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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 192(3): 1267-76, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24367025

RESUMO

Cytokines secreted at sites of inflammation impact the onset, progression, and resolution of inflammation. In this article, we investigated potential proresolving mechanisms of IFN-γ in models of inflammatory bowel disease. Guided by initial microarray analysis, in vitro studies revealed that IFN-γ selectively induced the expression of IL-10R1 on intestinal epithelia. Further analysis revealed that IL-10R1 was expressed predominantly on the apical membrane of polarized epithelial cells. Receptor activation functionally induced canonical IL-10 target gene expression in epithelia, concomitant with enhanced barrier restitution. Furthermore, knockdown of IL-10R1 in intestinal epithelial cells results in impaired barrier function in vitro. Colonic tissue isolated from murine colitis revealed that levels of IL-10R1 and suppressor of cytokine signaling 3 were increased in the epithelium and coincided with increased tissue IFN-γ and IL-10 cytokines. In parallel, studies showed that treatment of mice with rIFN-γ was sufficient to drive expression of IL-10R1 in the colonic epithelium. Studies of dextran sodium sulfate colitis in intestinal epithelial-specific IL-10R1-null mice revealed a remarkable increase in disease susceptibility associated with increased intestinal permeability. Together, these results provide novel insight into the crucial and underappreciated role of epithelial IL-10 signaling in the maintenance and restitution of epithelial barrier and of the temporal regulation of these pathways by IFN-γ.


Assuntos
Células Epiteliais/metabolismo , Interferon gama/farmacologia , Subunidade alfa de Receptor de Interleucina-10/biossíntese , Interleucina-10/fisiologia , Mucosa Intestinal/metabolismo , Animais , Linhagem Celular , Polaridade Celular , Colite/induzido quimicamente , Colite/metabolismo , Citocinas/biossíntese , Citocinas/genética , Sulfato de Dextrana/toxicidade , Dextranos/farmacocinética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/ultraestrutura , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Regulação da Expressão Gênica , Humanos , Interferon gama/biossíntese , Interferon gama/genética , Interferon gama/fisiologia , Subunidade alfa de Receptor de Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Proteínas Supressoras da Sinalização de Citocina/genética
2.
FASEB J ; 28(1): 256-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24029533

RESUMO

Hypoxia has been widely implicated in many pathological conditions, including those associated with inflammation and tumorigenesis. A number of recent studies have implicated hypoxia in the control of vasculogenesis and permeability, the basis for which is not fully understood. Here we examine the transcriptional regulation of angiogenesis and permeability by hypoxia in endothelial cells. Guided by a global profiling approach in cultured endothelial cells, these studies revealed the selective induction of human gravin (protein kinase A anchoring protein 12) by hypoxia. Analysis of the cloned gravin promoter identified a functional hypoxia-responsive region including 2 binding sites for hypoxia-inducible factor (HIF). Site-directed mutagenesis identified the most distal HIF-binding site as essential for the induction of gravin by hypoxia. Further studies examining gravin gain and loss of function confirmed strong dependence of gravin in control of microvascular endothelial tube formation, wherein gravin functions as a "braking" system for angiogenesis. Additional studies in confluent endothelia revealed that gravin functionally couples to control endothelial barrier function in response to protein kinase A (PKA) agonists. Taken together, these results demonstrate transcriptional coordination of gravin by HIF-1α and amplified PKA-dependent endothelial responses. These findings provide an important link between hypoxia and metabolic conditions associated with inflammation and angiogenesis.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ciclo Celular/genética , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular , Humanos , Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mutagênese Sítio-Dirigida
3.
J Immunol ; 186(3): 1790-8, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21199896

RESUMO

Numerous studies have revealed that hypoxia and inflammation occur coincidentally in mucosal disorders, such as inflammatory bowel disease. During inflammation, epithelial-expressed hypoxia-inducible factor (HIF) serves an endogenously protective function. In this study, we sought to explore how mucosal immune responses influence HIF-dependent end points. Guided by a screen of relevant inflammatory mediators, we identified IFN-γ as a potent repressor of HIF-dependent transcription in human intestinal epithelial cells. Analysis of HIF levels revealed that HIF-1ß, but not HIF-1α, is selectively repressed by IFN-γ in a JAK-dependent manner. Cloning and functional analysis of the HIF-1ß promoter identified a prominent region for IFN-γ-dependent repression. Further studies revealed that colonic IFN-γ and HIF-1ß levels were inversely correlated in a murine colitis model. Taken together, these studies demonstrated that intestinal epithelial HIF is attenuated by IFN-γ through transcriptional repression of HIF-1ß. These observations are relevant to the pathophysiology of colitis (i.e., that loss of HIF signaling during active inflammation may exacerbate disease pathogenesis).


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/antagonistas & inibidores , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Colite/imunologia , Interferon gama/fisiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Proteínas Repressoras/fisiologia , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto/fisiologia , Células CACO-2 , Linhagem Celular Tumoral , Células Cultivadas , Clonagem Molecular , Colite/enzimologia , Colite/patologia , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Feminino , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Mucosa Intestinal/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Pró-Colágeno-Prolina Dioxigenase/fisiologia , Transdução de Sinais/imunologia
4.
J Immunol ; 186(11): 6505-14, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21515785

RESUMO

Tissues of the mucosa are lined by an epithelium that provides barrier and transport functions. It is now appreciated that inflammatory responses in inflammatory bowel diseases are accompanied by striking shifts in tissue metabolism. In this paper, we examined global metabolic consequences of mucosal inflammation using both in vitro and in vivo models of disease. Initial analysis of the metabolic signature elicited by inflammation in epithelial models and in colonic tissue isolated from murine colitis demonstrated that levels of specific metabolites associated with cellular methylation reactions are significantly altered by model inflammatory systems. Furthermore, expression of enzymes central to all cellular methylation, S-adenosylmethionine synthetase and S-adenosylhomocysteine hydrolase, are increased in response to inflammation. Subsequent studies showed that DNA methylation is substantially increased during inflammation and that epithelial NF-κB activity is significantly inhibited following treatment with a reversible S-adenosylhomocysteine hydrolase inhibitor, DZ2002. Finally, these studies demonstrated that inhibition of cellular methylation in a murine model of colitis results in disease exacerbation while folate supplementation to promote methylation partially ameliorates the severity of murine colitis. Taken together, these results identify a global change in methylation, which during inflammation, translates to an overall protective role in mucosal epithelia.


Assuntos
Colite/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Metabolômica/métodos , Adenina/análogos & derivados , Adenina/farmacologia , Adenosil-Homocisteinase/genética , Adenosil-Homocisteinase/metabolismo , Animais , Western Blotting , Butiratos/farmacologia , Linhagem Celular Tumoral , Colite/genética , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Metilação de DNA/efeitos dos fármacos , Sulfato de Dextrana/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica/métodos , Células HeLa , Humanos , Inflamação/genética , Interferon gama/metabolismo , Interferon gama/farmacologia , Mucosa Intestinal/patologia , Espectroscopia de Ressonância Magnética , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mucosite/genética , Mucosite/metabolismo , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Proc Natl Acad Sci U S A ; 107(32): 14298-303, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660763

RESUMO

Resolvin-E1 (RvE1) has been demonstrated to promote inflammatory resolution in numerous disease models. Given the importance of epithelial cells to coordination of mucosal inflammation, we hypothesized that RvE1 elicits an epithelial resolution signature. Initial studies revealed that the RvE1-receptor (ChemR23) is expressed on intestinal epithelial cells (IECs) and that microarray profiling of cells exposed to RvE1 revealed regulation of inflammatory response gene expression. Notably, RvE1 induced intestinal alkaline phosphatase (ALPI) expression and significantly enhanced epithelial ALPI enzyme activity. One role recently attributed to ALPI is the detoxification of bacterial LPS. In our studies, RvE1-exposed epithelia detoxified LPS (assessed by attenuation of NF-kappaB signaling). Furthermore, in epithelial-bacterial interaction assays, we determined that ALPI retarded the growth of Escherichia coli. To define these features in vivo, we used a murine dextran sulfate sodium (DSS) model of colitis. Compared with vehicle controls, administration of RvE1 resulted in significant improvement of disease activity indices (e.g., body weight, colon length) concomitant with increased ALPI expression in the intestinal epithelium. Moreover, inhibition of ALPI activity resulted in increased severity of colitis in DSS-treated animals and partially abrogated the protective influence of RvE1. Together, these data implicate a previously unappreciated role for ALPI in RvE1-mediated inflammatory resolution.


Assuntos
Fosfatase Alcalina/genética , Ácido Eicosapentaenoico/análogos & derivados , Inflamação/prevenção & controle , Mucosa Intestinal/enzimologia , Lipopolissacarídeos/antagonistas & inibidores , Animais , Colite/prevenção & controle , Ácido Eicosapentaenoico/farmacologia , Células Epiteliais/química , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/análise , Ativação Transcricional
6.
FASEB J ; 25(6): 1856-64, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21350119

RESUMO

In intact mucosal tissues, epithelial cells are anatomically positioned in proximity to a number of subepithelial cell types, including endothelia. A number of recent studies have suggested that imbalances between energy supply and demand can result in "inflammatory hypoxia." Given these associations, we hypothesized that endothelial-derived, hypoxia-inducible mediators might influence epithelial function. Guided by cDNA microarray analysis of human microvascular endothelial cells (HMEC-1 line) subjected to hypoxia (pO(2) 20 torr, 8 h), we identified adrenomedullin (ADM) as a prominent hypoxia-inducible factor (HIF) that acts on epithelial cells through cell surface receptors. We assessed the functional ability for exogenous ADM to signal in human intestinal Caco2 cells in vitro by demonstrating a dose-dependent induction of Erk1/2phosphorylation. Further analysis revealed that ADM deneddylates cullin-2 (Cul2), whose action has been demonstrated to control the activity of HIF. Caco2 cells stably expressing a hypoxic response element (HRE)-driven luciferase promoter confirmed that ADM activates the HIF signaling pathway. Extensions of these studies revealed an increase in canonical HIF-1-dependent genes following stimulation with ADM. To define physiological relevance, we investigated the effect of ADM in a DSS model of murine colitis. Administration of ADM resulted in reduced inflammatory indices and less severe histological inflammation compared to vehicle controls. Analysis of tissue and serum cytokines showed a marked and significant inhibition of colitis-associated TNF-α, IL-1ß, and KC. Analysis of circulating ADM demonstrated an increase in serum ADM in murine models of colitis. Taken together, these results identify ADM as an endogenously generated vascular mediator that functions as a mucosal protective factor through fine tuning of HIF activity.


Assuntos
Adrenomedulina/fisiologia , Células Endoteliais/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Adrenomedulina/metabolismo , Adrenomedulina/farmacologia , Animais , Células CACO-2 , Linhagem Celular , Colite/induzido quimicamente , Colite/tratamento farmacológico , Sulfato de Dextrana/toxicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Fator 1 Induzível por Hipóxia/genética , Mucosa Intestinal/citologia , Camundongos , Camundongos Endogâmicos C57BL , Consumo de Oxigênio , Receptores de Adrenomedulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
7.
J Immunol ; 184(9): 5271-9, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20348420

RESUMO

Sepsis and septic acute lung injury are among the leading causes for morbidity and mortality of critical illness. Extracellular adenosine is a signaling molecule implicated in the cellular adaptation to hypoxia, ischemia, or inflammation. Therefore, we pursued the role of the A2B adenosine receptor (AR) as potential therapeutic target in endotoxin-induced acute lung injury. We gained initial insight from in vitro studies of cultured endothelia or epithelia exposed to inflammatory mediators showing time-dependent induction of the A2BAR (up to 12.9 + or - 3.4-fold, p < 0.05). Similarly, murine studies of endotoxin-induced lung injury identified an almost 4.6-fold induction of A2BAR transcript and corresponding protein induction with LPS exposure. Studies utilizing A2BAR promoter constructs and RNA protection assays indicated that A2BAR induction involved mRNA stability. Functional studies of LPS-induced lung injury revealed that pharmacological inhibition or genetic deletion of the A2BAR was associated with dramatic increases in lung inflammation and histologic tissue injury. Studies of A2BAR bone marrow chimeric mice suggested pulmonary A2BAR signaling in lung protection. Finally, studies with a specific A2BAR agonist (BAY 60-6583) demonstrated attenuation of lung inflammation and pulmonary edema in wild-type but not in gene-targeted mice for the A2BAR. These studies suggest the A2BAR as potential therapeutic target in the treatment of endotoxin-induced forms of acute lung injury.


Assuntos
Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/terapia , Mediadores da Inflamação/fisiologia , Lipopolissacarídeos/administração & dosagem , Receptor A2B de Adenosina/fisiologia , Transdução de Sinais/imunologia , Acetamidas/administração & dosagem , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Agonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Administração por Inalação , Aminopiridinas/administração & dosagem , Animais , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Linhagem Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/antagonistas & inibidores , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Purinas/administração & dosagem , Receptor A2B de Adenosina/deficiência , Sepse/genética , Sepse/imunologia , Sepse/terapia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
8.
J Immunol ; 182(8): 4957-64, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19342675

RESUMO

Inflammatory diseases influence tissue metabolism, significantly altering the profile of extracellular adenine nucleotides. A number of studies have suggested that adenosine (Ado) may function as an endogenously generated anti-inflammatory molecule. Given the central role of intestinal epithelial cells to the development of colitis, we hypothesized that specific Ado receptors would contribute to disease resolution in mucosal inflammation as modeled by dextran sodium sulfate (DSS) colitis. Initial profiling studies revealed that murine intestinal epithelial cells express predominantly the Ado A2B receptor (AA2BR) and to a lesser extent AA2AR. Guided by these results, we examined the contribution of AA2BR to colitis. Initial studies indicated that the severity of colitis was increased in Aa2br(-/-) mice relative to Aa2br(+/+) controls, as reflected by increased weight loss, colonic shortening, and disease activity indices. Likewise, enteral administration of the selective AA2BR inhibitor PSB1115 to Aa2br(+/+) mice resulted in a similar increase in severity of DSS colitis. Cytokine profiling of colonic tissue revealed specific deficiencies in IL-10 in Aa2br(-/-) mice relative to controls. Extensions of these findings in cultured human intestinal epithelial cells revealed that stable Ado analogs induce IL-10 mRNA and protein and that such increases can be blocked with PSB1115. Taken together, these studies indicate a central regulatory role for AA2BR-modulated IL-10 in the acute inflammatory phase of DSS colitis, thereby implicating AA2BR as an endogenously protective molecule expressed on intestinal epithelial cells.


Assuntos
Colite/metabolismo , Receptor A2B de Adenosina/metabolismo , Animais , Células CACO-2 , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptor A2B de Adenosina/deficiência , Receptor A2B de Adenosina/genética
9.
FASEB J ; 23(5): 1338-46, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19103643

RESUMO

Because of localized vascular damage and increased tissue oxygen demand, wound healing occurs in a relatively hypoxic microenvironment. These features are particularly relevant to wound healing and fibrosis in chronic inflammatory conditions, such as Crohn's disease and ulcerative colitis. In these studies, we sought to identify the contribution of hypoxia to mechanisms of wound repair in a model of the intestinal submucosa. Initial studies revealed that hypoxia promotes wound healing, as modeled by an increase in intestinal fibroblast-mediated collagen gel contraction. Guided by results from transcriptional profiling, we identified the selective induction of fibroblast integrin beta1 (ITGB1) by hypoxia. Further analysis revealed that hypoxia, as well as pharmacological activators of hypoxia-inducible factor (HIF), induce fibroblast beta1 integrin mRNA, protein, and function by as much as 4-fold. Cloning and analysis of the beta1 integrin gene promoter revealed a 10 +/- 0.8-fold increase in promoter activity in response to hypoxia, and subsequent studies identified a functional DNA binding region for HIF in the ITGB1 gene promoter. Mutational analysis of the HIF binding site within the ITGB1 promoter resulted in a significant loss of ITGB1 hypoxia-inducibility. As proof of principle, studies in a murine model of colitis revealed a correlation between colitic disease severity and tissue ITGB1 expression (R(2)=0.80). Taken together, these results demonstrate that hypoxia induces fibroblast ITGB1 expression and function by transcriptional mechanisms dependent on HIF.


Assuntos
Fator 1 Induzível por Hipóxia/fisiologia , Hipóxia/fisiopatologia , Integrina beta1/genética , Cicatrização/fisiologia , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/fisiopatologia , Colágeno/metabolismo , Células HeLa , Humanos , Camundongos
10.
Trends Cardiovasc Med ; 18(3): 103-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18436149

RESUMO

Nucleotide signaling is currently an area of intense investigation. Extracellular adenosine triphosphate (ATP) liberated during hypoxia or inflammation can either signal directly to purinergic receptors or, after phosphohydrolytic metabolism, can activate surface adenosine receptors. Given the association of polymorphonuclear leukocytes (PMNs) with adenine nucleotide/nucleoside signaling in the inflammatory milieu, it was recently demonstrated that PMNs actively release ATP via a connexin 43 hemichannel-dependent mechanism. Here, we review the mechanisms of ATP release and subsequent functional implications of ATP metabolism at the interface between PMN and vascular endothelial cells during inflammation and in hypoxia.


Assuntos
Trifosfato de Adenosina/fisiologia , Endotélio Vascular/fisiologia , Ativação de Neutrófilo/fisiologia , Neutrófilos/fisiologia , Nucleotídeos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Hipóxia Celular/fisiologia , Endotélio Vascular/metabolismo , Humanos , Neutrófilos/metabolismo , Transdução de Sinais
11.
Mol Cancer Res ; 5(7): 737-48, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17606477

RESUMO

We have shown previously that interleukin-8 (IL-8) and IL-8 receptor expression is elevated in tumor cells of human prostate biopsy tissue and correlates with increased cyclin D1 expression. Using PC3 and DU145 cell lines, we sought to determine whether IL-8 signaling regulated cyclin D1 expression in androgen-independent prostate cancer (AIPC) cells and to characterize the signaling pathways underpinning this response and that of IL-8-promoted proliferation. Administration of recombinant human IL-8 induced a rapid, time-dependent increase in cyclin D1 expression in AIPC cells, a response attenuated by the translation inhibitor cycloheximide but not by the RNA synthesis inhibitor, actinomycin D. Suppression of endogenous IL-8 signaling using neutralizing antibodies to IL-8 or its receptors also attenuated basal cyclin D1 expression in AIPC cells. Immunoblotting using phospho-specific antibodies confirmed that recombinant human IL-8 induced rapid time-dependent phosphorylation of Akt and the mammalian target of rapamycin substrate proteins, 4E-BP1 and ribosomal S6 kinase, resulting in a downstream phosphorylation of the ribosomal S6 protein (rS6). LY294002 and rapamycin each abrogated the IL-8-promoted phosphorylation of rS6 and attenuated the rate of AIPC cell proliferation. Our results indicate that IL-8 signaling (a) regulates cyclin D1 expression at the level of translation, (b) regulates the activation of proteins associated with the translation of capped and 5'-oligopyrimidine tract transcripts, and (c) activates signal transduction pathways underpinning AIPC cell proliferation. This study provides a molecular basis to support the correlation of IL-8 expression with that of cyclin D1 in human prostate cancer and suggests a mechanism by which this chemokine promotes cell proliferation.


Assuntos
Ciclina D1/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interleucina-8/farmacologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Biossíntese de Proteínas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Androgênios/metabolismo , Proteínas de Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ativação Enzimática/efeitos dos fármacos , Humanos , Interleucina-8/metabolismo , Masculino , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase D/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR
12.
J Crohns Colitis ; 11(9): 1101-1112, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28472424

RESUMO

BACKGROUND AND AIMS: Inflammatory bowel disease [IBD] is characterised by a disruption of immune homeostasis, which is tightly regulated to protect against harmful pathogens yet not react to commensal antigens. Animal studies indicate that regulatory T cells [Treg] modulate the immune response to prevent IBD development. Lactoferrin [LF] is an endogenous anti-inflammatory pleiotropic protein secreted at high concentrations in colostrum and at mucosal sites. However, the effect of LF on specific T lymphocyte populations has not been studied. Here, we identify a novel mechanism by which a recombinant human LF, VEN-120, regulates T cell populations in health and disease. METHODS: Two murine models of intestinal inflammation, the dextran sodium sulphate colitis model and the TNFΔARE/+ model of ileitis, were used to study the anti-inflammatory and T cell modulating ability of VEN-120. Flow cytometry was used to evaluate T cell populations within the lamina propria and mesenteric lymph nodes, and to evaluate the effect of VEN-120 on CD4+ T cells in vitro. RESULTS: VEN-120 reduced inflammation in both models of IBD, accompanied by increased Tregs in the intestinal lamina propria. Treatment of CD4+ T cells in vitro resulted in an upregulation of Treg genes and skewing towards a Treg population. This in vitro T cell skewing translated to an increase of Treg homing to the intestinal lamina propria and associated lymph tissue in healthy mice. CONCLUSIONS: These data provide a novel immunological mechanism by which VEN-120 modulates T cells to restrict inflammatory T cell-driven disease.


Assuntos
Colite/imunologia , Ileíte/imunologia , Doenças Inflamatórias Intestinais/imunologia , Lactoferrina/imunologia , Fenótipo , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/imunologia
13.
Mol Biol Cell ; 21(4): 538-46, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20032301

RESUMO

Mucosal surfaces, such as the lung and intestine, are lined by a monolayer of epithelia that provides tissue barrier and transport function. It is recently appreciated that a common feature of inflammatory processes within the mucosa is hypoxia (so-called inflammatory hypoxia). Given the strong association between bacterial translocation and mucosal inflammatory disease, we hypothesized that intestinal epithelial hypoxia influences bacterial translocation. Initial studies revealed that exposure of cultured intestinal epithelia to hypoxia (pO(2), 20 torr; 24-48 h) resulted in a increase of up to 40-fold in the translocation of some strains of Gram-positive bacteria, independently of epithelial barrier function. A screen of relevant pathway inhibitors identified a prominent role for the platelet-activating factor receptor (PAFr) in hypoxia-associated bacterial translocation, wherein pharmacologic antagonists of PAFr blocked bacterial translocation by as much as 80 +/- 6%. Extensions of these studies revealed that hypoxia prominently induces PAFr through a hypoxia-inducible factor (HIF)-dependent mechanism. Indeed, HIF and PAFr loss of function studies (short hairpin RNA) revealed that apically expressed PAFr is central to the induction of translocation for the Gram-positive bacteria Enterococcus faecalis. Together, these findings reveal that some strains of Gram-positive bacteria exploit HIF-regulated PAFr as a means for translocation through intestinal epithelial cells.


Assuntos
Bactérias Gram-Positivas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mucosa Intestinal , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Transporte Biológico/fisiologia , Células CACO-2 , Membrana Celular/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Glicoproteínas da Membrana de Plaquetas/genética , Interferência de RNA , Receptores Acoplados a Proteínas G/genética , Ativação Transcricional
14.
J Immunol ; 180(6): 4246-55, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322237

RESUMO

Inflammatory diseases influence tissue metabolism, altering regulation of extracellular adenine nucleotides, with a resultant protective influence of adenosine. Ecto-5'-nucleotidase (CD73) is a central surface enzyme generating extracellular adenosine. Thus, we hypothesized that CD73 is protective in mucosal inflammation as modeled by trinitrobenzene sulfonate (TNBS) colitis. Initial studies revealed a >3-fold induction of CD73 mRNA levels after TNBS colitis. Additionally, the severity of colitis was increased, as determined by weight loss and colonic shortening, in cd73(-/-) mice relative to cd73(+/+) controls. Likewise, enteral administration of the selective CD73 inhibitor alpha,beta-methylene ADP to cd73(+/+) mice resulted in a similar increase in severity of TNBS colitis. Gene array profiling of cytokine mRNA expression, verified by real-time PCR, revealed a >90% down-regulation of IFN-alphaA in cd73(-/-) mice and alpha,beta-methylene ADP-treated cd73(+/+) mice, compared with cd73(+/+) mice. Exogenous administration of recombinant IFN-alphaA partially protected TNBS-treated cd73(-/-) mice. Cytokine profiling revealed similar increases in both IFN-gamma and TNF-alpha mRNA in colitic animals, independent of genotype. However, IL-10 mRNA increased in wild-type mice on day 3 after TNBS administration, whereas cd73(-/-) mice mounted no IL-10 response. This IL-10 response was restored in the cd73(-/-) mice by exogenous IFN-alphaA. Further cytokine profiling revealed that this IL-10 induction is preceded by a transient IFN-alphaA induction on day 2 after TNBS exposure. Together, these studies indicate a critical regulatory role for CD73-modulated IFNalphaA in the acute inflammatory phase of TNBS colitis, thereby implicating IFN-alphaA as a protective element of adenosine signaling during mucosal inflammation.


Assuntos
5'-Nucleotidase/fisiologia , Mediadores da Inflamação/fisiologia , Interferon-alfa/administração & dosagem , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , 5'-Nucleotidase/antagonistas & inibidores , 5'-Nucleotidase/biossíntese , 5'-Nucleotidase/deficiência , Doença Aguda , Difosfato de Adenosina/administração & dosagem , Difosfato de Adenosina/análogos & derivados , Animais , Colite/induzido quimicamente , Colite/imunologia , Colite/patologia , Progressão da Doença , Regulação para Baixo/imunologia , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/biossíntese , Interferon-alfa/genética , Interleucina-10/biossíntese , Interleucina-10/deficiência , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Ácido Trinitrobenzenossulfônico/toxicidade
15.
Wound Repair Regen ; 14(1): 61-5, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16476073

RESUMO

This study examined the mechanism of wound repair in the early chick amnion following surgical puncture. The chick amnion is a bilayered membrane with ultrastructural features similar to the human amnion, and thus may provide a model of the consequences of amnion puncture following first trimester amniocentesis in humans. Chick amnion was wounded on day 4-5 of incubation. The rate of wounding was measured as 40 microm2/minute by hourly measurements of wound area from initial wounding to a point where the wound appeared to be fully closed. Morphological changes were examined by scanning electron microscopy of amniotic membranes with the overlying chorionic membrane removed. Cells bordering the wound arranged themselves circumferentially during the healing period and eventually came together as a cellular pile up as the healing process was complete. The presence of an actin cable was revealed by fluorescein isothiocyanate-labeled phalloidin. The actin was circumferentially arranged around the wound margin and appeared within 10 minutes after wounding. Treatment of the amnion with the inhibitor of endogenous Rho, C3 exotransferase, inhibited actin cable formation, suggesting that formation of an actin cable within the amnion during wound healing is Rho dependent.


Assuntos
Actinas/fisiologia , Proteínas de Fase Aguda/fisiologia , Âmnio/lesões , Cicatrização/fisiologia , Âmnio/ultraestrutura , Animais , Embrião de Galinha , Microscopia Eletrônica de Varredura , Transdução de Sinais/fisiologia , Coloração e Rotulagem , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA