Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 11(2): 357-368, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28812548

RESUMO

Interactions between the microbiota and distal gut are important for the maintenance of a healthy intestinal barrier; dysbiosis of intestinal microbial communities has emerged as a likely contributor to diseases that arise at the level of the mucosa. Intraepithelial lymphocytes (IELs) are positioned within the epithelial barrier, and in the small intestine they function to maintain epithelial homeostasis. We hypothesized that colon IELs promote epithelial barrier function through the expression of cytokines in response to interactions with commensal bacteria. Profiling of bacterial 16S ribosomal RNA revealed that candidate bacteria in the order Bacteroidales are sufficient to promote IEL presence in the colon that in turn produce interleukin-6 (IL-6) in a MyD88 (myeloid differentiation primary response 88)-dependent manner. IEL-derived IL-6 is functionally important in the maintenance of the epithelial barrier as IL-6-/- mice were noted to have increased paracellular permeability, decreased claudin-1 expression, and a thinner mucus gel layer, all of which were reversed by transfer of IL-6+/+ IELs, leading to protection of mice in response to Citrobacter rodentium infection. Therefore, we conclude that microbiota provide a homeostatic role for epithelial barrier function through regulation of IEL-derived IL-6.


Assuntos
Bacteroidaceae/fisiologia , Citrobacter rodentium/imunologia , Colo/imunologia , Disbiose/imunologia , Infecções por Enterobacteriaceae/imunologia , Microbioma Gastrointestinal/imunologia , Interleucina-6/metabolismo , Mucosa Intestinal/fisiologia , Linfócitos Intraepiteliais/fisiologia , Animais , Permeabilidade da Membrana Celular/genética , Homeostase , Imunidade Inata , Interleucina-6/genética , Linfócitos Intraepiteliais/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , RNA Ribossômico 16S/genética , Simbiose
2.
Mucosal Immunol ; 11(2): 415-426, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28832026

RESUMO

Barrier dysfunction has been implicated in the pathophysiology of eosinophilic esophagitis (EoE). Transforming growth factor-ß1 (TGF-ß1), a potent pleiotropic molecule, is increased in EoE; however, no study has evaluated its influence on esophageal epithelial barrier. We hypothesized that TGF-ß1 regulates barrier dysfunction in EoE. We aimed to determine the role of TGF-ß1 in the epithelial barrier in models of EoE. To examine the impact of TGF-ß1 on esophageal barrier, immortalized human esophageal epithelial (EPC2-hTERT) cells were exposed to TGF-ß1 during the three-dimensional air-liquid interface (3D-ALI) model in vitro. TGF-ß1 exposure diminished EPC2-hTERT barrier function as measured by transepithelial electrical resistance (TEER) and 3 kDa Fluorescein isothiocyanate dextran paracellular flux (FITC Flux), and hematoxylin and eosin (H&E) assessment revealed prominent cellular separation. In analysis of epithelial barrier molecules, TGF-ß1 led to the specific reduction in expression of the tight-junction molecule, claudin-7 (CLDN7), and this was prevented by TGF-ß-receptor I inhibitor. Short hairpin ribonucleic acid (shRNA)-mediated CLDN7 knockdown diminished epithelial barrier function, whereas CLDN7 overexpression resulted in protection from TGF-ß1-mediated barrier dysfunction. In pediatric EoE biopsies CLDN7 expression was decreased and altered localization was observed with immunofluorescence analysis, and the TGF-ß1 downstream transcription factor, phosphorylated SMAD2/3 (pSMAD2/3), was increased. Our data suggest that TGF-ß1 participates in esophageal epithelial barrier dysfunction through CLDN7 dysregulation.


Assuntos
Claudinas/metabolismo , Esofagite Eosinofílica/imunologia , Eosinófilos/imunologia , Células Epiteliais/fisiologia , Esôfago/patologia , Junções Íntimas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Biópsia , Técnicas de Cultura de Células , Células Cultivadas , Criança , Claudinas/genética , Regulação para Baixo , Impedância Elétrica , Células Epiteliais/patologia , Humanos , RNA Interferente Pequeno/genética
3.
Mucosal Immunol ; 10(5): 1133-1144, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28098246

RESUMO

IL-10 is a potent anti-inflammatory cytokine that inhibits the production of proinflammatory mediators. Signaling by IL-10 occurs through the IL-10 receptor (IL-10R), which is expressed in numerous cell types, including intestinal epithelial cells (IECs), where it is associated with development and maintenance of barrier function. Guided by an unbiased metabolomics screen, we identified tryptophan (Trp) metabolism as a major modifying pathway in interferon-γ (IFNγ)-dominant murine colitis. In parallel, we demonstrated that IFNγ induction of indoleamine 2,3-dioxygenase 1, an enzyme that catalyzes the conversion of Trp to kynurenine (Kyn), induces IL-10R1 expression. Based on these findings, we hypothesized that IL-10R1 expression on IEC is regulated by Trp metabolites. Analysis of the promoter region of IL-10R1 revealed a functional aryl hydrocarbon response element, which is induced by Kyn in luciferase-based IL-10R1 promoter assays. Additionally, this analysis confirmed that IL-10R1 protein levels were increased in response to Kyn in IEC in vitro. Studies using in vitro wounding assays revealed that Kyn accelerates IL-10-dependent wound closure. Finally, reduction of murine dextran sodium sulfate colitis through Kyn administration correlates with colonic IL-10R1 expression. Taken together, these results provide evidence on the importance of IL-10 signaling in intestinal epithelia and implicate AHR in the regulation of IL-10R1 expression in the colon.


Assuntos
Colite/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Subunidade alfa de Receptor de Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Triptofano/metabolismo , Animais , Sulfato de Dextrana , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Subunidade alfa de Receptor de Interleucina-10/genética , Cinurenina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Cicatrização
5.
Mucosal Immunol ; 8(6): 1324-38, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25850656

RESUMO

Central to inflammatory bowel disease (IBD) pathogenesis is loss of mucosal barrier function. Emerging evidence implicates extracellular adenosine signaling in attenuating mucosal inflammation. We hypothesized that adenosine-mediated protection from intestinal barrier dysfunction involves tissue-specific signaling through the A2B adenosine receptor (Adora2b) at the intestinal mucosal surface. To address this hypothesis, we combined pharmacologic studies and studies in mice with global or tissue-specific deletion of the Adora2b receptor. Adora2b(-/-) mice experienced a significantly heightened severity of colitis, associated with a more acute onset of disease and loss of intestinal epithelial barrier function. Comparison of mice with Adora2b deletion on vascular endothelial cells (Adora2b(fl/fl)VeCadCre(+)) or intestinal epithelia (Adora2b(fl/fl)VillinCre(+)) revealed a selective role for epithelial Adora2b signaling in attenuating colonic inflammation. In vitro studies with Adora2b knockdown in intestinal epithelial cultures or pharmacologic studies highlighted Adora2b-driven phosphorylation of vasodilator-stimulated phosphoprotein (VASP) as a specific barrier repair response. Similarly, in vivo studies in genetic mouse models or treatment studies with an Adora2b agonist (BAY 60-6583) recapitulate these findings. Taken together, our results suggest that intestinal epithelial Adora2b signaling provides protection during intestinal inflammation via enhancing mucosal barrier responses.


Assuntos
Colite/patologia , Células Epiteliais/metabolismo , Mucosa Intestinal/patologia , Receptor A2B de Adenosina/metabolismo , Transdução de Sinais , Doença Aguda , Animais , Western Blotting , Colite/metabolismo , Modelos Animais de Doenças , Células Epiteliais/patologia , Citometria de Fluxo , Imunofluorescência , Marcação In Situ das Extremidades Cortadas , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia
6.
Mucosal Immunol ; 7(1): 114-23, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23695513

RESUMO

Pharmacological stabilization of hypoxia-inducible factor (HIF) through prolyl hydroxylase (PHD) inhibition limits mucosal damage associated with models of murine colitis. However, little is known about how PHD inhibitors (PHDi) influence systemic immune function during mucosal inflammation or the relative importance of immunological changes to mucosal protection. We hypothesized that PHDi enhances systemic innate immune responses to colitis-associated bacteremia. Mice with colitis induced by trinitrobenzene sulfonic acid were treated with AKB-4924, a new HIF-1 isoform-predominant PHDi, and clinical, immunological, and biochemical endpoints were assessed. Administration of AKB-4924 led to significantly reduced weight loss and disease activity compared with vehicle controls. Treated groups were pyrexic but did not become subsequently hypothermic. PHDi treatment augmented epithelial barrier function and led to an approximately 50-fold reduction in serum endotoxin during colitis. AKB-4924 also decreased cytokines involved in pyrogenesis and hypothermia, significantly reducing serum levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α while increasing IL-10. Treatment offered no protection against colitis in epithelial-specific HIF-1α-deficient mice, strongly implicating epithelial HIF-1α as the tissue target for AKB-4924-mediated protection. Taken together, these results indicate that inhibition of prolyl hydroxylase with AKB-4924 enhances innate immunity and identifies that the epithelium is a central site of inflammatory protection afforded by PHDi in murine colitis.


Assuntos
Colite/imunologia , Colite/metabolismo , Imunidade Inata/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Animais , Colite/induzido quimicamente , Colite/tratamento farmacológico , Modelos Animais de Doenças , Endotoxemia/tratamento farmacológico , Feminino , Fator 1 Induzível por Hipóxia/agonistas , Subunidade alfa do Fator 1 Induzível por Hipóxia/agonistas , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Permeabilidade/efeitos dos fármacos , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Piridonas/administração & dosagem , Piridonas/farmacologia , Ácido Trinitrobenzenossulfônico/efeitos adversos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
7.
Mucosal Immunol ; 6(6): 1110-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23462909

RESUMO

Antimicrobial peptides are secreted by the intestinal epithelium to defend from microbial threats. The role of human ß defensin-1 (hBD-1) is notable because its gene (beta-defensin 1 (DEFB1)) is constitutively expressed and its antimicrobial activity is potentiated in the low-oxygen environment that characterizes the intestinal mucosa. Hypoxia-inducible factor (HIF) is stabilized even in healthy intestinal mucosa, and we identified that epithelial HIF-1α maintains expression of murine defensins. Extension to a human model revealed that basal HIF-1α is critical for the constitutive expression of hBD-1. Chromatin immunoprecipitation identified HIF-1α binding to a hypoxia response element in the DEFB1 promoter whose importance was confirmed by site-directed mutagenesis. We used 94 human intestinal samples to identify a strong expression correlation between DEFB1 and the canonical HIF-1α target GLUT1. These findings indicate that basal HIF-1α is critical for constitutive expression of enteric DEFB1 and support targeting epithelial HIF for restoration and maintenance of intestinal integrity.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/imunologia , Mucosa Intestinal/imunologia , beta-Defensinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células CACO-2 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Regiões Promotoras Genéticas/genética , Ligação Proteica , Ativação Transcricional , beta-Defensinas/genética
8.
Mucosal Immunol ; 6(4): 762-75, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23187315

RESUMO

Acute lung injury (ALI) is associated with high morbidity and mortality in critically ill patients. At present, the functional contribution of airway mucins to ALI is unknown. We hypothesized that excessive mucus production could be detrimental during lung injury. Initial transcriptional profiling of airway mucins revealed a selective and robust induction of MUC5AC upon cyclic mechanical stretch exposure of pulmonary epithelia (Calu-3). Additional studies confirmed time- and stretch-dose-dependent induction of MUC5AC transcript or protein during cyclic mechanical stretch exposure in vitro or during ventilator-induced lung injury in vivo. Patients suffering from ALI showed a 58-fold increase in MUC5AC protein in their bronchoalveolar lavage. Studies of the MUC5AC promoter implicated nuclear factor κB in Muc5ac induction during ALI. Moreover, mice with gene-targeted deletion of Muc5ac⁻/⁻ experience attenuated lung inflammation and pulmonary edema during injurious ventilation. We observed that neutrophil trafficking into the lungs of Muc5ac⁻/⁻ mice was selectively attenuated. This implicates that endogenous Muc5ac production enhances pulmonary neutrophil trafficking during lung injury. Together, these studies reveal a detrimental role for endogenous Muc5ac production during ALI and suggest pharmacological strategies to dampen mucin production in the treatment of lung injury.


Assuntos
Mucina-5AC/genética , Mucina-5AC/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Animais , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Estresse Mecânico , Transcrição Gênica , Migração Transendotelial e Transepitelial/genética , Migração Transendotelial e Transepitelial/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia
9.
Cell Mol Biol (Noisy-le-grand) ; 49(1): 77-87, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12839339

RESUMO

Epithelial cells which line mucosal surfaces (e.g. lung, intestine) critically function as a semi-permeable barrier to the outside world. Mucosal organs are highly vascular with extensive metabolic demands, and for this reason, are particularly susceptible to diminished blood flow and resultant tissue hypoxia. Recent work from a number of groups have defined the critical molecular and cellular determinants of barrier function in hypoxic/ischemic tissues. Here, we will briefly highlight some of these studies from both a basic and clinical viewpoint and provide a perspective on future work related to tigh tjunction function in mucosal hypoxia.


Assuntos
Hipóxia/metabolismo , Mucosa Intestinal/metabolismo , Rim/metabolismo , Pulmão/metabolismo , Fatores de Transcrição , Junções Aderentes/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mucosa/metabolismo , Proteínas Nucleares/metabolismo
10.
Biochem Soc Trans ; 31(Pt 4): 795-800, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12887308

RESUMO

Among the antimicrobial proteins and peptides of humans is the cationic 55 kDa bactericidal/permeability-increasing protein (BPI), which possesses antibacterial, endotoxin-neutralizing and opsonic activity against Gram-negative bacteria. Although identified originally as an abundant constituent of neutrophil granules, we have recently identified functional expression of BPI by human mucosal epithelia. BPI expression was markedly up-regulated by exposure of epithelia to lipoxins, endogenous anti-inflammatory eicosanoids that are generated in vivo in the context of aspirin treatment (aspirin-triggered lipoxins). Epithelial BPI was found to be surface expressed and fully functional, as measured by antibacterial activity against Salmonella typhimurium as well as lipopolysaccharide (LPS; endotoxin)-neutralizing activity. These results suggest a role for BPI as an effector of epithelial antibacterial activity and as a modulator of epithelial responses to LPS. Both BPI and the lipoxins are currently the subject of intensive biopharmaceutical development, raising the possibility that therapeutic use of BPI or modulation of epithelial BPI expression may be a useful adjunctive therapy for conditions in which epithelial inflammation is associated with Gram-negative infections and/or endotoxin.


Assuntos
Proteínas Sanguíneas/biossíntese , Células Epiteliais/metabolismo , Proteínas de Membrana , Peptídeos Catiônicos Antimicrobianos , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Humanos , Lipoxinas/metabolismo , Modelos Moleculares , Mucosa/citologia , Conformação Proteica
11.
Cell Mol Life Sci ; 59(5): 754-60, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12088276

RESUMO

Epithelial cells which line mucosal surfaces (e.g. lung, intestine) play a central role in the coordination of the inflammatory response. In both the healthy and diseased mucosa, epithelia lie anatomically positioned in close proximity to a number of other cell types, including leukocytes, fibroblasts, smooth muscle cells and vascular endothelia. This complex architecture supports a unique microenvironment for biochemical cell-cell crosstalk. Our previous studies and work by others have elucidated lipid mediator signaling networks emanating from epithelial cell-cell interactive pathways, and have defined a number of targets for development of effective therapeutics. This short review will focus on recently defined pathways of lipid mediator function in the mucosa, particularly with regard to the role of the epithelium.


Assuntos
Comunicação Celular , Células Epiteliais/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Quimiocinas/metabolismo , Ciclo-Oxigenase 2 , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Inflamação/fisiopatologia , Isoenzimas/metabolismo , Leucócitos/metabolismo , Proteínas de Membrana , Estrutura Molecular , Mucosa/citologia , Mucosa/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo
12.
J Immunol ; 166(12): 7543-8, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11390509

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone-binding proteins that regulate transcriptional responses to peroxisome proliferators and structurally diverse fatty acids. PPARs have been implicated in a wide variety of functions, including lipid homeostasis and inflammatory responses. In this study, we examined the expression of PPAR-alpha in response to ambient hypoxia. Initial studies using microarray analysis of intestinal epithelial mRNA revealed that hypoxia rapidly down-regulates PPAR-alpha mRNA and protein in epithelial cells in vitro and in vivo. Subsequent studies revealed that the PPAR-alpha gene bears a DNA consensus motif for the transcription factor hypoxia-inducible factor 1 (HIF-1). EMSA analysis revealed that ambient hypoxia induces HIF-1alpha binding to the HIF-1 consensus domain of PPAR-alpha in parallel to HIF-1 nuclear accumulation, and antisense depletion of HIF-1alpha resulted in a loss of PPAR-alpha down-regulation. The PPAR-alpha ligand pirinixic acid (WY14643) functionally promoted IFN-gamma-induced ICAM-1 expression in normoxic epithelia, and this response was lost in cells pre-exposed to ambient hypoxia. Such results indicate that HIF-1-dependent down-regulation of PPAR-alpha may provide an adaptive response to proinflammatory stimuli during cellular hypoxia. These studies provide unique insight into the regulation of PPAR-alpha expression and, importantly, provide an example of a down-regulatory pathway mediated by HIF-1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Hipóxia/metabolismo , Proteínas Nucleares/fisiologia , Peroxissomos/fisiologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/biossíntese , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/biossíntese , Animais , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Regulação para Baixo/genética , Humanos , Hipóxia/genética , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
13.
Blood ; 97(10): 3251-8, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11342456

RESUMO

During episodes of inflammation, neutrophils (polymorphonuclear leukocytes [PMNs]) encounter subendothelial matrix substrates that may require additional signaling pathways as directives for movement through the extracellular space. Using an in vitro endothelial and epithelial model, inhibitors of phosphoinositide 3-kinase (PI3K) were observed to promote chemoattractant-stimulated migration by as much as 8 +/- 0.3-fold. Subsequent studies indicated that PMNs respond in a similar manner to RGD-containing matrix substrates and that PMN-matrix interactions are potently inhibited by antibodies directed against beta(3)- but not beta(1)-integrin antibodies, and that PI3K inhibitors block beta(3)-integrin dependence. Biochemical analysis of intracellular beta(3)-integrin uncoupling by PI3K inhibitors revealed diminished beta(3)-integrin tyrosine phosphorylation and decreased association with p72(syk). Similarly, the p72(syk) inhibitor piceatannol promoted PMN transmatrix migration, whereas HIV-tat peptide-facilitated loading of peptides corresponding to the beta(3)-integrin cytoplasmic tail identified the functional tyrosine residues for this activity. These data indicate that PI3K-regulated beta(3)-integrin represents a natural "braking" mechanism for PMNs during transit through the extracellular matrix.


Assuntos
Antígenos CD/fisiologia , Quimiotaxia de Leucócito , Matriz Extracelular/fisiologia , Neutrófilos/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Glicoproteínas da Membrana de Plaquetas/fisiologia , Androstadienos/farmacologia , Anticorpos Monoclonais/farmacologia , Antígenos CD/química , Antígenos CD/imunologia , Linhagem Celular , Citoplasma/química , Endotélio Vascular/fisiologia , Inibidores Enzimáticos/farmacologia , Precursores Enzimáticos/antagonistas & inibidores , Precursores Enzimáticos/fisiologia , Epitélio/fisiologia , Humanos , Integrina beta3 , Peptídeos e Proteínas de Sinalização Intracelular , Leucotrieno B4/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Peroxidase/análise , Inibidores de Fosfoinositídeo-3 Quinase , Fosfotirosina/metabolismo , Glicoproteínas da Membrana de Plaquetas/química , Glicoproteínas da Membrana de Plaquetas/imunologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/fisiologia , Quinase Syk , Wortmanina
14.
J Exp Med ; 193(9): 1027-34, 2001 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-11342587

RESUMO

Mucosal organs such as the intestine are supported by a rich and complex underlying vasculature. For this reason, the intestine, and particularly barrier-protective epithelial cells, are susceptible to damage related to diminished blood flow and concomitant tissue hypoxia. We sought to identify compensatory mechanisms that protect epithelial barrier during episodes of intestinal hypoxia. Initial studies examining T84 colonic epithelial cells revealed that barrier function is uniquely resistant to changes elicited by hypoxia. A search for intestinal-specific, barrier-protective factors revealed that the human intestinal trefoil factor (ITF) gene promoter bears a previously unappreciated binding site for hypoxia-inducible factor (HIF)-1. Hypoxia resulted in parallel induction of ITF mRNA and protein. Electrophoretic mobility shift assay analysis using ITF-specific, HIF-1 consensus motifs resulted in a hypoxia-inducible DNA binding activity, and loading cells with antisense oligonucleotides directed against the alpha chain of HIF-1 resulted in a loss of ITF hypoxia inducibility. Moreover, addition of anti-ITF antibody resulted in a loss of barrier function in epithelial cells exposed to hypoxia, and the addition of recombinant human ITF to vascular endothelial cells partially protected endothelial cells from hypoxia-elicited barrier disruption. Extensions of these studies in vivo revealed prominent hypoxia-elicited increases in intestinal permeability in ITF null mice. HIF-1-dependent induction of ITF may provide an adaptive link for maintenance of barrier function during hypoxia.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Substâncias de Crescimento/biossíntese , Mucosa Intestinal/fisiologia , Mucinas , Proteínas Musculares , Neuropeptídeos , Proteínas Nucleares/metabolismo , Fatores de Transcrição , Animais , Células CACO-2 , Hipóxia Celular , Linhagem Celular , Colo/metabolismo , Colo/fisiologia , Proteínas de Ligação a DNA/genética , Cães , Expressão Gênica , Substâncias de Crescimento/genética , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mucosa Intestinal/metabolismo , Camundongos , Proteínas Nucleares/genética , Peptídeos/genética , Fator Trefoil-2 , Fator Trefoil-3
15.
Am J Physiol Gastrointest Liver Physiol ; 280(4): G746-54, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11254502

RESUMO

Neutrophil (PMN) transepithelial migration is a major effector of epithelial defense in inflammatory diseases involving mucosal surfaces. However, major receptor-ligand interactions between epithelial cells and PMN remain incompletely characterized. To better define the molecular events involved in PMN interactions with epithelial cells, we produced a monoclonal antibody called g82 that inhibited PMN transepithelial migration in the physiological basolateral-to-apical direction. The g82 antigen localized to the apical surface of human colonic epithelium and was significantly upregulated under inflammatory conditions. Immunoprecipitation revealed two polypeptides of M(r) 207 and 32 kDa. F(ab')(2) fragments from g82 IgG had no effect on transmigration, suggesting Fc dependence. Further experiments confirmed dependence on the PMN Fc receptor CD32A and that the observed effects were secondary to a failure of PMN to detach from the apical epithelial surface. These Fc-mediated events were epitope specific since binding, isotype-matched antibodies did not affect detachment. These results identify a new mechanism for retention of PMN at the apical epithelial surface following transepithelial migration. This pathway may be important in pathogen clearance and mucosal pathophysiology associated with autoimmunity.


Assuntos
Fragmentos Fc das Imunoglobulinas/fisiologia , Infiltração de Neutrófilos/fisiologia , Neutrófilos/fisiologia , Animais , Soluções Tampão , Adesão Celular/fisiologia , Células Cultivadas , Colo/citologia , Colo/imunologia , Células Epiteliais/fisiologia , Epitélio/fisiologia , Técnica Direta de Fluorescência para Anticorpo , Humanos , Fragmentos Fc das Imunoglobulinas/biossíntese , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Precipitina
16.
J Exp Med ; 192(8): 1197-204, 2000 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-11034610

RESUMO

Aspirin therapy inhibits prostaglandin biosynthesis without directly acting on lipoxygenases, yet via acetylation of cyclooxygenase 2 (COX-2) it leads to bioactive lipoxins (LXs) epimeric at carbon 15 (15-epi-LX, also termed aspirin-triggered LX [ATL]). Here, we report that inflammatory exudates from mice treated with omega-3 polyunsaturated fatty acid and aspirin (ASA) generate a novel array of bioactive lipid signals. Human endothelial cells with upregulated COX-2 treated with ASA converted C20:5 omega-3 to 18R-hydroxyeicosapentaenoic acid (HEPE) and 15R-HEPE. Each was used by polymorphonuclear leukocytes to generate separate classes of novel trihydroxy-containing mediators, including 5-series 15R-LX(5) and 5,12,18R-triHEPE. These new compounds proved to be potent inhibitors of human polymorphonuclear leukocyte transendothelial migration and infiltration in vivo (ATL analogue > 5,12,18R-triHEPE > 18R-HEPE). Acetaminophen and indomethacin also permitted 18R-HEPE and 15R-HEPE generation with recombinant COX-2 as well as omega-5 and omega-9 oxygenations of other fatty acids that act on hematologic cells. These findings establish new transcellular routes for producing arrays of bioactive lipid mediators via COX-2-nonsteroidal antiinflammatory drug-dependent oxygenations and cell-cell interactions that impact microinflammation. The generation of these and related compounds provides a novel mechanism(s) for the therapeutic benefits of omega-3 dietary supplementation, which may be important in inflammation, neoplasia, and vascular diseases.


Assuntos
Aspirina/farmacologia , Endotélio Vascular/fisiologia , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-3/fisiologia , Inflamação/fisiopatologia , Isoenzimas/metabolismo , Neutrófilos/fisiologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Acetaminofen/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular , Células Cultivadas , Ciclo-Oxigenase 2 , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/metabolismo , Ácido Eicosapentaenoico/farmacologia , Humanos , Técnicas In Vitro , Indometacina/farmacologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos , Microcirculação , Microssomos/enzimologia , Receptores do Leucotrieno B4/fisiologia , Proteínas Recombinantes/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais
17.
J Immunol ; 165(9): 5262-8, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11046060

RESUMO

During episodes of inflammation, multiple cell types release adenine nucleotides in the form of ATP, ADP, 5'-AMP, and adenosine. In particular, following activation, polymorphonuclear leukocytes release larger quantities of 5'-AMP. Extracellular 5'-AMP is metabolized to adenosine by surface-expressed 5'-ectonucleotidase (CD73). Adenosine liberated by this process activates surface adenosine A(2B) receptors, results in endothelial junctional reorganization, and promotes barrier function. We hypothesized that adenosine signaling to endothelia provides a paracrine loop for regulated expression of CD73 and enhanced endothelial barrier function. Using an in vitro microvascular endothelial model, we investigated the influence of 5'-AMP; adenosine; and adenosine analogues on CD73 transcription, surface expression, and function. Initial experiments revealed that adenosine and adenosine analogues induce CD73 mRNA (RT-PCR), surface expression (immunoprecipitation of surface biotinylated CD73), and function (HPLC analysis of etheno-AMP conversion to ethenoadenosine) in a time- and concentration-dependent fashion. Subsequent studies revealed that similar exposure conditions increase surface protein through transcriptional induction of CD73. Analysis of DNA-binding activity by EMSA identified a functional role for CD73 cAMP response element and, moreover, indicated that multiple cAMP agonists induce transcriptional activation of functional CD73. Induced CD73 functioned to enhance 5'-AMP-mediated promotion of endothelial barrier (measured as a paracellular flux of 70-kDa FITC-labeled tracer). These results provide an example of transcriptional induction of enzyme (CD73) by enzymatic product (adenosine) and define a paracrine pathway for the regulated expression of vascular endothelial CD73 and barrier function.


Assuntos
5'-Nucleotidase/metabolismo , Adenosina/fisiologia , Permeabilidade Capilar/fisiologia , Endotélio Vascular/enzimologia , Comunicação Parácrina/fisiologia , 5'-Nucleotidase/biossíntese , 5'-Nucleotidase/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Adulto , Células Cultivadas , AMP Cíclico/agonistas , AMP Cíclico/metabolismo , AMP Cíclico/fisiologia , Derme/irrigação sanguínea , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Ativação Enzimática , Humanos , RNA Mensageiro/metabolismo , Receptores Purinérgicos P1/metabolismo , Ativação Transcricional
18.
Proc Natl Acad Sci U S A ; 97(22): 12091-6, 2000 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-11035795

RESUMO

Hypoxia activates a number of gene products through degradation of the transcriptional coactivator cAMP response element binding protein (CREB). Other transcriptional regulators (e.g., beta-catenin and NF-kappa B) are controlled through phosphorylation-targeted proteasomal degradation, and thus, we hypothesized a similar degradative pathway for CREB. Differential display analysis of mRNA derived from hypoxic epithelia revealed a specific and time-dependent repression of protein phosphatase 1 (PP1), a serine phosphatase important in CREB dephosphorylation. Subsequent studies identified a previously unappreciated proteasomal-targeting motif within the primary structure of CREB (DSVTDS), which functions as a substrate for PP1. Ambient hypoxia resulted in temporally sequential CREB serine phosphorylation, ubiquitination, and degradation (in vitro and in vivo). HIV-tat peptide-facilitated loading of intact epithelia with phosphopeptides corresponding to this proteasome targeting motif resulted in inhibition of CREB ubiquitination. Further studies revealed that PP1 inhibitors mimicked hypoxia-induced gene expression, whereas proteasome inhibitors reversed the hypoxic phenotype. Thus, hypoxia establishes conditions that target CREB to proteasomal degradation. These studies may provide unique insight into a general mechanism of transcriptional regulation by hypoxia.


Assuntos
Hipóxia Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA , Células HeLa , Humanos , Camundongos , Modelos Animais , Fosforilação , Complexo de Endopeptidases do Proteassoma , Transcrição Gênica
19.
J Leukoc Biol ; 68(2): 251-9, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10947070

RESUMO

Neutrophil-induced damage to the protective epithelium has been implicated in mucosal disorders associated with hypoxia, and such damage may be initiated by epithelial-derived chemokines. Because chemokines can bind to membrane proteoglycans, we hypothesized that chemokines may associate with epithelial surfaces and activate polymorphonuclear neutrophils (PMN). Epithelial hypoxia (pO2 20 torr) resulted in a time-dependent induction of interleukin-8 (IL-8) mRNA, soluble protein, as well as surface protein. Such surface IL-8 expression was demonstrated to be dependent on heparinase III expression, and extensions of these experiments indicated that hypoxia induces epithelial perlecan expression in parallel with IL-8. Finally, co-incubation of post-hypoxic epithelia with human PMN induced IL-8-dependent expression of the PMN beta2-integrin CD11b/18. These data indicate that chemokines liberated from epithelia may exist in a surface-bound, bioactive form and that hypoxia may regulate proteoglycan expression.


Assuntos
Proteoglicanas de Heparan Sulfato/biossíntese , Interleucina-8/biossíntese , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Ativação de Neutrófilo , Polissacarídeo-Liases/biossíntese , Antígenos CD18/metabolismo , Comunicação Celular , Hipóxia Celular , Linhagem Celular , Técnicas de Cocultura , Humanos
20.
Mol Immunol ; 37(14): 861-8, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11257307

RESUMO

Recent studies have shown that the CD1 family of proteins present various glycolipid antigens to subsets of T cells. CD1d is expressed on human intestinal epithelial cells (IEC) and exists in two biochemical forms: 37-kDa, beta2-microglobulin (beta2m) independent, nonglycosylated, and 47-kDa, beta2m dependent, glycosylated forms. The biosynthetic pathways and the mechanisms of generation of these two biochemically distinct forms of CD1d in human IEC are unknown. Using a human colonic cell line, T84, transfected with CD1d, the biosynthesis of CD1d was investigated. Pulse-chase metabolic labeling studies of T84 transfected with wild type CD1d demonstrated that CD1d was a stable protein over a 4-day chase period. During the first 24 h of the chase, a novel 65-kDa glycoprotein was co-immunoprecipitated with CD1d. Microsequencing of this protein identified the glycoprotein as the alpha and beta subunits of the resident endoplasmic reticulum protein, prolyl-4-hydroxylase (P4H), an enzyme responsible for hydroxyl modification of proline residues. To study if either one or both biochemical forms of CD1d contained hydroxyproline residues, amino acid composition analysis of the 37 and 48 kDa was performed, and demonstrated that only the 37-kDa, but not the 48-kDa form of CD1d, contained hydroxyproline residues. These studies demonstrate that CD1d exhibits a prolonged association with P4H and that the 37-kDa form contains hydroxyproline residues. This suggests that P4H association with CD1d during its biosynthesis results in a novel post-translational modification of CD1d.


Assuntos
Antígenos CD1/biossíntese , Pró-Colágeno-Prolina Dioxigenase/análise , Antígenos CD1/análise , Antígenos CD1d , Humanos , Mucosa Intestinal/enzimologia , Peso Molecular , Processamento de Proteína Pós-Traducional , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...