyclooxygenase-2 (Cox-2)-prostaglandin E2 (PGE2) pathway, which we show here to be essential for the epithelial homeostatic response. Exogenous PGE2 administration rescues mice with complete or IMF-specific Tpl2 ablation from defects in crypt function and susceptibility to colitis. We also show that Tpl2 expression is decreased in IMFs isolated from the inflamed ileum of IBD patients indicating that Tpl2 function in IMFs may be highly relevant to human disease. The IMF-mediated mechanism we propose also involves the IBD-associated genes IL1R1, MAPK1, and the PGE2 receptor-encoding PTGER4. Our results establish a previously unidentified myofibroblast-specific innate pathway that regulates intestinal homeostasis and may underlie IBD susceptibility in humans.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Epitélio/metabolismo , Homeostase , Intestinos/patologia , MAP Quinase Quinase Quinases/metabolismo , Miofibroblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Colite/enzimologia , Colite/imunologia , Colite/patologia , Sulfato de Dextrana , Dinoprostona/administração & dosagem , Dinoprostona/farmacologia , Suscetibilidade a Doenças , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/patologia , Homeostase/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/patologia , Doenças Inflamatórias Intestinais/enzimologia , Doenças Inflamatórias Intestinais/patologia , MAP Quinase Quinase Quinases/deficiência , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Fenótipo , Proteínas Proto-Oncogênicas/deficiência , Transdução de Sinais/efeitos dos fármacos
3.
J Immunol ; 193(7): 3779-91, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25187663

RESUMO

The Citrobacter rodentium model mimics the pathogenesis of infectious colitis and requires sequential contributions from different immune cell populations, including innate lymphoid cells (ILCs) and CD4(+) lymphocytes. In this study, we addressed the role of STAT3 activation in CD4(+) cells during host defense in mice against C. rodentium. In mice with defective STAT3 in CD4(+) cells (Stat3(ΔCD4)), the course of infection was unchanged during the innate lymphoid cell-dependent early phase, but significantly altered during the lymphocyte-dependent later phase. Stat3(ΔCD4) mice exhibited intestinal epithelial barrier defects, including downregulation of antimicrobial peptides, increased systemic distribution of bacteria, and prolonged reduction in the overall burden of C. rodentium infection. Immunomonitoring of lamina propria cells revealed loss of virtually all IL-22-producing CD4(+) lymphocytes, suggesting that STAT3 activation was required for IL-22 production not only in Th17 cells, but also in Th22 cells. Notably, the defective host defense against C. rodentium in Stat3(∆CD4) mice could be fully restored by specific overexpression of IL-22 through a minicircle vector-based technology. Moreover, expression of a constitutive active STAT3 in CD4(+) cells shaped strong intestinal epithelial barrier function in vitro and in vivo through IL-22, and it promoted protection from enteropathogenic bacteria. Thus, our work indicates a critical role of STAT3 activation in Th17 and Th22 cells for control of the IL-22-mediated host defense, and strategies expanding STAT3-activated CD4(+) lymphocytes may be considered as future therapeutic options for improving intestinal barrier function in infectious colitis.


Assuntos
Citrobacter rodentium/imunologia , Colite/imunologia , Infecções por Enterobacteriaceae/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Fator de Transcrição STAT3/imunologia , Células Th17/imunologia , Animais , Colite/genética , Colite/microbiologia , Colite/patologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/patologia , Interleucinas/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Transgênicos , Células Th17/patologia , Interleucina 22
4.
Gut ; 64(4): 601-10, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25379949

RESUMO

OBJECTIVES: Intestinal epithelial cells (IEC) express toll-like receptors (TLR) that facilitate microbial recognition. Stimulation of TLR ligands induces a transient increase in epithelial cell shedding, a mechanism that serves the antibacterial and antiviral host defence of the epithelium and promotes elimination of intracellular pathogens. Although activation of the extrinsic apoptosis pathway has been described during inflammatory shedding, its functional involvement is currently unclear. DESIGN: We investigated the functional involvement of caspase-8 signalling in microbial-induced intestinal cell shedding by injecting Lipopolysaccharide (LPS) to mimic bacterial pathogens and poly(I:C) as a probe for RNA viruses in vivo. RESULTS: TLR stimulation of IEC was associated with a rapid activation of caspase-8 and increased epithelial cell shedding. In mice with an epithelial cell-specific deletion of caspase-8 TLR stimulation caused Rip3-dependent epithelial necroptosis instead of apoptosis. Mortality and tissue damage were more severe in mice in which IECs died by necroptosis than apoptosis. Inhibition of receptor-interacting protein (Rip) kinases rescued the epithelium from TLR-induced gut damage. TLR3-induced necroptosis was directly mediated via TRIF-dependent pathways, independent of Tnf-α and type III interferons, whereas TLR4-induced tissue damage was critically dependent on Tnf-α. CONCLUSIONS: Together, our data demonstrate an essential role for caspase-8 in maintaining the gut barrier in response to mucosal pathogens by permitting inflammatory shedding and preventing necroptosis of infected cells. These data suggest that therapeutic strategies targeting the cell death machinery represent a promising new option for the treatment of inflammatory and infective enteropathies.


Assuntos
Caspase 8/fisiologia , Células Epiteliais/fisiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Camundongos , Transdução de Sinais
5.
Gastroenterology ; 145(6): 1369-79, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24036366

RESUMO

BACKGROUND & AIMS: The intestinal epithelium generates a barrier that protects mammals from potentially harmful intestinal contents, such as pathogenic bacteria. Dysregulation of epithelial cell death has been implicated in barrier dysfunction and in the pathogenesis of intestinal inflammation. We investigated mechanisms of cell-death regulation in the intestinal epithelium of mice. METHODS: Conditional knockout mice (either inducible or permanent) with deletion of cellular FLICE-inhibitory protein (cFlip) or caspase-8 in the intestinal epithelium were analyzed by histology and high-resolution endoscopy. We assessed the effects of cFlip or caspase-8 deficiency on intestinal homeostasis. RESULTS: Expression of cFlip in the intestinal epithelium was required for constitutive activation of caspase-8 under steady-state conditions. Intestinal expression of cFlip was required for development; disruption of the gene encoding cFlip from the intestinal epithelium (cFlip(fl/fl) VillinCre(+) mice) resulted in embryonic lethality. When cFlip was deleted from the intestinal epithelium of adult mice (cFlip(iΔIEC) mice), the animals died within a few days from severe tissue destruction, epithelial cell death, and intestinal inflammation. Death of cFlip-depleted intestinal epithelial cells was regulated extrinsically and required the presence of death receptor ligands, such as tumor necrosis factor-α and CD95 ligand, but was independent of receptor-interacting protein 3. cFlip deficiency was associated with strong up-regulation of caspase-8 and caspase-3 activity and excessive apoptosis in intestinal crypts. CONCLUSIONS: cFlip is required for intestinal tissue homeostasis in mice. It controls the level of activation of caspase-8 to promote survival of intestinal epithelial cells.


Assuntos
Apoptose/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Caspase 8/fisiologia , Homeostase/fisiologia , Imunidade/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/deficiência , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Caspase 3/fisiologia , Sobrevivência Celular/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/fisiologia , Modelos Animais , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
6.
Clin Dev Immunol ; 2012: 278059, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22291845

RESUMO

Genetic polymorphisms of autophagy-related genes have been associated with an increased risk to develop inflammatory bowel disease (IBD). Autophagy is an elementary process participating in several cellular events such as cellular clearance and nonapoptotic programmed cell death. Furthermore, autophagy may be involved in intestinal immune homeostasis due to its participation in the digestion of intracellular pathogens and in antigen presentation. In the present study, the role of autophagy in the intestinal epithelial layer was investigated. The intestinal epithelium is essential to maintain gut homeostasis, and defects within this barrier have been associated with the pathogenesis of IBD. Therefore, mice with intestinal epithelial deletion of Atg7 were generated and investigated in different mouse models. Knockout mice showed reduced size of granules and decreased levels of lysozyme in Paneth cells. However, this was dispensable for gut immune homeostasis and had no effect on susceptibility in mouse models of experimentally induced colitis.


Assuntos
Colite/imunologia , Imunidade Inata , Mucosa Intestinal/imunologia , Proteínas Associadas aos Microtúbulos/deficiência , Celulas de Paneth/patologia , Animais , Autofagia/imunologia , Proteína 7 Relacionada à Autofagia , Biomarcadores/metabolismo , Colite/genética , Colite/patologia , Grânulos Citoplasmáticos/imunologia , Grânulos Citoplasmáticos/patologia , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Homeostase , Imuno-Histoquímica , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Muramidase/genética , Muramidase/metabolismo , Celulas de Paneth/imunologia
7.
Mucosal Immunol ; 12(2): 413-424, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30446727

RESUMO

Intestinal epithelial cells (IECs) constitute an important barrier between host and pathogen. Immune mechanisms that provide protection against gastrointestinal helminths often require IL-4Rα-induced activation of STAT6-regulated genes in IECs. However, it is not known whether STAT6 activation in IECs enhances protective immunity against helminths. Furthermore, the regulation of proliferation and differentiation processes of the intestinal epithelium by IEC-intrinsic STAT6 signaling remains unclear. To address these questions, we generated mice with specific expression of a constitutively active version of STAT6 in IECs. These VillinCre_STAT6vt mice show accumulation of secretory IECs, increased proliferation of IECs and lengthening of the small intestine. They rapidly expelled Nippostrongylus brasiliensis worms even in the absence of T cells. Furthermore, primary infection with Heligmosomoides polygyrus resulted in larval trapping in the submucosa and the fecundity of adult worms was severely impaired. Our results reveal an important IEC-intrinsic role of STAT6-regulated genes for intestinal homeostasis and protective immunity against helminths.


Assuntos
Células Epiteliais/fisiologia , Intestinos/patologia , Nematospiroides dubius/fisiologia , Nippostrongylus/fisiologia , Fator de Transcrição STAT6/metabolismo , Infecções por Strongylida/imunologia , Animais , Secreções Corporais , Diferenciação Celular , Feminino , Homeostase , Interações Hospedeiro-Patógeno , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição STAT6/genética , Transdução de Sinais
8.
Front Immunol ; 10: 2481, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695698

RESUMO

Neutrophils respond to various stimuli by decondensing and releasing nuclear chromatin characterized by citrullinated histones as neutrophil extracellular traps (NETs). This achieves pathogen immobilization or initiation of thrombosis, yet the molecular mechanisms of NET formation remain elusive. Peptidyl arginine deiminase-4 (PAD4) achieves protein citrullination and has been intricately linked to NET formation. Here we show that citrullination represents a major regulator of proteolysis in the course of NET formation. Elevated cytosolic calcium levels trigger both peptidylarginine deiminase-4 (PAD4) and calpain activity in neutrophils resulting in nuclear decondensation typical of NETs. Interestingly, PAD4 relies on proteolysis by calpain to achieve efficient nuclear lamina breakdown and chromatin decondensation. Pharmacological or genetic inhibition of PAD4 and calpain strongly inhibit chromatin decondensation of human and murine neutrophils in response to calcium ionophores as well as the proteolysis of nuclear proteins like lamin B1 and high mobility group box protein 1 (HMGB1). Taken together, the concerted action of PAD4 and calpain induces nuclear decondensation in the course of calcium-mediated NET formation.


Assuntos
Calpaína/imunologia , Citrulinação/imunologia , Armadilhas Extracelulares/imunologia , Neutrófilos/imunologia , Lâmina Nuclear/imunologia , Animais , Calpaína/genética , Citrulinação/genética , Armadilhas Extracelulares/genética , Humanos , Camundongos , Camundongos Knockout , Neutrófilos/citologia , Lâmina Nuclear/genética , Proteína-Arginina Desiminase do Tipo 4/genética , Proteína-Arginina Desiminase do Tipo 4/imunologia
9.
Cell Mol Gastroenterol Hepatol ; 4(1): 33-46, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28560287

RESUMO

The intestinal epithelium can be easily disrupted during gut inflammation as seen in inflammatory bowel disease (IBD), such as ulcerative colitis or Crohn's disease. For a long time, research into the pathophysiology of IBD has been focused on immune cell-mediated mechanisms. Recent evidence, however, suggests that the intestinal epithelium might play a major role in the development and perpetuation of IBD. It is now clear that IBD can be triggered by disturbances in epithelial barrier integrity via dysfunctions in intestinal epithelial cell-intrinsic molecular circuits that control the homeostasis, renewal, and repair of intestinal epithelial cells. The intestinal epithelium in the healthy individual represents a semi-permeable physical barrier shielding the interior of the body from invasions of pathogens on the one hand and allowing selective passage of nutrients on the other hand. However, the intestinal epithelium must be considered much more than a simple physical barrier. Instead, the epithelium is a highly dynamic tissue that responds to a plenitude of signals including the intestinal microbiota and signals from the immune system. This epithelial response to these signals regulates barrier function, the composition of the microbiota, and mucosal immune homeostasis within the lamina propria. The epithelium can thus be regarded as a translator between the microbiota and the immune system and aberrant signal transduction between the epithelium and adjacent immune cells might promote immune dysregulation in IBD. This review summarizes the important cellular and molecular barrier components of the intestinal epithelium and emphasizes the mechanisms leading to barrier dysfunction during intestinal inflammation.

10.
J Exp Med ; 214(6): 1655-1662, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28476895

RESUMO

Cancer cells often acquire capabilities to evade cell death induced by current chemotherapeutic treatment approaches. Caspase-8, a central initiator of death receptor-mediated apoptosis, for example, is frequently inactivated in human cancers via multiple mechanisms such as mutation. Here, we show an approach to overcome cell death resistance in caspase-8-deficient colorectal cancer (CRC) by induction of necroptosis. In both a hereditary and a xenograft mouse model of caspase-8-deficient CRC, second mitochondria-derived activator of caspase (SMAC) mimetic treatment induced massive cell death and led to regression of tumors. We further demonstrate that receptor-interacting protein kinase 3 (RIP3), which is highly expressed in mouse models of CRC and in a subset of human CRC cell lines, is the deciding factor of cancer cell susceptibility to SMAC mimetic-induced necroptosis. Thus, our data implicate that it may be worthwhile to selectively evaluate the efficacy of SMAC mimetic treatment in CRC patients with caspase-8 deficiency in clinical trials for the development of more effective personalized therapy.


Assuntos
Apoptose , Neoplasias Colorretais/patologia , Animais , Caspase 8/metabolismo , Colo/patologia , Neoplasias Colorretais/enzimologia , Enterócitos/metabolismo , Células HT29 , Humanos , Camundongos , Proteínas Mitocondriais/metabolismo , Necrose , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cell Cycle ; 15(21): 2875-2881, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27715398

RESUMO

As an inhibitor of apoptosis (IAP) family member, Survivin is known for its role during regulation of apoptosis. More recently its function as a cell cycle regulator has become evident. Survivin was shown to play a pivotal role during embryonic development and is highly expressed in regenerative tissue as well as in many cancer types. We examined the function of Survivin during mouse intestinal organogenesis and in gut pathophysiology. We found high expression of Survivin in experimentally induced colon cancer in mice but also in colon tumors of humans. Moreover, Survivin was regulated by TGF-ß and was found to be highly expressed during mucosal healing following intestinal inflammation. We identified that expression of Survivin is essential early on in life, as specific deletion of Survivin in Villin expressing cells led to embryonic death around day 12 post coitum. Together with our recent study on the role of Survivin in the gut of adult mice our data demonstrate that Survivin is an essential guardian of embryonic gut development and adult gut homeostasis protecting the epithelium from cell death promoting the proliferation of intestinal stem and progenitor cells.


Assuntos
Proteínas Inibidoras de Apoptose/metabolismo , Intestinos/citologia , Proteínas Repressoras/metabolismo , Nicho de Células-Tronco , Fator de Crescimento Transformador beta/metabolismo , Animais , Desenvolvimento Embrionário , Enterócitos/metabolismo , Feminino , Deleção de Genes , Homeostase , Humanos , Inflamação/metabolismo , Inflamação/patologia , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Masculino , Camundongos , Mitose , Survivina
12.
Cell Rep ; 14(5): 1062-1073, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26832409

RESUMO

A tightly regulated balance of proliferation and cell death of intestinal epithelial cells (IECs) is essential for maintenance of gut homeostasis. Survivin is highly expressed during embryogenesis and in several cancer types, but little is known about its role in adult gut tissue. Here, we show that Survivin is specifically expressed in transit-amplifying cells and Lgr5(+) stem cells. Genetic loss of Survivin in IECs resulted in destruction of intestinal integrity, mucosal inflammation, and death of the animals. Survivin deletion was associated with decreased epithelial proliferation due to defective chromosomal segregation. Moreover, Survivin-deficient animals showed induced phosphorylation of p53 and H2AX and increased levels of cell-intrinsic apoptosis in IECs. Consequently, induced deletion of Survivin in Lgr5(+) stem cells led to cell death. In summary, Survivin is a key regulator of gut tissue integrity by regulating epithelial homeostasis in the stem cell niche.


Assuntos
Células Epiteliais/patologia , Homeostase , Proteínas Inibidoras de Apoptose/deficiência , Intestinos/imunologia , Mitose , Proteínas Repressoras/deficiência , Células-Tronco/patologia , Animais , Morte Celular , Divisão Celular , Sobrevivência Celular , Deleção de Genes , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Intestinos/ultraestrutura , Camundongos , Proteínas Repressoras/metabolismo , Nicho de Células-Tronco , Survivina
13.
PLoS One ; 10(3): e0118401, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25799189

RESUMO

Gastrointestinal infections with EHEC and EPEC are responsible for outbreaks of diarrheal diseases and represent a global health problem. Innate first-line-defense mechanisms such as production of mucus and antimicrobial peptides by intestinal epithelial cells are of utmost importance for host control of gastrointestinal infections. For the first time, we directly demonstrate a critical role for Stat3 activation in intestinal epithelial cells upon infection of mice with Citrobacter rodentium - a murine pathogen that mimics human infections with attaching and effacing Escherichia coli. C. rodentium induced transcription of IL-6 and IL-22 in gut samples of mice and was associated with activation of the transcription factor Stat3 in intestinal epithelial cells. C. rodentium infection induced expression of several antimicrobial peptides such as RegIIIγ and Pla2g2a in the intestine which was critically dependent on Stat3 activation. Consequently, mice with specific deletion of Stat3 in intestinal epithelial cells showed increased susceptibility to C. rodentium infection as indicated by high bacterial load, severe gut inflammation, pronounced intestinal epithelial cell death and dissemination of bacteria to distant organs. Together, our data implicate an essential role for Stat3 activation in intestinal epithelial cells during C. rodentium infection. Stat3 concerts the host response to bacterial infection by controlling bacterial growth and suppression of apoptosis to maintain intestinal epithelial barrier function.


Assuntos
Citrobacter rodentium/imunologia , Colite/prevenção & controle , Colo/imunologia , Infecções por Enterobacteriaceae/prevenção & controle , Células Epiteliais/imunologia , Intestinos/imunologia , Fator de Transcrição STAT3/fisiologia , Animais , Colite/etiologia , Colo/microbiologia , Colo/patologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA