Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 159(1): 273-288, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32169429

RESUMO

BACKGROUND & AIMS: We investigated mechanisms of hepatic stellate cell (HSC) activation, which contributes to liver fibrogenesis. We aimed to determine whether activated HSCs increase glycolysis, which is regulated by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), and whether this pathway might serve as a therapeutic target. METHODS: We performed studies with primary mouse HSCs, human LX2 HSCs, human cirrhotic liver tissues, rats and mice with liver fibrosis (due to bile duct ligation [BDL] or administration of carbon tetrachloride), and CPEB4-knockout mice. Glycolysis was inhibited in cells and mice by administration of a small molecule antagonist of PFKFB3 (3-[3-pyridinyl]-1-[4-pyridinyl]-2-propen-1-one [3PO]). Cells were transfected with small interfering RNAs that knock down PFKFB3 or CPEB4. RESULTS: Up-regulation of PFKFB3 protein and increased glycolysis were early and sustained events during HSC activation and accompanied by increased expression of markers of fibrogenesis; incubation of HSCs with 3PO or knockdown of PFKFB3 reduced their activation and proliferation. Mice with liver fibrosis after BDL had increased hepatic PFKFB3; injection of 3PO immediately after the surgery prevented HSC activation and reduced the severity of liver fibrosis compared with mice given vehicle. Levels of PFKFB3 protein were increased in fibrotic liver tissues from patients compared with non-fibrotic liver. Up-regulation of PFKFB3 in activated HSCs did not occur via increased transcription, but instead via binding of CPEB4 to cytoplasmic polyadenylation elements within the 3'-untranslated regions of PFKFB3 messenger RNA. Knockdown of CPEB4 in LX2 HSCs prevented PFKFB3 overexpression and cell activation. Livers from CPEB4-knockout had decreased PFKFB3 and fibrosis after BDL or administration of carbon tetrachloride compared with wild-type mice. CONCLUSIONS: Fibrotic liver tissues from patients and rodents (mice and rats) have increased levels of PFKFB3 and glycolysis, which are essential for activation of HSCs. Increased expression of PFKFB3 is mediated by binding of CPEB4 to its untranslated messenger RNA. Inhibition or knockdown of CPEB4 or PFKFB3 prevents HSC activation and fibrogenesis in livers of mice.


Assuntos
Células Estreladas do Fígado/patologia , Cirrose Hepática Experimental/patologia , Cirrose Hepática/patologia , Fosfofrutoquinase-2/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Tetracloreto de Carbono/toxicidade , Linhagem Celular , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Glicólise , Humanos , Fígado/citologia , Fígado/patologia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/genética , Masculino , Camundongos , Camundongos Knockout , Fosfofrutoquinase-2/genética , Cultura Primária de Células , Proteínas de Ligação a RNA/genética , Ratos , Regulação para Cima
2.
Adv Exp Med Biol ; 1122: 73-100, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30937864

RESUMO

This review chapter describes the current knowledge about the nature of pericytes in the gut, their interaction with endothelial cells in blood vessels, and their pathophysiological functions in the setting of chronic liver disease. In particular, it focuses on the role of these vascular cell types and related molecular signaling pathways in pathological angiogenesis associated with liver disease and in the establishment of the gut-vascular barrier and the potential implications in liver disease through the gut-liver axis.


Assuntos
Trato Gastrointestinal/citologia , Neovascularização Patológica , Pericitos/citologia , Transdução de Sinais , Vasos Sanguíneos/citologia , Células Endoteliais/citologia , Humanos , Hepatopatias
3.
Sci Rep ; 7(1): 14791, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29093528

RESUMO

Development of portosystemic collateral vessels and gastroesophageal varices is responsible for the most serious clinical consequences of portal hypertension, but effective clinical therapies are limited. Here we developed and investigated the therapeutic potential of an innovative liposomally-formulated short-interfering RNA (siRNA) technology based on clinical stage components, capable to attenuate production of the endothelial kinase insert domain receptor (KDR), which controls portosystemic collateralization and contributes to disease progression and aggravation. These siRNAs were first validated in vitro, and then, their therapeutic potential on portosystemic collateralization and pathological angiogenesis was tested in vivo in mouse models of portal hypertension (portal vein-ligation). siRNAKDR-lipoplexes efficiently transported siRNAKDR to vascular endothelial cells in mesenteric microvenules and portal vein of portal hypertensive mice, where collaterogenesis and angiogenesis take place. This systemic treatment significantly downregulated pathological KDR overexpression, without causing complete KDR knockout, preserving homeostatic baseline KDR levels and thus limiting adverse effects. siRNAKDR-lipoplex-induced endothelial-specific KDR knockdown drastically reduced by 73% the portosystemic collateralization, and impaired the pathologic angiogenic potential of vascular endothelial cells at different levels (cell proliferation, sprouting and remodeling). Targeting endothelial KDR with therapeutic siRNAKDR-lipoplexes could be a promising and plausible treatment modality for attenuating the formation of portosystemic collaterals in a clinical setting.


Assuntos
Endotélio Vascular , Hipertensão Portal , Neovascularização Patológica , RNA Interferente Pequeno , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Animais , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Hipertensão Portal/genética , Hipertensão Portal/metabolismo , Hipertensão Portal/patologia , Hipertensão Portal/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
4.
Gut ; 66(7): 1306-1320, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-26984852

RESUMO

OBJECTIVE: Pathological neovascularisation is intimately involved in portal hypertension (PH). Here, we determined the contribution of vascular stem/progenitor cells (VSPCs) to neovessel growth in PH and whether the RNA-binding protein cytoplasmic polyadenylation element binding protein-4 (CPEB4) was behind the mechanism controlling VSPC function. DESIGN: To identify and monitor VSPCs in PH rats (portal vein-ligated), we used a combinatorial approach, including sphere-forming assay, assessment of self-renewal, 5-bromo-2'-desoxyuridine label retention technique, in vitro and in vivo stem/progenitor cell (SPC) differentiation and vasculogenic capability, cell sorting, as well as immunohistochemistry, immunofluorescence and confocal microscopy expression analysis. We also determined the role of CPEB4 on VSPC proliferation using genetically engineered mouse models. RESULTS: We demonstrated the existence in the mesenteric vascular bed of VSPCs displaying capability to form cellular spheres in suspension culture, self-renewal ability, expression of molecules commonly found in SPCs, slow-cycling features, in addition to other cardinal properties exhibited by SPCs, like capacity to differentiate into endothelial cells and pericytes with remarkable vasculogenic activity. Such VSPCs showed, after PH induction, an early switch in proliferation, and differentiated in vivo into endothelial cells and pericytes, contributing, structurally and functionally, to abnormal neovessel formation. Quantification of VSPC-dependent neovessel formation in PH further illustrated the key role played by VSPCs. We also demonstrated that CPEB4 regulates the proliferation of the activated VSPC progeny upon PH induction. CONCLUSIONS: These findings demonstrate that VSPC-derived neovessel growth (ie, vasculogenesis) and angiogenesis cooperatively stimulate mesenteric neovascularisation in PH and identify VSPC and CPEB4 as potential therapeutic targets.


Assuntos
Hipertensão Portal/patologia , Neovascularização Patológica , Proteínas de Ligação a RNA/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Proliferação de Células , Camundongos , Ratos
5.
Gastroenterology ; 150(4): 982-97.e30, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26627607

RESUMO

BACKGROUND & AIMS: Vascular endothelial growth factor (VEGF) regulates angiogenesis, yet therapeutic strategies to disrupt VEGF signaling can interfere with physiologic angiogenesis. In a search for ways to inhibit pathologic production or activities of VEGF without affecting its normal production or functions, we investigated the post-transcriptional regulation of VEGF by the cytoplasmic polyadenylation element-binding proteins CPEB1 and CPEB4 during development of portal hypertension and liver disease. METHODS: We obtained transjugular liver biopsies from patients with hepatitis C virus-associated cirrhosis or liver tissues removed during transplantation; healthy human liver tissue was obtained from a commercial source (control). We also performed experiments with male Sprague-Dawley rats and CPEB-deficient mice (C57BL6 or mixed C57BL6/129 background) and their wild-type littermates. Secondary biliary cirrhosis was induced in rats by bile duct ligation, and portal hypertension was induced by partial portal vein ligation. Liver and mesenteric tissues were collected and analyzed in angiogenesis, reverse transcription polymerase chain reaction, polyA tail, 3' rapid amplification of complementary DNA ends, Southern blot, immunoblot, histologic, immunohistochemical, immunofluorescence, and confocal microscopy assays. CPEB was knocked down with small interfering RNAs in H5V endothelial cells, and translation of luciferase reporters constructs was assessed. RESULTS: Activation of CPEB1 promoted alternative nuclear processing within noncoding 3'-untranslated regions of VEGF and CPEB4 messenger RNAs in H5V cells, resulting in deletion of translation repressor elements. The subsequent overexpression of CPEB4 promoted cytoplasmic polyadenylation of VEGF messenger RNA, increasing its translation; the high levels of VEGF produced by these cells led to their formation of tubular structures in Matrigel assays. We observed increased levels of CPEB1 and CPEB4 in cirrhotic liver tissues from patients, compared with control tissue, as well as in livers and mesenteries of rats and mice with cirrhosis or/and portal hypertension. Mice with knockdown of CPEB1 or CPEB4 did not overexpress VEGF or have signs of mesenteric neovascularization, and developed less-severe forms of portal hypertension after portal vein ligation. CONCLUSIONS: We identified a mechanism of VEGF overexpression in liver and mesentery that promotes pathologic, but not physiologic, angiogenesis, via sequential and nonredundant functions of CPEB1 and CPEB4. Regulation of CPEB4 by CPEB1 and the CPEB4 autoamplification loop induces pathologic angiogenesis. Strategies to block the activities of CPEBs might be developed to treat chronic liver and other angiogenesis-dependent diseases.


Assuntos
Hipertensão Portal/metabolismo , Cirrose Hepática Biliar/metabolismo , Cirrose Hepática/metabolismo , Neovascularização Patológica , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Regiões 3' não Traduzidas , Adulto , Animais , Estudos de Casos e Controles , Linhagem Celular , Doença Crônica , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Hipertensão Portal/genética , Hipertensão Portal/patologia , Cirrose Hepática/patologia , Cirrose Hepática/virologia , Cirrose Hepática Biliar/genética , Cirrose Hepática Biliar/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Poliadenilação , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transfecção , Fatores de Poliadenilação e Clivagem de mRNA/deficiência , Fatores de Poliadenilação e Clivagem de mRNA/genética
6.
Gut ; 64(4): 657-66, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24848263

RESUMO

OBJECTIVE: Antiangiogenic strategies have been proposed as a promising new approach for the therapy of portal hypertension and chronic liver disease. Pigment epithelium-derived factor (PEDF) is a powerful endogenous angiogenesis inhibitor whose role in portal hypertension remains unknown. Therefore, we aimed at determining the involvement of PEDF in cirrhotic portal hypertension and the therapeutic efficacy of its supplementation. DESIGN: PEDF expression profiling and its relationship with vascular endothelial growth factor (VEGF), neovascularisation and fibrogenesis was determined in bile duct-ligated (BDL) rats and human cirrhotic livers. The ability of exogenous PEDF overexpression by adenovirus-mediated gene transfer (AdPEDF) to inhibit angiogenesis, fibrogenesis and portal pressure was also evaluated in BDL rats, following prevention and intervention trials. RESULTS: PEDF was upregulated in cirrhotic human and BDL rat livers. PEDF and VEGF protein expression and localisation in mesentery and liver increased in parallel with portal hypertension progression, being closely linked in time and space with mesenteric neovascularisation and liver fibrogenesis in BDL rats. Furthermore, AdPEDF increased PEDF bioavailability in BDL rats, shifting the net balance in the local abundance of positive (VEGF) and negative (PEDF) angiogenesis drivers in favour of attenuation of portal hypertension-associated pathological neovascularisation. The antiangiogenic effects of AdPEDF targeted only pathological angiogenesis, without affecting normal vasculature, and were observed during early stages of disease. AdPEDF also significantly decreased liver fibrogenesis (through metalloproteinase upregulation), portosystemic collateralisation and portal pressure in BDL rats. CONCLUSIONS: This study provides compelling experimental evidence indicating that PEDF could be a novel therapeutic agent worthy of assessment in portal hypertension and cirrhosis.


Assuntos
Proteínas do Olho/fisiologia , Proteínas do Olho/uso terapêutico , Hipertensão Portal/etiologia , Hipertensão Portal/prevenção & controle , Cirrose Hepática/prevenção & controle , Neovascularização Patológica/prevenção & controle , Fatores de Crescimento Neural/fisiologia , Fatores de Crescimento Neural/uso terapêutico , Serpinas/fisiologia , Serpinas/uso terapêutico , Animais , Ductos Biliares , Humanos , Ligadura , Masculino , Pressão na Veia Porta , Ratos , Ratos Sprague-Dawley
7.
Hepatology ; 60(2): 633-47, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24390792

RESUMO

UNLABELLED: Pathological angiogenesis represents a critical hallmark for chronic liver diseases. Understanding the mechanisms regulating angiogenesis is essential to develop new therapeutic strategies that specifically target pathological angiogenesis without affecting physiological angiogenesis. Here we investigated the contribution and therapeutic impact of the endogenous angioinhibitor vasohibin-1 in portal hypertension and cirrhosis. The spatiotemporal expression profiling of vasohibin-1 and its relationship with vascular endothelial growth factor (VEGF), angiogenesis, and fibrogenesis was determined through the analysis of human cirrhotic liver specimens, widely accepted in vivo animal models of portal hypertension and cirrhosis, and in vitro angiogenesis assays. Effects of vasohibin-1 overexpression by adenoviral-mediated gene transfer on angiogenesis, fibrogenesis, and portal hypertension-associated hemodynamic alterations were also studied in rats. We found that vasohibin-1 and VEGF are up-regulated, in mesentery and liver, in cirrhotic and precirrhotic portal hypertensive rats and cirrhosis patients. Our results are consistent with vasohibin-1/VEGF cascades being spatially and temporally coordinated through a negative-feedback loop driving pathological angiogenesis. Paradoxically, further overexpression of vasohibin-1 by adenoviral gene transfer exerts multifold beneficial effects in portal hypertension and cirrhosis: reduction of pathologic angiogenesis, attenuation of liver fibrogenesis partly mediated through inhibition of hepatic stellate cell activation, and significant decreases in portocollateralization, splanchnic blood flow, portohepatic resistance, and portal pressure. The explanation for this apparent contradiction is that, unlike endogenous vasohibin-1, the ectopic overexpression is not regulated by VEGF and therefore disrupts the negative-feedback loop, thus generating constant, but lower levels of VEGF synthesis sufficient to maintain vascular homeostasis but not pathological angiogenesis. CONCLUSION: Our study provides evidence that vasohibin-1 regulates portal hypertension-associated pathological angiogenesis and highlights that increasing vasohibin-1 might be a promising novel therapeutic strategy for portal hypertension and cirrhosis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cirrose Hepática/metabolismo , Neovascularização Patológica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenoviridae/genética , Animais , Proteínas de Ciclo Celular/genética , Doença Crônica , Retroalimentação Fisiológica/fisiologia , Expressão Gênica/fisiologia , Células Estreladas do Fígado/metabolismo , Homeostase/fisiologia , Humanos , Hiperemia/metabolismo , Hiperemia/fisiopatologia , Hipertensão Portal/metabolismo , Hipertensão Portal/fisiopatologia , Cirrose Hepática/fisiopatologia , Masculino , Neovascularização Patológica/fisiopatologia , Ratos , Ratos Sprague-Dawley , Circulação Esplâncnica/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética
8.
J Hepatol ; 52(4): 529-39, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20206401

RESUMO

BACKGROUND & AIMS: Splenomegaly is a frequent hallmark of portal hypertension that, in some cases, can be very prominent and cause symptoms like abdominal pain, splenic infarction, and cytopenia. This study characterizes the pathogenetic mechanisms leading to spleen enlargement in portal hypertensive rats and focuses on mTOR pathway as a potential modulator of splenomegaly in portal hypertension. METHODS: Characterization of splenomegaly was performed by histological, hematological, immunohistochemical and Western blot analyses in rats with portal hypertension induced by portal vein ligation, and compared with sham-operated animals. The contribution of the mTOR signaling pathway to splenomegaly was determined in rats with fully developed portal hypertension and control rats by treatment with rapamycin or vehicle. RESULTS: Our results illustrate that splenomegaly in portal hypertensive rats arises as a consequence of the interplay of several factors, including not only spleen congestion, as traditionally thought, but also enlargement and hyperactivation of the splenic lymphoid tissue, as well as increased angiogenesis and fibrogenesis. Since mTOR signaling plays a central role in immunological processes, angiogenesis and fibrogenesis, we next determined the involvement of mTOR in splenomegaly. Interestingly, mTOR signaling was overactivated in the spleen of portal hypertensive rats, and mTOR blockade by rapamycin profoundly ameliorated splenomegaly, causing a 44% decrease in spleen size. This effect was most likely accounted for the inhibitory action of rapamycin on lymphocyte proliferation, neovascularization and fibrosis. CONCLUSIONS: These findings shed light on the pathogenesis of splenomegaly in portal hypertension, and identify mTOR signaling as a potential target for therapeutic intervention in this disease.


Assuntos
Hipertensão Portal/metabolismo , Hipertensão Portal/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Esplenomegalia/metabolismo , Esplenomegalia/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Fibrose , Hematócrito , Hipertensão Portal/tratamento farmacológico , Imunossupressores/farmacologia , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Masculino , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Baço/irrigação sanguínea , Baço/metabolismo , Baço/patologia , Esplenomegalia/tratamento farmacológico , Serina-Treonina Quinases TOR
9.
Hepatology ; 49(4): 1245-56, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19137587

RESUMO

UNLABELLED: Portal hypertension, the most important complication in patients with cirrhosis of the liver, is a serious and life-threatening disease for which there are few therapeutic options. Because angiogenesis is a pathological hallmark of portal hypertension, the goal of this study was to determine the effects of sorafenib-a potent inhibitor of proangiogenic vascular endothelial growth factor receptor 2 (VEGFR-2), platelet-derived growth factor receptor beta (PDGFR-beta), and Raf kinases-on splanchnic, intrahepatic, systemic, and portosystemic collateral circulations in two different experimental models of portal hypertension: rats with prehepatic portal hypertension induced by partial portal vein ligation and rats with intrahepatic portal hypertension and secondary biliary cirrhosis induced by bile duct ligation. Such a comprehensive approach is necessary for any translational research directed toward defining the efficacy and potential clinical application of new therapeutic agents. Sorafenib administered orally once a day for 2 weeks in experimental models of portal hypertension and cirrhosis effectively inhibited VEGF, PDGF, and Raf signaling pathways, and produced several protective effects by inducing an approximately 80% decrease in splanchnic neovascularization and a marked attenuation of hyperdynamic splanchnic and systemic circulations, as well as a significant 18% decrease in the extent of portosystemic collaterals. In cirrhotic rats, sorafenib treatment also resulted in a 25% reduction in portal pressure, as well as a remarkable improvement in liver damage and intrahepatic fibrosis, inflammation, and angiogenesis. Notably, beneficial effects of sorafenib against tissue damage and inflammation were also observed in splanchnic organs. CONCLUSION: Taking into account the limitations of translating animal study results into humans, we believe that our findings will stimulate consideration of sorafenib as an effective therapeutic agent in patients suffering from advanced portal hypertension.


Assuntos
Benzenossulfonatos/uso terapêutico , Hipertensão Portal/tratamento farmacológico , Cirrose Hepática/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Circulação Esplâncnica/efeitos dos fármacos , Animais , Benzenossulfonatos/farmacologia , Circulação Colateral/efeitos dos fármacos , Enterite/tratamento farmacológico , Enterite/etiologia , Heme Oxigenase-1/metabolismo , Hepatite/tratamento farmacológico , Hepatite/etiologia , Hipertensão Portal/etiologia , Fígado/metabolismo , Cirrose Hepática/complicações , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Niacinamida/análogos & derivados , Óxido Nítrico Sintase Tipo III/metabolismo , Compostos de Fenilureia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Sorafenibe
10.
J Hepatol ; 50(2): 296-305, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19070926

RESUMO

BACKGROUND/AIMS: Angiogenesis is a pathological hallmark of portal hypertension. Although VEGF is considered to be the most important proangiogenic factor in neoangiogenesis, this process requires the coordinated action of a variety of factors. Identification of novel molecules involved in angiogenesis is highly relevant, since they may represent potential new targets to suppress pathological neovascularization in angiogenesis-related diseases like portal hypertension. The apelin/APJ signaling pathway plays a crucial role in angiogenesis. Therefore, we determined whether the apelin system modulates angiogenesis-driven processes in portal hypertension. METHODS: Partial portal vein-ligated rats were treated with the APJ antagonist F13A for seven days. Splanchnic neovascularization and expression of angiogenesis mediators (Western blotting) was determined. Portosystemic collateral formation (microspheres), and hemodynamic parameters (flowmetry) were also assessed. RESULTS: Apelin and its receptor APJ were overexpressed in the splanchnic vasculature of portal hypertensive rats. F13A effectively decreased, by 52%, splanchnic neovascularization and expression of proangiogenic factors VEGF, PDGF and angiopoietin-2 in portal hypertensive rats. F13A also reduced, by 35%, the formation of portosystemic collateral vessels. CONCLUSIONS: This study provides the first experimental evidence showing that the apelin/APJ system contributes to portosystemic collateralization and splanchnic neovascularization in portal hypertensive rats, presenting a potential novel therapeutic target for portal hypertension.


Assuntos
Proteínas de Transporte/fisiologia , Neovascularização Fisiológica , Sistema Porta/fisiopatologia , Transdução de Sinais/fisiologia , Circulação Esplâncnica , Animais , Apelina , Receptores de Apelina , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Circulação Colateral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Heme Oxigenase (Desciclizante)/genética , Hipertensão Portal/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Cadeias Leves de Miosina/genética , Óxido Nítrico Sintase Tipo III/genética , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
11.
J Hepatol ; 49(1): 43-51, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18457899

RESUMO

BACKGROUND/AIMS: Splanchnic vasodilation initiates the hyperdynamic syndrome in portal hypertension. We aimed to explore molecular mechanisms involved in the development of mesenteric vasodilation in portal hypertension. METHODS: Superior mesenteric artery (SMA) samples from portal vein ligated (PVL) and sham rats were compared in a time course experiment using DNA microarrays. Selected genes were quantified by qRT-PCR in PVL and cirrhotic rats. Inmunohistochemistry of tyrosine hydroxylase (Th) and norepinephrine was assessed in SMA sections of PVL and sham rats. Western blot analysis of Th, dopamine beta-hydroxylase (Dbh) and synaptosome-associated protein (Snap-25) was performed in SMA and jejunum samples from the animal models. RESULTS: Fifty differentially expressed genes implicated in neurotransmission, especially adrenergic, were detected in SMA samples from PVL rats. Sequential analysis showed a profound down-regulation at 14 days in PVL rats. These down-regulated genes were confirmed by RT-PCR in SMA from PVL and cirrhotic rats. Th and NE detection by immunohistochemistry was reduced in PVL compared to sham. Th, Dbh and Snap-25 expression was lower in SMA from 14-day PVL and cirrhotic rats compared to sham and control rats, respectively. CONCLUSIONS: Genetic down-regulation of genes related to the adrenergic system might have a role in splanchnic vasodilation of portal hypertension.


Assuntos
Perfilação da Expressão Gênica , Hipertensão Portal/genética , Hipertensão Portal/fisiopatologia , Norepinefrina/fisiologia , Circulação Esplâncnica/fisiologia , Vasodilatação/fisiologia , Animais , Tetracloreto de Carbono/toxicidade , Modelos Animais de Doenças , Dopamina beta-Hidroxilase/genética , Dopamina beta-Hidroxilase/metabolismo , Regulação para Baixo/genética , Hipertensão Portal/induzido quimicamente , Imuno-Histoquímica , Ligadura , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/fisiopatologia , Masculino , Artéria Mesentérica Superior/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Veia Porta , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
12.
J Cell Mol Med ; 12(5A): 1690-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18194463

RESUMO

BACKGROUND: Angiogenesis is an important determinant of the pathophysiology of portal hypertension contributing to the formation of portosystemic collateral vessels and the hyperdynamic splanchnic circulation associated to this syndrome. Somatostatin and its analogues, like octreotide, have been shown to be powerful inhibitors of experimental angiogenesis. AIM: To determine whether octreotide has angioinhibitory effects in portal hypertensive rats. METHODS: Partial portal vein-ligated (PPVL) rats were treated with octreotide or vehicle during 4 or 7 days. Splanchnic neovascularization and VEGF expression were determined by histological analysis and western blotting. Expression of the somatostatin receptor subtype 2 (SSTR2), which mediates the anti-angiogenic effects of octreotide, was also analyzed. Formation of portosystemic collaterals (radioactive microspheres) and hemodynamic parameters were also measured. RESULTS: Octreotide treatment during 4 days markedly and significantly decreased splanchnic neovascularization, VEGF expression by 63% and portal pressure by 15%, whereas portosystemic collateralization and splanchnic blood flow were not modified. After 1 week of octreotide injection, portal pressure was reduced by 20%, but inhibition of angiogenesis escaped from octreotide therapy, a phenomenon that could be related to the finding that expression of SSTR2 receptor decreased progressively (up to 78% reduction) during the evolution of portal hypertension. CONCLUSION: This study provides the first experimental evidence showing that octreotide may be an effective anti-angiogenic therapy early after induction of portal hypertension, but not in advanced stages most likely due to SSTR2 down-regulation during the progression of portal hypertension in rats. These findings shed light on new mechanisms of action of octreotide in portal hypertension.


Assuntos
Hipertensão Portal/tratamento farmacológico , Hipertensão Portal/patologia , Neovascularização Patológica/tratamento farmacológico , Octreotida/uso terapêutico , Somatostatina/análogos & derivados , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Hemodinâmica/efeitos dos fármacos , Hipertensão Portal/genética , Hipertensão Portal/metabolismo , Masculino , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Octreotida/química , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Hepatology ; 46(4): 1208-17, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17654489

RESUMO

UNLABELLED: Vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) pathways are crucial to angiogenesis, a process that contributes significantly to the pathogenesis of portal hypertension. This study determined the effects of inhibition of VEGF and/or PDGF signaling on hyperdynamic splanchnic circulation and portosystemic collateralization in rats with completely established portal hypertension, thus mimicking the situation in patients. Portal vein-ligated rats were treated with rapamycin (VEGF signaling inhibitor), Gleevec (PDGF signaling inhibitor), or both simultaneously when portal hypertension was already fully developed. Hemodynamic studies were performed by transit-time flowmetry. The extent of portosystemic collaterals was measured by radioactive microspheres. The expression of angiogenesis mediators was determined by Western blotting and immunohistochemistry. Combined inhibition of VEGF and PDGF signaling significantly reduced splanchnic neovascularization (i.e., CD31 and VEGFR-2 expression) and pericyte coverage of neovessels (that is, alpha-smooth muscle actin and PDGFR-beta expression) and translated into hemodynamic effects as marked as a 40% decrease in portal pressure, a 30% decrease in superior mesenteric artery blood flow, and a 63% increase in superior mesenteric artery resistance, yielding a significant reversal of the hemodynamic changes provoked by portal hypertension in rats. Portosystemic collateralization was reduced as well. CONCLUSIONS: Our results provide new insights into how angiogenesis regulates portal hypertension by demonstrating that the maintenance of increased portal pressure, hyperkinetic circulation, splanchnic neovascularization, and portosystemic collateralization is regulated by VEGF and PDGF in portal hypertensive rats. Importantly, these findings also suggest that an extended antiangiogenic strategy (that is, targeting VEGF/endothelium and PDGF/pericytes) may be a novel approach to the treatment of portal hypertension.


Assuntos
Hipertensão Portal/fisiopatologia , Neovascularização Patológica/metabolismo , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Circulação Esplâncnica/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Benzamidas , Modelos Animais de Doenças , Quimioterapia Combinada , Hipertensão Portal/tratamento farmacológico , Hipertensão Portal/metabolismo , Mesilato de Imatinib , Imunossupressores/uso terapêutico , Ligadura , Masculino , Artéria Mesentérica Superior/fisiopatologia , Neovascularização Patológica/tratamento farmacológico , Piperazinas/uso terapêutico , Fator de Crescimento Derivado de Plaquetas/metabolismo , Veia Porta/fisiopatologia , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/fisiologia , Transdução de Sinais/fisiologia , Sirolimo/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Gut ; 56(4): 560-4, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16854998

RESUMO

BACKGROUND: Recent studies have shown the presence of vascular endothelial growth factor (VEGF)-dependent splanchnic angiogenesis in experimental models of portal hypertension, and the role of such neovascularisation on the development of both portosystemic collaterals and hyperdynamic splanchnic circulation. However, the mechanisms modulating angiogenesis in portal hypertension are unknown. Experimental evidence indicates that NAD(P)H oxidase is required for VEGF-induced angiogenesis. Interestingly, we have recently shown that splanchnic NAD(P)H oxidase activity is significantly increased in portal hypertensive rats. Therefore, it could be possible that activated NAD(P)H oxidases modulate angiogenesis in portal hypertension. AIM: To determine the effects of chronic NAD(P)H oxidase inhibition on angiogenesis and splanchnic haemodynamics in portal hypertensive rats. METHODS: Partial portal vein-ligated and sham-operated rats were treated with the NAD(P)H oxidase inhibitor apocynin, or with vehicle for 5 days. Then, the expression of angiogenesis markers (western blotting), the formation of portosystemic collaterals (radioactive microspheres) and the production of superoxide anion (lucigenin-enhanced chemiluminescence) were determined. Mean arterial pressure, portal pressure, and superior mesenteric arterial blood flow and resistance were also measured. RESULTS: In portal hypertensive rats, NAD(P)H oxidase blockade significantly decreased portosystemic collateral formation, and superior mesenteric arterial flow. It also reduced the splanchnic expression of VEGF, VEGF receptor-2 and CD31, and attenuated the increased production of superoxide, compared with vehicle. CONCLUSIONS: NAD(P)H oxidase plays an important role in experimental portal hypertension, modulating splanchnic angiogenesis, the formation of portosystemic collaterals and the development of splanchnic hyperdynamic circulation. These results suggest that NAD(P)H oxidase may represent a new target in the treatment of portal hypertension.


Assuntos
Hiperemia/enzimologia , Hipertensão Portal/enzimologia , NADPH Oxidases/fisiologia , Neovascularização Patológica/enzimologia , Circulação Esplâncnica , Acetofenonas/farmacologia , Indutores da Angiogênese/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Circulação Colateral/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hiperemia/fisiopatologia , Hiperemia/prevenção & controle , Hipertensão Portal/patologia , Hipertensão Portal/fisiopatologia , Masculino , Artéria Mesentérica Superior/fisiopatologia , NADPH Oxidases/antagonistas & inibidores , Neovascularização Patológica/fisiopatologia , Neovascularização Patológica/prevenção & controle , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Circulação Esplâncnica/efeitos dos fármacos , Superóxidos/metabolismo
15.
J Hepatol ; 44(6): 1033-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16458992

RESUMO

BACKGROUND/AIMS: The pathophysiological significance of heme oxygenase-1 up-regulation in portal hypertension is not completely understood. In this study, we determined the role of heme oxygenase-1 on oxidative stress, inflammation, angiogenesis, and splanchnic hemodynamics in rats with portal hypertension induced by partial portal vein ligation. METHODS: Rats were treated with the heme oxygenase inhibitor SnMP or vehicle for 7 days. Then, oxidative stress was quantified by superoxide anion production, and inflammatory response was assessed by immunofluorescence. Expression of angiogenesis mediators was determined by western blotting, and the extent of portosystemic collaterals by radioactive microspheres. Hemodynamic studies were performed by flowmetry. RESULTS: Oxidative stress was significantly increased in the mesentery of portal hypertensive rats, as compared with sham-operated controls. In portal hypertensive rats, chronic heme oxygenase inhibition (1) potentiated oxidative stress and inflammation, (2) significantly decreased VEGF expression, without modifying the extent of collaterals or the splanchnic neovascularization, and (3) significantly decreased superior mesenteric artery blood flow and portal pressure. CONCLUSIONS: This study demonstrates that heme oxygenase plays an important (beneficial) role attenuating oxidative stress and inflammation, but it also plays a detrimental role in stimulating VEGF production, and contributing to the development of hyperdynamic splanchnic circulation in rats with portal hypertension.


Assuntos
Heme Oxigenase-1/fisiologia , Hipertensão Portal/enzimologia , Inflamação/enzimologia , Estresse Oxidativo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Masculino , Metaloporfirinas/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Protoporfirinas/farmacologia , Ratos , Ratos Sprague-Dawley , Regulação para Cima
16.
J Hepatol ; 43(1): 98-103, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15893841

RESUMO

BACKGROUND/AIMS: Portal hypertension is characterized by the development of a hyperdynamic splanchnic circulation. To determine whether this process is angiogenesis-dependent, we assessed the effects of SU5416, a specific inhibitor of VEGF receptor-2, in portal hypertensive rats. METHODS: Rats with portal hypertension induced by partial portal vein ligation were treated with SU5416 or vehicle during 5 days. Then, hemodynamic studies were performed using radioactive microspheres. Protein expressions of CD31, VEGF receptor-2 and VEGF were also determined by Western blotting. RESULTS: Treatment of portal hypertensive rats with SU5416 resulted in a significant and marked decrease (by 44%) in portal venous inflow, and increases in splanchnic arteriolar resistance (by 68%) and portal venous resistance (by 93%). In addition, SU5416 administration significantly inhibited the formation of portal-systemic collateral vessels (52% inhibition), as well as the splanchnic CD31 and VEGF receptor-2 protein expressions in portal hypertensive rats, compared with those receiving vehicle. CONCLUSIONS: This study demonstrates that the development of hyperdynamic splanchnic circulation and the formation of portal-systemic collateral vessels in portal hypertensive rats are angiogenesis-dependent processes that can be markedly inhibited by blockade of the VEGF signaling pathway. Therefore, modulation of angiogenesis may represent a potential target in the treatment of portal hypertension.


Assuntos
Circulação Colateral , Hipertensão Portal/complicações , Hipertensão Portal/fisiopatologia , Neovascularização Patológica/etiologia , Sistema Porta/fisiopatologia , Circulação Esplâncnica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Arteríolas/fisiopatologia , Circulação Colateral/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Indóis/farmacologia , Masculino , Molécula-1 de Adesão Celular Endotelial a Plaquetas/efeitos dos fármacos , Veia Porta/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Pirróis/farmacologia , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Circulação Esplâncnica/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Resistência Vascular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...