Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Gastroenterology ; 138(1): 185-96, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19786028

RESUMO

BACKGROUND & AIMS: Necrotizing enterocolitis (NEC), the leading cause of gastrointestinal death from gastrointestinal disease in preterm infants, is characterized by exaggerated TLR4 signaling and decreased enterocyte proliferation through unknown mechanisms. Given the importance of beta-catenin in regulating proliferation of many cell types, we hypothesize that TLR4 impairs enterocyte proliferation in NEC via impaired beta-catenin signaling. METHODS: Enterocyte proliferation was detected in IEC-6 cells or in ileum or colon from wild-type, TLR4-mutant, or TLR4(-/-) mice after induction of NEC or endotoxemia. beta-Catenin signaling was assessed by cell fractionation or immunoconfocal microscopy to detect its nuclear translocation. Activation and inhibition of beta-catenin were achieved via cDNA or small interfering RNA, respectively. TLR4 in the intestinal mucosa was inhibited with adenoviruses expressing dominant-negative TLR4. RESULTS: TLR4 activation significantly impaired enterocyte proliferation in the ileum but not colon in newborn but not adult mice and in IEC-6 enterocytes. beta-Catenin activation reversed these effects in vitro. To determine the mechanisms involved, TLR4 activation phosphorylated the upstream inhibitory kinase GSK3beta, causing beta-catenin degradation. NEC in both mouse and humans was associated with decreased beta-catenin and increased mucosal GSK3beta expression. Strikingly, the inhibition of enterocyte beta-catenin signaling in NEC could be reversed, and enterocyte proliferation restored, through adenoviral-mediated inhibition of TLR4 signaling in the small intestinal mucosa. CONCLUSION: We now report a novel pathway linking TLR4 with inhibition of beta-catenin signaling via GSK3beta activation, leading to reduced enterocyte proliferation in vitro and in vivo. These data provide additional insights into the pathogenesis of diseases of intestinal inflammation such as NEC.


Assuntos
Enterocolite Necrosante/metabolismo , Enterócitos/citologia , Enterócitos/metabolismo , Receptor 4 Toll-Like/metabolismo , beta Catenina/metabolismo , Adenoviridae/genética , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Colo/patologia , Enterocolite Necrosante/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Íleo/patologia , Recém-Nascido , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/genética
2.
Gastroenterology ; 139(3): 904-17, 917.e1-6, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20580721

RESUMO

BACKGROUND & AIMS: Factors that regulate enterocyte apoptosis in necrotizing enterocolitis (NEC) remain incompletely understood, although Toll-like receptor-4 (TLR4) signaling in enterocytes plays a major role. Nucleotide-binding oligomerization domain-2 (NOD2) is an immune receptor that regulates other branches of the immune system, although its effects on TLR4 in enterocytes and its role in NEC remain unknown. We now hypothesize that activation of NOD2 in the newborn intestine inhibits TLR4, and that failure of NOD2 signaling leads to NEC through increased TLR4-mediated enterocyte apoptosis. METHODS: The effects of NOD2 on enterocyte TLR4 signaling and intestinal injury and repair were assessed in enterocytes lacking TLR4 or NOD2, in mice with intestinal-specific wild-type or dominant-negative TLR4 or NOD2, and in mice with NEC. A protein array was performed on NOD2-activated enterocytes to identify novel effector molecules involved. RESULTS: TLR4 activation caused apoptosis in newborn but not adult small intestine or colon, and its intestinal expression was influenced by NOD2. NOD2 activation inhibited TLR4 in enterocytes, but not macrophages, and reversed the effects of TLR4 on intestinal mucosal injury and repair. Protection from TLR4-induced enterocyte apoptosis by NOD2 required a novel pathway linking NOD2 with the apoptosis mediator second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low PI (SMAC-DIABLO), both in vitro and in vivo. Strikingly, activation of NOD2 reduced SMAC-DIABLO expression, attenuated the extent of enterocyte apoptosis, and reduced the severity of NEC. CONCLUSIONS: These findings reveal a novel inhibitory interaction between TLR4 and NOD2 signaling in enterocytes leading to the regulation of enterocyte apoptosis and suggest a therapeutic role for NOD2 in the protection of intestinal diseases such as NEC.


Assuntos
Endotoxemia/metabolismo , Enterocolite Necrosante/metabolismo , Mucosa Intestinal/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Apoptose , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/metabolismo , Linhagem Celular , Movimento Celular , Modelos Animais de Doenças , Endotoxemia/genética , Endotoxemia/patologia , Enterocolite Necrosante/genética , Enterocolite Necrosante/patologia , Enterocolite Necrosante/prevenção & controle , Enterócitos/metabolismo , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/agonistas , Proteína Adaptadora de Sinalização NOD2/deficiência , Proteína Adaptadora de Sinalização NOD2/genética , Análise Serial de Proteínas , Ratos , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Transdução Genética
3.
J Immunol ; 182(1): 636-46, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19109197

RESUMO

Necrotizing enterocolitis (NEC) is a common and often fatal inflammatory disorder affecting preterm infants that develops upon interaction of indigenous bacteria with the premature intestine. We now demonstrate that the developing mouse intestine shows reciprocal patterns of expression of TLR4 and TLR9, the receptor for bacterial DNA (CpG-DNA). Using a novel ultrasound-guided in utero injection system, we administered LPS directly into the stomachs of early and late gestation fetuses to induce TLR4 signaling and demonstrated that TLR4-mediated signaling within the developing intestine follows its expression pattern. Murine and human NEC were associated with increased intestinal TLR4 and decreased TLR9 expression, suggesting that reciprocal TLR4 and TLR9 signaling may occur in the pathogenesis of NEC. Enteral administration of adenovirus expressing mutant TLR4 to neonatal mice reduced the severity of NEC and increased TLR9 expression within the intestine. Activation of TLR9 with CpG-DNA inhibited LPS-mediated TLR4 signaling in enterocytes in a mechanism dependent upon the inhibitory molecule IRAK-M. Strikingly, TLR9 activation with CpG-DNA significantly reduced NEC severity, whereas TLR9-deficient mice exhibited increased NEC severity. Thus, the reciprocal nature of TLR4 and TLR9 signaling within the neonatal intestine plays a role in the development of NEC and provides novel therapeutic approaches to this disease.


Assuntos
Enterocolite Necrosante/imunologia , Enterocolite Necrosante/terapia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/fisiologia , Receptor Toll-Like 9/biossíntese , Receptor Toll-Like 9/fisiologia , Animais , Linhagem Celular , Regulação para Baixo/imunologia , Enterocolite Necrosante/embriologia , Enterocolite Necrosante/metabolismo , Enterócitos/imunologia , Enterócitos/metabolismo , Terapia Genética , Humanos , Recém-Nascido , Mucosa Intestinal/embriologia , Mucosa Intestinal/crescimento & desenvolvimento , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Ratos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/uso terapêutico , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética , Regulação para Cima/imunologia
4.
J Immunol ; 181(12): 8534-8543, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19050272

RESUMO

The pathways that lead to the internalization of pathogens via phagocytosis remain incompletely understood. We now demonstrate a previously unrecognized role for the gap junction protein connexin43 (Cx43) in the regulation of phagocytosis by macrophages and in the host response to bacterial infection of the peritoneal cavity. Primary and cultured macrophages were found to express Cx43, which localized to the phagosome upon the internalization of IgG-opsonized particles. The inhibition of Cx43 using small interfering RNA or by obtaining macrophages from Cx43 heterozygous or knockout mice resulted in significantly impaired phagocytosis, while transfection of Cx43 into Fc-receptor expressing HeLa cells, which do not express endogenous Cx43, conferred the ability of these cells to undergo phagocytosis. Infection of macrophages with adenoviruses expressing wild-type Cx43 restored phagocytic ability in macrophages from Cx43 heterozygous or deficient mice, while infection with viruses that expressed mutant Cx43 had no effect. In understanding the mechanisms involved, Cx43 was required for RhoA-dependent actin cup formation under adherent particles, and transfection with constitutively active RhoA restored a phagocytic phenotype after Cx43 inactivation. Remarkably, mortality was significantly increased in a mouse model of bacterial peritonitis after Cx43 inhibition and in Cx43 heterozygous mice compared with untreated and wild-type counterparts. These findings reveal a novel role for Cx43 in the regulation of phagocytosis and rearrangement of the F-actin cytoskeleton, and they implicate Cx43 in the regulation of the host response to microbial infection.


Assuntos
Conexina 43/fisiologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/mortalidade , Macrófagos/imunologia , Macrófagos/microbiologia , Peritonite/imunologia , Peritonite/mortalidade , Animais , Linhagem Celular , Conexina 43/biossíntese , Conexina 43/deficiência , Conexina 43/genética , Infecções por Escherichia coli/patologia , Feminino , Células HeLa , Humanos , Fígado/citologia , Fígado/embriologia , Fígado/imunologia , Macrófagos/patologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/patologia , Fagossomos/imunologia , Fagossomos/metabolismo , Fagossomos/microbiologia , Análise de Sobrevida
5.
J Leukoc Biol ; 83(3): 493-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18160540

RESUMO

Emerging evidence suggests that the innate immune system, comprised of Toll-like receptors (TLRs) and their associated molecules, plays a pivotal role in the regulation of intestinal inflammation and in the response to invading pathogens. Although TLRs are thought to have predominantly beneficial effects in pathogen recognition and bacterial clearance by leukocytes, their dysregulation and unique signaling effects within intestinal epithelia in the setting of inflammation may have devastating consequences. For instance, activation of TLR4 in enterocytes leads to an inhibition of enterocyte migration and proliferation as well as the induction of enterocyte apoptosis-factors that would be expected to promote intestinal injury while inhibiting intestinal repair. TLR signaling has been shown to be abnormal in several intestinal inflammatory diseases, including Crohn's disease, ulcerative colitis, and necrotizing enterocolitis. This review serves to examine the evidence regarding the patterns of expression and signaling of TLRs in the intestinal mucosa at basal levels and during physiologic stressors to gain insights into the pathogenesis of intestinal inflammation. We conclude that the data reviewed suggest that epithelial TLR signaling-acting in concert with TLR signaling by leukocytes-participates in the development of intestinal inflammation. We further conclude that the evidence reviewed provides a rationale for the development of novel, epithelial-specific, TLR-based agents in the management of diseases of intestinal inflammation.


Assuntos
Inflamação/fisiopatologia , Enteropatias/fisiopatologia , Mucosa Intestinal/fisiopatologia , Receptores Toll-Like/análise , Biomarcadores/análise , Enterócitos/fisiologia , Humanos , Inflamação/diagnóstico , Inflamação/prevenção & controle , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/fisiopatologia , Enteropatias/diagnóstico , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/fisiologia
6.
Mol Med ; 14(9-10): 645-59, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18584047

RESUMO

Diseases of mucosal inflammation represent important causes of morbidity and mortality, and have led to intense research efforts to understand the factors that lead to their development. It is well accepted that a breakdown of the normally impermeant epithelial barrier of the intestine, the lung, and the kidney is associated with the development of inflammatory disease in these organs, yet significant controversy exists as to how this breakdown actually occurs, and how such a breakdown may lead to inflammation. In this regard, much work has focused upon the role of the epithelium as an "innocent bystander," a target of a leukocyte-mediated inflammatory cascade that leads to its destruction in the mucosal inflammatory process. However, recent evidence from a variety of laboratories indicates that the epithelium is not merely a passive component in the steps that lead to mucosal inflammation, but is a central participant in the process. In addressing this controversy, we and others have determined that epithelial cells express Toll-like receptors (TLRs) of the innate immune system, and that activation of TLRs by endogenous and exogenous ligands may play a central role in determining the balance between a state of "mucosal homeostasis," as is required for optimal organ function, and "mucosal injury," leading to mucosal inflammation and barrier breakdown. In particular, activation of TLRs within intestinal epithelial cells leads to the development of cellular injury and impairment in mucosal repair in the pathogenesis of intestinal inflammation, while activation of TLRs in the lung and kidney may participate in the development of pneumonitis and nephritis respectively. Recent work in support of these concepts is extensively reviewed, while essential areas of further study that are required to determine the significance of epithelial TLR signaling during states of health and disease are outlined.


Assuntos
Células Epiteliais/metabolismo , Inflamação/fisiopatologia , Mucosa/imunologia , Mucosa/fisiopatologia , Transdução de Sinais , Receptores Toll-Like/metabolismo , Células Epiteliais/imunologia , Regulação da Expressão Gênica , Humanos , Enteropatias/imunologia , Enteropatias/fisiopatologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/fisiopatologia , Sistema Respiratório/imunologia , Sistema Respiratório/metabolismo , Doenças Respiratórias/imunologia , Doenças Respiratórias/fisiopatologia , Transdução de Sinais/fisiologia
7.
J Leukoc Biol ; 82(5): 1257-65, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17675562

RESUMO

Phagocytosis is the process by which microbial pathogens are engulfed by macrophages and neutrophils and represents the first line of defense against bacterial infection. The importance of phagocytosis for bacterial clearance is of particular relevance to systemic inflammatory diseases, which are associated with the development of hypoxia, yet the precise effects of hypoxia on phagocytosis remain largely unexplored. We now hypothesize that hypoxia inhibits phagocytosis in macrophages and sought to determine the mechanisms involved. Despite our initial prediction, hypoxia significantly increased the phagocytosis rate of particles in vitro by RAW264.7 and primary peritoneal macrophages and increased phagocytosis of labeled bacteria in vivo by hypoxic mice compared with normoxic controls. In understanding the mechanisms involved, hypoxia caused no changes in RhoA-GTPase signaling but increased the phosphorylation of p38-MAPK significantly. Inhibition of p38 reversed the effects of hypoxia on phagocytosis, suggesting a role for p38 in the hypoxic regulation of phagocytosis. Hypoxia also significantly increased the expression of hypoxia-inducible factor-1alpha (HIF-1alpha) in macrophages, which was reversed after p38 inhibition, suggesting a link between p38 activation and HIF-1alpha expression. It is striking that small interfering RNA knockdown of HIF-1alpha reversed the effects of hypoxia on phagocytosis, and overexpression of HIF-1alpha caused a surprising increase in phagocytosis compared with nontransfected controls, demonstrating a specific role for HIF-1alpha in the regulation of phagocytosis. These data indicate that hypoxia enhances phagocytosis in macrophages in a HIF-1alpha-dependent manner and shed light on an important role for HIF-1alpha in host defense.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia , Macrófagos Peritoneais/metabolismo , Fagocitose , Animais , Apoptose , Diferenciação Celular , Células Cultivadas , Escherichia coli/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Fosforilação , RNA Interferente Pequeno/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
8.
J Perinatol ; 22(3): 204-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11948382

RESUMO

OBJECTIVE: To determine whether dexamethasone use increases the risk for Candida sepsis (CS) in very low birth weight premature infants (<1250 g). DESIGN: Retrospective chart review of all infants with a birth weight <1250 g, admitted to the neonatal intensive care unit of the MetroHealth Medical Center, Cleveland, Ohio between January 1, 1996 and December 31, 1999. Infant groups with (n=65) and without (n=229) CS were compared. RESULTS: Two hundred and ninety four infants with a birth weight <1250 g were identified. CS was diagnosed at a median age of 18 days, and 6 of 65 (10%) infants died directly from Candida-related complications. Candida albicans (n=30, 60%) and Candida parapsilosis (n=14, 25%) were the predominant isolates. Use of dexamethasone in infants at risk for chronic lung disease before 14 days of age (p=0.001), duration of antibiotics (p=0.001), and total duration of parenteral nutrition and intralipid (p=0.0001) were all significantly greater in infants who developed CS. Regression analysis showed that duration of antibiotics before the diagnosis of Candida infection (r(2)=0.69, p=0.0002) and duration of dexamethasone (r(2)=0.93, p=0.0002) correlated with Candida infection. Early dexamethasone use was also related to the age at diagnosis of Candida infection (r(2)=0.51, p=0.01). CONCLUSIONS: Dexamethasone therapy and prolonged duration of antibiotics are associated with Candida infection in premature infants.


Assuntos
Candidíase/induzido quimicamente , Dexametasona/efeitos adversos , Glucocorticoides/efeitos adversos , Recém-Nascido de muito Baixo Peso , Sepse/induzido quimicamente , Estudos de Casos e Controles , Dexametasona/uso terapêutico , Glucocorticoides/uso terapêutico , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/prevenção & controle , Pneumopatias/prevenção & controle , Sepse/microbiologia
10.
Dis Model Mech ; 1(2-3): 94-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19048070

RESUMO

Necrotizing enterocolitis (NEC) is the leading cause of death and long-term disability from gastrointestinal disease in preterm infants, and is characterized by acute and chronic intestinal inflammation that may lead to systemic sepsis and multi-system organ failure. NEC typically develops in the preterm infant after the administration of tube feeds, although it may occasionally be seen in full-term babies. Despite extensive clinical experience in the management of patients with NEC, the underlying cellular and molecular mechanisms leading to its development remain incompletely understood. Several animal models have therefore been developed in a variety of species in order to study the pathogenesis of NEC and to develop more effective treatment strategies. This review seeks to examine the pros and cons of animal models that have been developed in the study of NEC over the past 30 years. It will highlight the various strengths and weaknesses of experimental approaches that have been used, and discuss potential directions for the development of such models for the future.


Assuntos
Modelos Animais de Doenças , Enterocolite Necrosante/fisiopatologia , Doenças do Recém-Nascido/fisiopatologia , Animais , Enterocolite Necrosante/terapia , Humanos , Recém-Nascido , Doenças do Recém-Nascido/terapia , Recém-Nascido Prematuro
11.
J Pediatr Surg ; 43(6): 1175-81, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18558203

RESUMO

BACKGROUND: The early signaling events in the development of necrotizing enterocolitis (NEC) remain undefined. We have recently shown that the endotoxin (lipopolysaccharide [LPS]) receptor toll-like receptor 4 (TLR4) on enterocytes is critical in the pathogenesis of experimental NEC. Given that the membrane receptor CD14 is known to facilitate the activation of TLR4, we now hypothesize that endotoxemia induces an early upregulation of CD14 in enterocytes and that this participates in the early intestinal inflammatory response in the development of NEC. METHODS: IEC-6 enterocytes were treated with LPS (50 microg/mL), and the subcellular localization of CD14 and TLR4 was assessed by confocal microscopy. C57/Bl6 or CD14-/- mice were treated with LPS (5 mg/kg), whereas experimental NEC was induced using a combination of gavage formula feeding and intermittent hypoxia. CD14 expression was determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and reverse transcriptase-polymerase chain reaction, and interleukin 6 was quantified by enzyme-linked immunosorbent assay and reverse transcriptase-polymerase chain reaction. RESULTS: Exposure of IEC-6 enterocytes to LPS led to an initial, transient increase in CD14 expression. The early increase in CD14 expression was associated with internalization of CD14 to a perinuclear compartment where increased colocalization with TLR4 was noted. The in vivo significance of these findings is suggested as treatment of mice with LPS led to an early increase in CD14 expression in the intestinal mucosa, whereas the persistent endotoxemia of experimental NEC was associated with decreased CD14 expression within enterocytes. CONCLUSIONS: LPS signaling in the enterocyte is marked by an early, transient increase in expression of CD14 and redistribution of the receptor. This process may contribute to the early activation of the intestinal inflammatory response that is observed in the development of NEC.


Assuntos
Endotoxemia/fisiopatologia , Enterocolite Necrosante/genética , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Endotoxinas , Enterocolite Necrosante/fisiopatologia , Enterócitos/citologia , Enterócitos/metabolismo , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatologia , Receptores de Lipopolissacarídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Probabilidade , Distribuição Aleatória , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Transdução de Sinais , Receptor 4 Toll-Like/genética , Regulação para Cima
12.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G559-69, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18635599

RESUMO

Necrotizing enterocolitis (NEC) is associated with the release of interferon-gamma (IFN) by enterocytes and delayed intestinal restitution. Our laboratory has recently demonstrated that IFN inhibits enterocyte migration by impairing enterocyte gap junctions, intercellular channels that are composed of connexin43 (Cx43) monomers and that are required for enterocyte migration to occur. The mechanisms by which IFN inhibits gap junctions are incompletely understood. Lipid rafts are cholesterol-sphingolipid-rich microdomains of the plasma membrane that play a central role in the trafficking and signaling of various proteins. We now hypothesize that Cx43 is present on enterocyte lipid rafts and that IFN inhibits enterocyte migration by displacing Cx43 from lipid rafts in enterocytes. We now confirm our previous observations that intestinal restitution is impaired in NEC and demonstrate that Cx43 is present on lipid rafts in IEC-6 enterocytes. We show that lipid rafts are required for enterocyte migration, that IFN displaces Cx43 from lipid rafts, and that the phorbol ester phorbol 12-myristate 13-acetate (PMA) restores Cx43 to lipid rafts after treatment with IFN in a protein kinase C-dependent manner. IFN also reversibly decreased the phosphorylation of Cx43 on lipid rafts, which was restored by PMA. Strikingly, restoration of Cx43 to lipid rafts by PMA or by transfection of enterocytes with adenoviruses expressing wild-type Cx43 but not mutant Cx43 is associated with the restoration of enterocyte migration after IFN treatment. Taken together, these findings suggest an important role for lipid raft-Cx43 interactions in the regulation of enterocyte migration during exposure to IFN, such as NEC.


Assuntos
Movimento Celular , Conexina 43/metabolismo , Enterocolite Necrosante/metabolismo , Enterócitos/metabolismo , Junções Comunicantes/metabolismo , Íleo/metabolismo , Interferon gama/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Linhagem Celular , Conexina 43/genética , Modelos Animais de Doenças , Enterocolite Necrosante/patologia , Enterócitos/efeitos dos fármacos , Enterócitos/enzimologia , Enterócitos/patologia , Junções Comunicantes/efeitos dos fármacos , Íleo/patologia , Microdomínios da Membrana/efeitos dos fármacos , Camundongos , Mutação , Fosforilação , Proteína Quinase C/metabolismo , Ratos , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Transfecção
13.
Am J Physiol Gastrointest Liver Physiol ; 294(1): G109-19, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17975131

RESUMO

Enterocytes exist in close association with tissue macrophages, whose activation during inflammatory processes leads to the release of nitric oxide (NO). Repair from mucosal injury requires the migration of enterocytes into the mucosal defect, a process that requires connexin43 (Cx43)-mediated gap junction communication between adjacent enterocytes. Enterocyte migration is inhibited during inflammatory conditions including necrotizing enterocolitis, in part, through impaired gap junction communication. We now hypothesize that activated macrophages inhibit gap junctions of adjacent enterocytes and seek to determine whether NO release from macrophages was involved. Using a coculture system of enterocytes and macrophages, we now demonstrate that "activation" of macrophages with lipopolysaccharide and interferon reduces the phosphorylation of Cx43 in adjacent enterocytes, an event known to inhibit gap junction communication. The effects of macrophages on enterocyte gap junctions could be reversed by treatment of macrophages with the inducible nitric oxide synthase (iNOS) inhibitor l-Lysine omega-acetamidine hydrochloride (l-NIL) and by incubation with macrophages from iNOS(-/-) mice, implicating NO in the process. Activated macrophages also caused a NO-dependent redistribution of connexin43 in adjacent enterocytes from the cell surface to an intracellular location, further suggesting NO release may inhibit gap junction function. Treatment of enterocytes with the NO donor S-nitroso-N-acetylpenicillamine (SNAP) markedly inhibited gap junction communication as determined using single cell microinjection of the gap junction tracer Lucifer yellow. Strikingly, activated macrophages inhibited enterocyte migration into a scraped wound, which was reversed by l-NIL pretreatment. These results implicate enterocyte gap junctions as a target of the NO-mediated effects of macrophages during intestinal inflammation, particularly where enterocyte migration is impaired.


Assuntos
Enterócitos/metabolismo , Junções Comunicantes/metabolismo , Gastroenterite/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Comunicação Parácrina , Animais , Linhagem Celular , Movimento Celular , Técnicas de Cocultura , Conexina 43/metabolismo , Enterócitos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Junções Comunicantes/efeitos dos fármacos , Gastroenterite/enzimologia , Interferons/metabolismo , Lipopolissacarídeos/farmacologia , Lisina/análogos & derivados , Lisina/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Fosforilação , Transporte Proteico , Ratos , S-Nitroso-N-Acetilpenicilamina/farmacologia , Fatores de Tempo
14.
J Immunol ; 179(7): 4808-20, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17878380

RESUMO

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in preterm infants and is characterized by translocation of LPS across the inflamed intestine. We hypothesized that the LPS receptor (TLR4) plays a critical role in NEC development, and we sought to determine the mechanisms involved. We now demonstrate that NEC in mice and humans is associated with increased expression of TLR4 in the intestinal mucosa and that physiological stressors associated with NEC development, namely, exposure to LPS and hypoxia, sensitize the murine intestinal epithelium to LPS through up-regulation of TLR4. In support of a critical role for TLR4 in NEC development, TLR4-mutant C3H/HeJ mice were protected from the development of NEC compared with wild-type C3H/HeOUJ littermates. TLR4 activation in vitro led to increased enterocyte apoptosis and reduced enterocyte migration and proliferation, suggesting a role for TLR4 in intestinal repair. In support of this possibility, increased NEC severity in C3H/HeOUJ mice resulted from increased enterocyte apoptosis and reduced enterocyte restitution and proliferation after mucosal injury compared with mutant mice. TLR4 signaling also led to increased serine phosphorylation of intestinal focal adhesion kinase (FAK). Remarkably, TLR4 coimmunoprecipitated with FAK, and small interfering RNA-mediated FAK inhibition restored enterocyte migration after TLR4 activation, demonstrating that the FAK-TLR4 association regulates intestinal healing. These findings demonstrate a critical role for TLR4 in the development of NEC through effects on enterocyte injury and repair, identify a novel TLR4-FAK association in regulating enterocyte migration, and suggest TLR4/FAK as a therapeutic target in this disease.


Assuntos
Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Mucosa Intestinal/metabolismo , Intestinos/patologia , Receptor 4 Toll-Like/metabolismo , Animais , Apoptose , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular , Endotoxinas/farmacologia , Enterocolite Necrosante/induzido quimicamente , Enterocolite Necrosante/genética , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Intestinos/lesões , Cinética , Receptores de Lipopolissacarídeos/metabolismo , Camundongos , Mutação/genética , Transdução de Sinais , Receptor 4 Toll-Like/genética , Regulação para Cima/efeitos dos fármacos
15.
Cardiol Young ; 12(5): 461-4, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15773449

RESUMO

UNLABELLED: A number of cytokines have been implicated in the pathophysiology of congestive heart failure. Genetic polymorphisms of several cytokine genes are known to result in altered gene expression, enabling us to characterize patients as "high" or "low" producers of specific cytokines. We speculate that the cytokine genotypes for a population of children who underwent heart transplantation for end-stage ventricular failure due to cardiomyopathy or congenital heart disease would be enriched for "high producers" of pro-inflammatory cytokines and "low producers" of anti-inflammatory cytokines. METHODS: Cytokine genotyping was performed for the following cytokines on 94 transplanted children using polymerase chain reaction-sequence specific technique: tumor necrosis factor-alpha (-308), interleukin 10 (-1082, -819, -592), interleukin 6 (-174), transforming growth factor-beta1 (codons 10 & 25), and interferon-gamma (+874). Patients with ventricular failure after transplantation for dilated cardiomyopathy, numbering 37, or for congenital heart disease, numbering 34, were compared to 15 children transplanted for structural disease, such as hypoplastic left heart syndrome, without ventricular failure, and to data from healthy children. An additional 8 children with restrictive or hypertrophic cardiomyopathy were also studied. RESULTS: No differences in genotypic distribution were seen between the groups, and all patients were comparable to genotypic distributions as assessed from published normal data. CONCLUSION: No evidence is found to support the hypothesis that these polymorphisms for cytokine genes influence progression to end-stage heart failure in children undergoing transplantation because of cardiomyopathy or congenital heart disease.


Assuntos
Citocinas/genética , Cardiopatias/genética , Polimorfismo Genético , Criança , Progressão da Doença , Genótipo , Cardiopatias/patologia , Humanos , Interferon gama/genética , Interleucina-10/genética , Interleucina-6/genética , Prognóstico , Fator de Crescimento Transformador beta/genética , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA