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











Base de dados
Intervalo de ano de publicação
1.
Nat Microbiol ; 4(2): 269-279, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30510170

RESUMO

Clostridium difficile infection (CDI) is mediated by two major exotoxins, toxin A (TcdA) and toxin B (TcdB), that damage the colonic epithelial barrier and induce inflammatory responses. The function of the colonic vascular barrier during CDI has been relatively understudied. Here we report increased colonic vascular permeability in CDI mice and elevated vascular endothelial growth factor A (VEGF-A), which was induced in vivo by infection with TcdA- and/or TcdB-producing C. difficile strains but not with a TcdA-TcdB- isogenic mutant. TcdA or TcdB also induced the expression of VEGF-A in human colonic mucosal biopsies. Hypoxia-inducible factor signalling appeared to mediate toxin-induced VEGF production in colonocytes, which can further stimulate human intestinal microvascular endothelial cells. Both neutralization of VEGF-A and inhibition of its signalling pathway attenuated CDI in vivo. Compared to healthy controls, CDI patients had significantly higher serum VEGF-A that subsequently decreased after treatment. Our findings indicate critical roles for toxin-induced VEGF-A and colonic vascular permeability in CDI pathogenesis and may also point to the pathophysiological significance of the gut vascular barrier in response to virulence factors of enteric pathogens. As an alternative to pathogen-targeted therapy, this study may enable new host-directed therapeutic approaches for severe, refractory CDI.


Assuntos
Toxinas Bacterianas/metabolismo , Permeabilidade Capilar , Clostridioides difficile/química , Infecções por Clostridium/patologia , Enterotoxinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Virulência/metabolismo , Animais , Toxinas Bacterianas/genética , Clostridioides difficile/patogenicidade , Infecções por Clostridium/metabolismo , Colo/metabolismo , Colo/patologia , Enterotoxinas/genética , Epitélio/metabolismo , Epitélio/patologia , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Neovascularização Patológica , Transdução de Sinais , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/sangue , Fatores de Virulência/genética
2.
J Immunol ; 196(10): 4311-21, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27076683

RESUMO

Neurotensin (NT) via its receptor 1 (NTR1) modulates the development of colitis, decreases HIF-1α/PHD2 interaction, stabilizes and increases HIF-1α transcriptional activity, and promotes intestinal angiogenesis. HIF-1α induces miR-210 expression, whereas miR-210 is strongly upregulated in response to NT in NCM460 human colonic epithelial cells overexpressing NTR1 (NCM460-NTR1). In this study, we examined whether NT activates a NTR1-HIF-1α-miR-210 cascade using in vitro (NCM460-NTR1 cells) and in vivo (transgenic mice overexpressing [HIF-1α-OE] or lacking HIF-1α [HIF-1α-knockout (KO)] in intestinal epithelial cells and mice lacking NTR1 [NTR1-KO]) models. Pretreatment of NCM460-NTR1 cells with the HIF-1α inhibitor PX-478 or silencing of HIF-1α (small interfering HIF-1α) attenuated miR-210 expression in response to NT. Intracolonic 2,4,6-trinitrobenzenesulfonic acid (TNBS) administration (2-d model) increased colonic miR-210 expression that was significantly reduced in NTR1-KO, HIF-1α-KO mice, and wild-type mice pretreated intracolonically with locked nucleic acid anti-miR-210. In contrast, HIF-1α-OE mice showed increased miR-210 expression at baseline that was further increased following TNBS administration. HIF-1α-OE mice had also exacerbated TNBS-induced neovascularization compared with TNBS-exposed wild-type mice. TNBS-induced neovascularization was attenuated in HIF-1α-KO mice, or mice pretreated intracolonically with anti-miR-210. Intracolonic anti-miR-210 also reduced colitis in response to TNBS (2 d). Importantly, miR-210 expression was increased in tissue samples from ulcerative colitis patients. We conclude that NT exerts its proinflammatory and proangiogenic effects during acute colitis via a NTR1-prolyl hydroxylase 2/HIF-1α-miR-210 signaling pathway. Our results also demonstrate that miR-210 plays a proinflammatory role in the development of colitis.


Assuntos
Colite Ulcerativa/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/metabolismo , Neovascularização Patológica/metabolismo , Neurotensina/metabolismo , Animais , Linhagem Celular Tumoral , Colo/irrigação sanguínea , Colo/patologia , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Compostos de Mostarda/administração & dosagem , NF-kappa B/metabolismo , Fenilpropionatos/administração & dosagem , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo , Transdução de Sinais , Ácido Trinitrobenzenossulfônico , Regulação para Cima/efeitos dos fármacos
3.
PLoS One ; 10(7): e0130938, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26222740

RESUMO

INTRODUCTION: Animal studies have shown that stress could induce epigenetic and transcriptomic alterations essential in determining the balance between adaptive or maladaptive responses to stress. We tested the hypothesis that chronic stress in rats deregulates coding and non-coding gene expression in the spinal cord, which may underline neuroinflammation and nociceptive changes previously observed in this model. METHODS: Male Wistar rats were exposed to daily stress or handled, for 10 days. At day 11, lumbar spinal segments were collected and processed for mRNA/miRNA isolation followed by expression profiling using Agilent SurePrint Rat Exon and Rat miRNA Microarray platforms. Differentially expressed gene lists were generated using the dChip program. Microarrays were analyzed using the Ingenuity Pathways Analysis (IPA) tool from Ingenuity Systems. Multiple methods were used for the analysis of miRNA-mRNA functional modules. Quantitative real time RT-PCR for Interleukin 6 signal transducer (gp130), the Signal Transducer And Activator Of Transcription 3 (STAT3), glial fibrillary acidic protein and mir-17-5p were performed to confirm levels of expression. RESULTS: Gene network analysis revealed that stress deregulated different inflammatory (IL-6, JAK/STAT, TNF) and metabolic (PI3K/AKT) signaling pathways. MicroRNA array analysis revealed a signature of 39 deregulated microRNAs in stressed rats. MicroRNA-gene network analysis showed that microRNAs are regulators of two gene networks relevant to inflammatory processes. Specifically, our analysis of miRNA-mRNA functional modules identified miR-17-5p as an important regulator in our model. We verified miR-17-5p increased expression in stress using qPCR and in situ hybridization. In addition, we observed changes in the expression of gp130 and STAT3 (involved in intracellular signaling cascades in response to gp130 activation), both predicted targets for miR-17-5p. A modulatory role of spinal mir17-5p in the modulation of visceral sensitivity was confirmed in vivo. CONCLUSION: Using an integrative high throughput approach, our findings suggest a link between miR-17-5p increased expression and gp130/STAT3 activation providing new insight into the possible mechanisms mediating the effect of chronic stress on neuroinflammation in the spinal cord.


Assuntos
Receptor gp130 de Citocina/biossíntese , Hiperalgesia/metabolismo , MicroRNAs/biossíntese , Fator de Transcrição STAT3/metabolismo , Medula Espinal/metabolismo , Estresse Psicológico/metabolismo , Animais , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Hiperalgesia/patologia , Janus Quinases/biossíntese , Masculino , Ratos , Ratos Wistar , Transdução de Sinais , Medula Espinal/patologia , Estresse Psicológico/patologia , Fator de Necrose Tumoral alfa/biossíntese
4.
Clin Exp Gastroenterol ; 8: 13-29, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25565877

RESUMO

BACKGROUND: Cathelicidin (LL-37 in humans and mCRAMP in mice) represents a family of endogenous antimicrobial and anti-inflammatory peptides. Cancer-associated fibroblasts can promote the proliferation of colon cancer cells and growth of colon cancer tumors. METHODS: We examined the role of cathelicidin in the development of colon cancer, using subcutaneous human HT-29 colon-cancer-cell-derived tumor model in nude mice and azoxymethane- and dextran sulfate-mediated colon cancer model in C57BL/6 mice. We also determined the indirect antitumoral mechanism of cathelicidin via the inhibition of epithelial-mesenchymal transition (EMT) of colon cancer cells and fibroblast-supported colon cancer cell proliferation. RESULTS: Intravenous administration of cathelicidin expressing adeno-associated virus significantly reduced the size of tumors, tumor-derived collagen expression, and tumor-derived fibroblast expression in HT-29-derived subcutaneous tumors in nude mice. Enema administration of the mouse cathelicidin peptide significantly reduced the size and number of colonic tumors in azoxymethane- and dextran sulfate-treated mice without inducing apoptosis in tumors and the adjacent normal colonic tissues. Cathelicidin inhibited the collagen expression and vimentin-positive fibroblast expression in colonic tumors. Cathelicidin did not directly affect HT-29 cell viability, but did significantly reduce tumor growth factor-ß1-induced EMT of colon cancer cells. Media conditioned by the human colonic CCD-18Co fibroblasts promoted human colon cancer HT-29 cell proliferation. Cathelicidin pretreatment inhibited colon cancer cell proliferation mediated by media conditioned by human colonic CCD-18Co fibroblasts. Cathelicidin disrupted tubulin distribution in colonic fibroblasts. Disruption of tubulin in fibroblasts reduced fibroblast-supported colon cancer cell proliferation. CONCLUSION: Cathelicidin effectively inhibits colon cancer development by interfering with EMT and fibroblast-supported colon cancer cell proliferation.

5.
Gut ; 64(7): 1095-104, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25112884

RESUMO

OBJECTIVE: Neurotensin (NT) mediates colonic inflammation through its receptor neurotensin receptor 1 (NTR1). NT stimulates miR-133α expression in colonic epithelial cells. We investigated the role of miR-133α in NT-associated colonic inflammation in vitro and in vivo. DESIGN: miR-133α and aftiphilin (AFTPH) levels were measured by quantitative PCR. Antisense (as)-miR-133α was administrated intracolonicaly prior to induction of 2, 4, 6-trinitrobenzene sulfonic acid (TNBS)-induced colitis and dextran sodium sulfate (DSS)-induced colitis. The effect of AFTPH was examined by gene silencing in vitro. RESULTS: NT increased miR-133α levels in NCM-460 overexpressing NTR1 (NCM460-NTR1) and HCT-116 cells. NT-induced p38, ERK1/2, c-Jun, and NF-κB activation, as well as IL-6, IL-8 and IL-1ß messenger RNA (mRNA) expression in NCM-460-NTR1 cells were reduced in miR-133α-silenced cells, while overexpression of miR-133α reversed these effects. MiR-133α levels were increased in TNBS (2 day) and DSS (5 day) colitis, while NTR1 deficient DSS-exposed mice had reduced miR-133α levels, compared to wild-type colitic mice. Intracolonic as-miR-133α attenuated several parameters of colitis as well expression of proinflammatory mediators in the colonic mucosa. In silico search coupled with qPCR identified AFTPH as a downstream target of miR-133α, while NT decreased AFTPH expression in NCM-460-NTR1 colonocytes. Gene silencing of AFTPH enhanced NT-induced proinflammatory responses and AFTPH levels were downregulated in experimental colitis. Levels of miR-133α were significantly upregulated, while AFTPH levels were downregulated in colonic biopsies of patients with ulcerative colitis compared to controls. CONCLUSIONS: NT-associated colitis and inflammatory signalling are regulated by miR-133α-AFTPH interactions. Targeting of miR-133α or AFTPH may represent a novel therapeutic approach in inflammatory bowel disease.


Assuntos
Colite/fisiopatologia , Colo/fisiologia , Células Epiteliais/fisiologia , Animais , Colo/citologia , Células HCT116 , Humanos , Camundongos , Camundongos Knockout , MicroRNAs , NF-kappa B/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de Neurotensina/genética , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
6.
Am J Pathol ; 184(12): 3405-14, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25307345

RESUMO

The expression of neurotensin (NT) and its receptor (NTR1) is up-regulated in experimental colitis and inflammatory bowel disease; NT/NTR1 interactions regulate gut inflammation. During active inflammation, metabolic shifts toward hypoxia lead to the activation of hypoxia-inducible factor (HIF)-1, which enhances vascular endothelial growth factor (VEGF) expression, promoting angiogenesis. We hypothesized that NT/NTR1 signaling regulates intestinal manifestations of hypoxia and angiogenesis by promoting HIF-1 transcriptional activity and VEGFα expression in experimental colitis. We studied NTR1 signaling in colitis-associated angiogenesis using 2,4,6-trinitrobenzenesulfonic acid-treated wild-type and NTR1-knockout mice. The effects of NT on HIF-1α and VEGFα were assessed on human colonic epithelial cells overexpressing NTR1 (NCM460-NTR1) and human intestinal microvascular-endothelial cells. NTR1-knockout mice had reduced microvascular density and mucosal integrity score compared with wild-type mice after 2,4,6-trinitrobenzenesulfonic acid treatment. VEGFα mRNA levels were increased in NCM460-NTR1 cells treated with 10(-7) mol/L NT, at 1 and 6 hours post-treatment. NT exposure in NCM460-NTR1 cells caused stabilization, nuclear translocation, and transcriptional activity of HIF-1α in a diacylglycerol kinase-dependent manner. NT did not stimulate tube formation in isolated human intestinal macrovascular endothelial cells but did so in human intestinal macrovascular endothelial cells cocultured with NCM460-NTR1 cells. Our results demonstrate the importance of an NTR1-HIF-1α-VEGFα axis in intestinal angiogenic responses and in the pathophysiology of colitis and inflammatory bowel disease.


Assuntos
Colo/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Neurotensina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Colite/patologia , Colo/citologia , Modelos Animais de Doenças , Células Endoteliais/citologia , Humanos , Imuno-Histoquímica , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/metabolismo , Intestinos/irrigação sanguínea , Masculino , Camundongos , Microcirculação , Neovascularização Patológica , Ácido Trinitrobenzenossulfônico/química , Regulação para Cima
8.
J Biol Chem ; 287(18): 15066-75, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22416137

RESUMO

The neuropeptide/hormone neurotensin (NT) mediates intestinal inflammation and cell proliferation by binding of its high affinity receptor, neurotensin receptor-1 (NTR1). NT stimulates IL-8 expression in NCM460 human colonic epithelial cells by both MAP kinase- and NF-κB-dependent pathways. Although the mechanism of NTR1 endocytosis has been studied, the relationship between NTR1 intracellular trafficking and inflammatory signaling remains to be elucidated. In the present study, we show that in NCM460 cells exposed to NT, ß-arrestin-1 (ßARR1), and ß-arrestin-2 (ßARR2) translocate to early endosomes together with NTR1. Endothelin-converting enzyme-1 (ECE-1) degrades NT in acidic conditions, and its activity is crucial for NTR1 recycling. Pretreatment of NCM460 cells with the ECE-1 inhibitor SM19712 or gene silencing of ßARR1 or ßARR2 inhibits NT-stimulated ERK1/2 and JNK phosphorylation, NF-κB p65 nuclear translocation and phosphorylation, and IL-8 secretion. Furthermore, NT-induced cell proliferation, but not IL-8 transcription, is attenuated by the JNK inhibitor, JNK(AII). Thus, NTR1 internalization and recycling in human colonic epithelial cells involves ßARRs and ECE-1, respectively. Our results also indicate that ßARRs and ECE-1-dependent recycling regulate MAP kinase and NF-κB signaling as well as cell proliferation in human colonocytes in response to NT.


Assuntos
Arrestinas/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Endocitose/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Metaloendopeptidases/metabolismo , Neurotensina/metabolismo , Proteólise , Receptores de Neurotensina/metabolismo , Arrestinas/genética , Ácido Aspártico Endopeptidases/genética , Linhagem Celular , Colo/citologia , Colo/metabolismo , Endocitose/efeitos dos fármacos , Enzimas Conversoras de Endotelina , Humanos , Inflamação/genética , Inflamação/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Metaloendopeptidases/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurotensina/genética , Receptores de Neurotensina/antagonistas & inibidores , Receptores de Neurotensina/genética , Sulfonamidas/farmacologia , Compostos de Sulfonilureia/farmacologia , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , beta-Arrestina 1 , beta-Arrestina 2 , beta-Arrestinas
9.
Gastroenterology ; 141(5): 1749-61.e1, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21806946

RESUMO

BACKGROUND & AIMS: Neurotensin promotes inflammation and colon cancer via the neurotensin-1 receptor (NTR1). MicroRNAs (miR) regulate protein synthesis by degrading or preventing translation of mRNAs. We analyzed expression of 365 different microRNAs by human colonic epithelial cells (NCM460) after activation of NTR1. METHODS: We performed microarray analysis of mRNA expression by neurotensin-stimulated NCM460 cells that overexpressed NTR1. Nuclear factor-κB (NF-κB) binding sites were identified and tumorigenesis was assessed using soft agar assays and xenograft analysis of severe combined immunodeficiency mice. Targets of neurotensin-regulated microRNAs were identified via bioinformatic, real-time polymerase chain reaction, and immunoblot analyses. We analyzed RNA samples from human normal colon and tumor samples. RESULTS: Neurotensin stimulated differential expression of 38 microRNAs, including miR-21 and miR-155, which have been associated with tumor growth and contain NF-κB binding sites. Neurotensin expression increased colony formation by HCT-116 cells. Blocking miR-21 and/or miR-155 prevented colony formation (P < .001). In mice, intraperitoneal administration of neurotensin increased the growth rate of HCT-116 xenograft tumors; blocking miR-21 and/or miR-155 slowed this tumor growth. Neurotensin activated Akt in HCT-116 cells; this effect was inhibited by blocking miR-21 and/or miR-155 (P < .001). Neurotensin activated AKT through miR-155-mediated suppression of the phosphatase protein phosphatase 2A catalytic subunit alpha (PPP2CA). Levels of phosphatase and tensin homolog (PTEN) and suppressor of cytokine signaling 1 (SOCS1) mRNA, potential targets of miR-21 and miR-155, respectively, were down-regulated by these miRs. Levels of NTR1, miR-21, and miR-155 increased significantly in human colon tumor samples, compared with normal tissues, whereas PPP2CA, SOCS1, and PTEN mRNAs were reduced significantly. CONCLUSIONS: NTR1 activation stimulates expression of miR-21 and miR-155 in colonocytes, via Akt and NF-κB, to down-regulate PTEN and SOCS1 and promote growth of tumors in mice. Levels of NTR1, miR-21, and miR-155 increase in human colon tumor samples and correlate with tumor stage.


Assuntos
Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , MicroRNAs/metabolismo , Neurotensina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteína Fosfatase 2/metabolismo , Receptores de Neurotensina/metabolismo , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transplante Heterólogo
10.
Gastroenterology ; 141(5): 1852-63.e1-3, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21762664

RESUMO

BACKGROUND & AIMS: Cathelicidin (encoded by Camp) is an antimicrobial peptide in the innate immune system. We examined whether macrophages express cathelicidin in colons of mice with experimental colitis and patients with inflammatory bowel disease, and we investigated its signaling mechanisms. METHODS: Quantitative, real-time, reverse-transcription polymerase chain reaction (PCR), bacterial 16S PCR, immunofluorescence, and small interfering RNA (siRNA) analyses were performed. Colitis was induced in mice using dextran sulfate sodium (DSS); levels of cathelicidin were measured in human primary monocytes. RESULTS: Expression of cathelicidin increased in the inflamed colonic mucosa of mice with DSS-induced colitis compared with controls. Cathelicidin expression localized to mucosal macrophages in inflamed colon tissues of patients and mice. Exposure of human primary monocytes to Escherichia coli DNA induced expression of Camp messenger RNA, which required signaling by extracellular signal-regulated kinase (ERK); expression was reduced by siRNAs against Toll-like receptor (TLR)9 and MyD88. Intracolonic administration of bacterial DNA to wild-type mice induced expression of cathelicidin in colons of control mice and mice with DSS-induced colitis. Colon expression of cathelicidin was significantly reduced in TLR9(-/-) mice with DSS-induced colitis. Compared with wild-type mice, Camp(-/-) mice developed a more severe form of DSS-induced colitis, particularly after intracolonic administration of E coli DNA. Expression of cathelicidin from bone marrow-derived immune cells regulated DSS induction of colitis in transplantation studies in mice. CONCLUSIONS: Cathelicidin protects against induction of colitis in mice. Increased expression of cathelicidin in monocytes and experimental models of colitis involves activation of TLR9-ERK signaling by bacterial DNA. This pathway might be involved in the pathogenesis of ulcerative colitis.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Colite/metabolismo , Colite/prevenção & controle , Transdução de Sinais/fisiologia , Receptor Toll-Like 9/metabolismo , Animais , Células Cultivadas , Colite/induzido quimicamente , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor Toll-Like 9/genética , Regulação para Cima , Catelicidinas
11.
Curr Pharm Des ; 17(16): 1576-82, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21548874

RESUMO

Until recently, fat was considered a relatively inactive tissue serving only as a depot for the storage of excess lipid around the body. Over the last decade, however, several studies have established fat as a metabolically active endocrine organ able to affect human pathophysiology at multiple levels. During this time adipose tissue has been shown to produce a number of hormones and inflammatory mediators collectively termed as adipokines. These molecules have been shown to be involved in the etiology of a number of inflammation-associated pathological conditions ranging from atherosclerosis and hypertension to diabetes and cancer. Despite the close physical association of abdominal fat and the intestine in the visceral cavity and the significant paracrine functions now attributed to adipose tissue, very little is known on the potential interactions between these tissues as they may relate to intestinal homeostasis. Considering the dramatic alterations in mesenteric fat depot size and placement during at least one intestinal disease, Crohn's disease, the potential involvement of fat tissue in the development as well as the progression of this and other pathological conditions should be considered. In this review we discuss the latest knowledge on neuropeptide-adipose tissue communication and the potential changes such interaction may induce in intra-abdominal fat tissue physiology. Finally we will discuss evidence on the potential pathways by which such changes in fat physiology may affect the development and progress of intestinal pathological conditions such as inflammatory bowel disease.


Assuntos
Doenças Inflamatórias Intestinais/fisiopatologia , Gordura Intra-Abdominal/fisiologia , Neuropeptídeos/fisiologia , Animais , Humanos
12.
J Biol Chem ; 286(8): 6092-9, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21212273

RESUMO

Neurotensin (NT) is a gastrointestinal neuropeptide that modulates intestinal inflammation and healing by binding to its high-affinity receptor NTR1. The dual role of NT in inflammation and healing is demonstrated in models of colitis induced by Clostridium difficile toxin A and dextran sulfate sodium, respectively, and involves NF-κB-dependent IL-8 expression and EGF receptor-mediated MAPK activation in human colonocytes. However, the detailed signaling pathways involved in these responses remain to be elucidated. We report here that NT/NTR1 coupling in human colonic epithelial NCM460 cells activates tyrosine phosphorylation of the insulin-like growth factor-1 receptor (IGF-1R) in a time- and dose-dependent manner. NT also rapidly induces Src tyrosine phosphorylation, whereas pretreatment of cells with the Src inhibitor PP2 before NT exposure decreases NT-induced IGF-1R phosphorylation. In addition, inhibition of IGF-1R activation by either its specific antagonist AG1024 or siRNA against IGF-1 significantly reduces NT-induced IL-8 expression and NF-κB-dependent reporter gene expression. Pretreatment with AG1024 also inhibits Akt activation and apoptosis induced by NT. Silencing of Akt expression by siRNA also substantially attenuates NT-induced IL-8 promoter activity and NF-κB-dependent reporter gene expression. This is the first report to indicate that NT transactivates IGF-1R and that this response is linked to Akt phosphorylation and NF-κB activation, contributing to both pro-inflammatory and tissue repair signaling pathways in response to NT in colonic epithelial cells. We propose that IGF-1R activation represents a previously unrecognized key pathway involved in the mechanisms by which NT and NTR1 modulate colonic inflammation and inflammatory bowel disease.


Assuntos
Colo/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Neurotensina/farmacologia , Receptor IGF Tipo 1/metabolismo , Apoptose/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Linhagem Celular , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Colo/patologia , Sulfato de Dextrana/efeitos adversos , Sulfato de Dextrana/farmacologia , Relação Dose-Resposta a Droga , Enterotoxinas/toxicidade , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Interleucina-8/biossíntese , Mucosa Intestinal/patologia , NF-kappa B/metabolismo , Neurotensina/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptor IGF Tipo 1/agonistas , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo , Fatores de Tempo , Tirfostinas/farmacologia
13.
Cell Cycle ; 9(24): 4931-40, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21150329

RESUMO

The role of nitric oxide (NO) in cancer has been controversial and is based on the levels of NO and the responsiveness of the tumor type. It remains unclear whether NO can inhibit the epithelial to mesenchymal transition (EMT) in cancer cells. EMT induction is mediated, in part, by the constitutive activation of the metastasis-inducer transcription factor, Snail and EMT can be inhibited by the metastasis-suppressor Raf-1 kinase inhibitor protein (RKIP) and E-cadherin. Snail is transcriptionally regulated by NF-κB and in turn, Snail represses RKIP transcription. Hence, we hypothesized that high levels of NO, that inhibit NF-κB activity, may also inhibit Snail and induce RKIP and leading to inhibition of EMT. We show that treatment of human prostate metastatic cell lines with the NO donor, DETANONOate, inhibits EMT and reverses both the mesenchymal phenotype and the cell invasive properties. Further, treatment with DETANONOate inhibits Snail expression and DNA-binding activity in parallel with the upregulation of RKIP and E-cadherin protein levels. The pivotal roles of Snail inhibition and RKIP induction in DETANONOate-mediated inhibition of EMT were corroborated by both Snail silencing by siRNA and by ectopic expression of RKIP. The in vitro findings were validated in vivo in mice bearing PC-3 xenografts and treated with DETANONOate. The present findings show, for the first time, the novel role of high subtoxic concentrations of NO in the inhibition of EMT. Thus, NO donors may exert therapeutic activities in the reversal of EMT and metastasis.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias/patologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Masculino , Camundongos , Neoplasias/metabolismo , Compostos Nitrosos/farmacologia , Fenótipo , Proteína de Ligação a Fosfatidiletanolamina/genética , Neoplasias da Próstata/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transplante Heterólogo
14.
J Biol Chem ; 280(8): 7262-72, 2005 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15590695

RESUMO

The four-transmembrane domain proteins synaptophysin and synaptogyrin represent the major constituents of synaptic vesicles. Our previous studies in PC12 cells demonstrated that synaptogyrin or its nonneuronal paralog cellugyrin targets efficiently to synaptic-like microvesicles (SLMVs) and dramatically increases the synaptophysin content of SLMVs (Belfort, G. M., and Kandror, K. V. (2003) J. Biol. Chem. 278, 47971-47978). Here, we explored the mechanism of these phenomena and found that ectopic expression of cellugyrin increases the number of SLMVs in PC12 cells. Mutagenesis studies revealed that cellugyrin's hydrophilic cytoplasmic domains are not involved in vesicle biogenesis, whereas small conserved hydrophobic hairpins in the first luminal loop and the carboxyl terminus of cellugyrin were found to be critical for the formation of SLMVs. In addition, the length but not the primary sequence of the second luminal loop was essential for SLMV biogenesis. We suggest that changing the length of this loop similar to disruption of the short hydrophobic hairpins alters the position of the vicinal transmembrane domains that may be crucial for protein function.


Assuntos
Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Vesículas Sinápticas/metabolismo , Animais , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/genética , Microscopia de Fluorescência , Modelos Moleculares , Mutagênese , Proteínas do Tecido Nervoso/genética , Células PC12 , Estrutura Terciária de Proteína , Ratos , Vesículas Sinápticas/química , Vesículas Sinápticas/ultraestrutura , Sinaptogirinas , Sinaptofisina/análise , Sinaptofisina/metabolismo , Transfecção
15.
Med Sci Monit ; 9(7): BR276-82, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12883446

RESUMO

BACKGROUND: To see if the higher levels of nitric oxide expired by asthmatics compared to healthy subjects might be of significance to airway function, the effect of nitric oxide and its second messenger, guanosine 3', 5'- cyclic monophosphate (cGMP), on the permeability of human nasal epithelial cells was studied. MATERIAL/METHODS: Cells from healthy and asthmatic donors, collected by swab biopsy, were plated on agar gel before being impaled with a microelectrode to measure their intracellular potential and membrane resistance. RESULTS: Exposure of cells to 300 mM sodium nitroprusside, a nitric oxide donor, caused a profound fall in both parameters in cells from non-asthmatics but no change in cells from asthmatic subjects. A similar response was seen when cells were exposed to 0.9 mM of the permeable form of cGMP, 8-Br-cGMP. Selective inhibition of ion transport pathways in healthy cells indicated that nitric oxide produced changes in permeability consistent with secretion of anions by the cells. CONCLUSIONS: Since anion secretion is associated with fluid secretion in the intact epithelium, we suggest that nitric oxide mediates a protective mechanism to remove foreign material from the airway surface. The defective response to nitric oxide seen in asthmatic cells may contribute to the disease by compromising the removal of allergens from the airway.


Assuntos
Asma/metabolismo , Células Epiteliais/metabolismo , Mucosa Nasal/citologia , Óxido Nítrico/metabolismo , Adulto , Amilorida/metabolismo , Animais , Compostos de Bário/metabolismo , Bloqueadores dos Canais de Cálcio/metabolismo , Cloretos/metabolismo , Diuréticos/metabolismo , Eletrofisiologia , Inibidores Enzimáticos/metabolismo , Células Epiteliais/citologia , Feminino , Humanos , Masculino , Potenciais da Membrana , Pessoa de Meia-Idade , Doadores de Óxido Nítrico/metabolismo , Nitroprussiato/metabolismo , Permeabilidade , ortoaminobenzoatos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA