Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 523(7558): 92-5, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-25970250

RESUMO

The tumour microenvironment may contribute to tumorigenesis owing to mechanical forces such as fibrotic stiffness or mechanical pressure caused by the expansion of hyper-proliferative cells. Here we explore the contribution of the mechanical pressure exerted by tumour growth onto non-tumorous adjacent epithelium. In the early stage of mouse colon tumour development in the Notch(+)Apc(+/1638N) mouse model, we observed mechanistic pressure stress in the non-tumorous epithelial cells caused by hyper-proliferative adjacent crypts overexpressing active Notch, which is associated with increased Ret and ß-catenin signalling. We thus developed a method that allows the delivery of a defined mechanical pressure in vivo, by subcutaneously inserting a magnet close to the mouse colon. The implanted magnet generated a magnetic force on ultra-magnetic liposomes, stabilized in the mesenchymal cells of the connective tissue surrounding colonic crypts after intravenous injection. The magnetically induced pressure quantitatively mimicked the endogenous early tumour growth stress in the order of 1,200 Pa, without affecting tissue stiffness, as monitored by ultrasound strain imaging and shear wave elastography. The exertion of pressure mimicking that of tumour growth led to rapid Ret activation and downstream phosphorylation of ß-catenin on Tyr654, imparing its interaction with the E-cadherin in adherens junctions, and which was followed by ß-catenin nuclear translocation after 15 days. As a consequence, increased expression of ß-catenin-target genes was observed at 1 month, together with crypt enlargement accompanying the formation of early tumorous aberrant crypt foci. Mechanical activation of the tumorigenic ß-catenin pathway suggests unexplored modes of tumour propagation based on mechanical signalling pathways in healthy epithelial cells surrounding the tumour, which may contribute to tumour heterogeneity.


Assuntos
Carcinogênese/patologia , Neoplasias do Colo/fisiopatologia , Pressão , Microambiente Tumoral , beta Catenina/genética , Transporte Ativo do Núcleo Celular , Animais , Células Epiteliais/citologia , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Imãs , Masculino , Nanopartículas Metálicas , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , beta Catenina/metabolismo
2.
Hepatology ; 61(3): 1041-55, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25043701

RESUMO

UNLABELLED: Liver fibrosis expanding from portal tracts and vascular remodeling are determinant factors in the progression of liver diseases to cirrhosis. In the present study, we examined the potential contribution of portal myofibroblasts (PMFs) to the vascular changes leading to cirrhosis. The analyses of liver cells based on the transcriptome of rat PMFs, compared to hepatic stellate cell HSC-derived myofibroblasts in culture, identified collagen, type XV, alpha 1 (COL15A1) as a marker of PMFs. Normal liver contained rare COL15A1-immunoreactive cells adjacent to the bile ducts and canals of Hering in the portal area. A marked increase in COL15A1 expression occurred together with that of the endothelial marker, von Willebrand factor, in human and rat liver tissue, at advanced stages of fibrosis caused by either biliary or hepatocellular injury. In cirrhotic liver, COL15A1-expressing PMFs adopted a perivascular distribution outlining vascular capillaries proximal to reactive ductules, within large fibrotic septa. The effect of PMFs on endothelial cells (ECs) was evaluated by in vitro and in vivo angiogenesis assays. PMF-conditioned medium increased the migration and tubulogenesis of liver ECs as well as human umbilical vein ECs and triggered angiogenesis within Matrigel plugs in mice. In coculture, PMFs developed intercellular junctions with ECs and enhanced the formation of vascular structures. PMFs released vascular endothelial growth factor (VEGF)A-containing microparticles, which activated VEGF receptor 2 in ECs and largely mediated their proangiogenic effect. Cholangiocytes potentiated the angiogenic properties of PMFs by increasing VEGFA expression and microparticle shedding in these cells. CONCLUSION: PMFs are key cells in hepatic vascular remodeling. They signal to ECs through VEGFA-laden microparticles and act as mural cells for newly formed vessels, driving scar progression from portal tracts into the parenchyma.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Cirrose Hepática/etiologia , Fígado/citologia , Miofibroblastos/fisiologia , Remodelação Vascular , Animais , Colágeno/análise , Humanos , Cirrose Hepática/patologia , Masculino , Neovascularização Fisiológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/fisiologia
3.
Cancer Res ; 73(13): 3974-86, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23633480

RESUMO

Insulin receptor (IR) exists as two isoforms resulting from the alternative splicing of IR pre-mRNA. IR-B promotes the metabolic effects of insulin, whereas IR-A rather signals proliferative effects. IR-B is predominantly expressed in the adult liver. Here, we show that the alternative splicing of IR pre-mRNA is dysregulated in a panel of 85 human hepatocellular carcinoma (HCC) while being normal in adjacent nontumor liver tissue. An IR-B to IR-A switch is frequently observed in HCC tumors regardless of tumor etiology. Using pharmacologic and siRNA approaches, we show that the autocrine or paracrine activation of the EGF receptor (EGFR)/mitogen-activated protein/extracellular signal-regulated kinase pathway increases the IR-A:IR-B ratio in HCC cell lines, but not in normal hepatocytes, by upregulating the expression of the splicing factors CUGBP1, hnRNPH, hnRNPA1, hnRNPA2B1, and SF2/ASF. In HCC tumors, there is a significant correlation between the expression of IR-A and that of splicing factors. Dysregulation of IR pre-mRNA splicing was confirmed in a chemically induced model of HCC in rat but not in regenerating livers after partial hepatectomy. This study identifies a mechanism responsible for the generation of mitogenic IR-A and provides a novel interplay between IR and EGFR pathways in HCC. Increased expression of IR-A during neoplastic transformation of hepatocytes could mediate some of the adverse effects of hyperinsulinemia on HCC.


Assuntos
Antígenos CD/genética , Carcinoma Hepatocelular/metabolismo , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas Experimentais/metabolismo , Receptor de Insulina/genética , Animais , Antígenos CD/metabolismo , Proteínas CELF1 , Transformação Celular Neoplásica/metabolismo , Expressão Gênica , Células Hep G2 , Hepatócitos/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Insulina/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Regeneração Hepática , Sistema de Sinalização das MAP Quinases , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ratos , Receptor de Insulina/metabolismo , Fatores de Processamento de Serina-Arginina
4.
Hepatology ; 58(4): 1401-12, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23696511

RESUMO

UNLABELLED: Alterations in apical junctional complexes (AJCs) have been reported in genetic or acquired biliary diseases. The vitamin D nuclear receptor (VDR), predominantly expressed in biliary epithelial cells in the liver, has been shown to regulate AJCs. The aim of our study was thus to investigate the role of VDR in the maintenance of bile duct integrity in mice challenged with biliary-type liver injury. Vdr(-/-) mice subjected to bile duct ligation (BDL) displayed increased liver damage compared to wildtype BDL mice. Adaptation to cholestasis, ascertained by expression of genes involved in bile acid metabolism and tissue repair, was limited in Vdr(-/-) BDL mice. Furthermore, evaluation of Vdr(-/-) BDL mouse liver tissue sections indicated altered E-cadherin staining associated with increased bile duct rupture. Total liver protein analysis revealed that a truncated form of E-cadherin was present in higher amounts in Vdr(-/-) mice subjected to BDL compared to wildtype BDL mice. Truncated E-cadherin was also associated with loss of cell adhesion in biliary epithelial cells silenced for VDR. In these cells, E-cadherin cleavage occurred together with calpain 1 activation and was prevented by the silencing of calpain 1. Furthermore, VDR deficiency led to the activation of the epidermal growth factor receptor (EGFR) pathway, while EGFR activation by EGF induced both calpain 1 activation and E-cadherin cleavage in these cells. Finally, truncation of E-cadherin was blunted when EGFR signaling was inhibited in VDR-silenced cells. CONCLUSION: Biliary-type liver injury is exacerbated in Vdr(-/-) mice by limited adaptive response and increased bile duct rupture. These results indicate that loss of VDR restricts the adaptation to cholestasis and diminishes bile duct integrity in the setting of biliary-type liver injury.


Assuntos
Sistema Biliar/patologia , Colestase/fisiopatologia , Células Epiteliais/patologia , Junções Intercelulares/patologia , Fígado/fisiopatologia , Receptores de Calcitriol/deficiência , Sequência de Aminoácidos , Animais , Ductos Biliares/fisiopatologia , Caderinas/análise , Caderinas/fisiologia , Calpaína/fisiologia , Colestase/patologia , Modelos Animais de Doenças , Receptores ErbB/fisiologia , Ligadura , Fígado/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Receptores de Calcitriol/fisiologia
5.
J Hepatol ; 57(1): 108-15, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22414764

RESUMO

BACKGROUND & AIMS: Sorafenib is the standard of care for the treatment of advanced hepatocellular carcinoma (HCC). However, primary and acquired resistance is observed in patients. We examined whether gefitinib, which inhibits both epidermal growth factor receptor (EGFR) and HER-3 phosphorylation, could improve HCC cell response to sorafenib. METHODS: Sorafenib and gefitinib were tested in HCC tumor xenografts and in sorafenib-sensitive and sorafenib-resistant HCC cell lines. Biomarkers relevant to the HER system were analyzed by Western blotting and ELISA. RNA interference was used to downregulate the HER system. Amphiregulin concentrations were measured by ELISA in sera from patients under sorafenib treatment. RESULTS: Sorafenib combined with gefitinib significantly inhibited tumor growth in mice and reduced cell viability in vitro compared to single agents. In cell lines cultured in 10% serum or treated with EGF, sorafenib alone inhibited phospho-STAT3 while it maintained or even increased phospho-ERK and/or phospho-AKT. The paradoxical effects of sorafenib were prevented by gefitinib or by downregulation of EGFR and HER-3 expression. In cells with acquired resistance to sorafenib, aberrant activation of EGFR/HER-3 receptors as well as overexpression of several EGFR ligands were observed. These enhanced autocrine/paracrine loops led to the constitutive activation of ERK and AKT and conferred increased sensitivity to gefitinib. Increased serum concentrations of amphiregulin were observed in 10 out of 14 patients under sorafenib treatment compared to baselines. CONCLUSIONS: Signaling pathways controlled by EGFR and HER-3 restrict sorafenib effects both in naive and sorafenib-resistant HCC cells. Consequently, gefitinib cooperates with sorafenib to increase antiproliferative response and to prevent resistance.


Assuntos
Antineoplásicos/farmacologia , Benzenossulfonatos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Receptores ErbB/metabolismo , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Piridinas/farmacologia , Receptor ErbB-3/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Anfirregulina , Animais , Antineoplásicos/uso terapêutico , Benzenossulfonatos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Família de Proteínas EGF , Receptores ErbB/antagonistas & inibidores , Feminino , Gefitinibe , Glicoproteínas/metabolismo , Células Hep G2 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Niacinamida/análogos & derivados , Compostos de Fenilureia , Piridinas/uso terapêutico , Quinazolinas/farmacologia , Sorafenibe , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Gastroenterology ; 142(7): 1581-91.e6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22370478

RESUMO

BACKGROUND & AIMS: Patients with cystic fibrosis (CF) have poorly defined defects in biliary function. We evaluated the effects of cystic fibrosis transmembrane conductance regulator (CFTR) deficiency on the enterohepatic disposition of bile acids (BAs). METHODS: Bile secretion and BA homeostasis were investigated in Cftr(tm1Unc) (Cftr-/-) and CftrΔF508 (ΔF508) mice. RESULTS: Cftr-/- and ΔF508 mice did not grow to normal size, but did not have liver abnormalities. The gallbladders of Cftr-/- mice were enlarged and had defects in emptying, based on (99m)technetium-mebrofenin scintigraphy or post-prandial variations in gallbladder volume; gallbladder contraction in response to cholecystokinin-8 was normal. Cftr-/- mice had abnormal gallbladder bile and duodenal acidity, and overexpressed the vasoactive intestinal peptide-a myorelaxant factor for the gallbladder. The BA pool was larger in Cftr-/- than wild-type mice, although there were no differences in fecal loss of BAs. Amounts of secondary BAs in portal blood, liver, and bile of Cftr-/- mice were much lower than normal. Expression of genes that are induced by BAs, including fibroblast growth factor-15 and BA transporters, was lower in the ileum but higher in the gallbladders of Cftr-/- mice, compared with wild-type mice, whereas enzymes that synthesize BA were down-regulated in livers of Cftr-/- mice. This indicates that BAs underwent a cholecystohepatic shunt, which was confirmed using cholyl-(Ne-NBD)-lysine as a tracer. In Cftr-/- mice, cholecystectomy reversed most changes in gene expression and partially restored circulating levels of secondary BAs. The ΔF508 mice overexpressed vasoactive intestinal peptide and had defects in gallbladder emptying and in levels of secondary BAs, but these features were less severe than in Cftr-/- mice. CONCLUSIONS: Cftr-/- and CftrΔF508 mice have defects in gallbladder emptying that disrupt enterohepatic circulation of BAs. These defects create a shunt pathway that restricts the amount of toxic secondary BAs that enter the liver.


Assuntos
Ácidos e Sais Biliares/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Esvaziamento da Vesícula Biliar/fisiologia , Homeostase/fisiologia , Animais , Bile , Colecistectomia , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Proteomics ; 10(5): 1017-28, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20049859

RESUMO

In chronic liver diseases, the accumulation of extracellular matrix leading to fibrosis is caused by myofibroblasts, the origins of which are debatable. We performed a comparative proteomic study to identify markers and gain insight into distinct functions of myofibroblasts derived either from hepatic stellate cells (HSCs) or from portal mesenchymal cells. After isolation from normal liver and culture in similar conditions, myofibroblastic HSCs (MF-HSCs) presented enlarged cytoplasms whereas portal myofibroblasts (PMFs) were more proliferative, and formed more stress fibers. The two cell types were subjected to comparative analyses by 2-D MS/MS. Six proteins were overexpressed in PMFs, with myofibroblast-related typical functions. Among them, cofilin-1 showed the greatest difference in expression and a lower pI than expected. Immunoblot demonstrated higher levels of phosphorylation, a modification of the protein implicated in stress fiber formation. Eleven proteins, mostly involved in stress response, were overexpressed in MF-HSCs. Cytoglobin had the highest level of overexpression, as confirmed by reverse transcription quantitative real-time PCR, immunoblot and immunocytochemical analyses. These results identify cytoglobin as the best marker for distinguishing MF-HSCs from PMFs and suggest different functions for the two cell populations in the liver wound healing response, with a prominent role for PMFs in scar formation.


Assuntos
Fibroblastos/metabolismo , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Sistema Porta/patologia , Proteômica/métodos , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Citoglobina , Eletroforese em Gel Bidimensional , Fibroblastos/patologia , Perfilação da Expressão Gênica , Globinas/química , Globinas/metabolismo , Células Estreladas do Fígado/patologia , Dados de Sequência Molecular , Peptídeos/química , Proteoma/química , Proteoma/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Am J Pathol ; 175(2): 627-35, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19628767

RESUMO

The insulin-like growth factor type 1 receptor (IGF-1R) controls aging and cellular stress, both of which play major roles in liver disease. Stimulation of insulin-like growth factor signaling can generate cell death in vitro. Here, we tested whether IGF-1R contributes to stress insult in the liver. Cholestatic liver injury was induced by bile duct ligation in control and liver-specific IGF-1R knockout (LIGFREKO) mice. LIGFREKO mice displayed less bile duct ligation-induced hepatocyte damage than controls, while no differences in bile acid serum levels or better adaptation to cholestasis by efflux transporters were found. We therefore tested whether stress pathways contributed to this phenomenon; oxidative stress, ascertained by both malondialdehyde content and heme oxygenase-1 expression, was similar in knockout and control animals. However, together with a lower level of eukaryotic initiation factor-2 alpha phosphorylation, the endoplasmic reticulum stress protein CHOP and its downstream pro-apoptotic target Bax were induced to lesser extents in LIGFREKO mice than in controls. Expression levels of cytokeratin 19, transforming growth factor-beta1, alpha-smooth muscle actin, and collagen alpha1(I) in LIGFREKO mice were all lower than in controls, indicating reduced ductular and fibrogenic responses and increased cholestasis tolerance in these mutants. This stress resistance phenotype was also evidenced by longer post-bile duct ligation survival in mutants than controls. These results indicate that IGF-1R contributes to cholestatic liver injury, and suggests the involvement of both CHOP and Bax in this process.


Assuntos
Colestase/patologia , Hepatopatias/patologia , Receptor IGF Tipo 1/metabolismo , Estresse Fisiológico , Animais , Colestase/complicações , Colestase/genética , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Deleção de Genes , Hepatócitos/patologia , Hepatopatias/etiologia , Camundongos , Camundongos Transgênicos , Receptor IGF Tipo 1/genética , Fator de Transcrição CHOP/metabolismo , Proteína X Associada a bcl-2/metabolismo
9.
Am J Pathol ; 174(3): 869-80, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19234136

RESUMO

Ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) anchors and regulates apical membrane proteins in epithelia. EBP50 is inducible by estrogen and may affect cell proliferation, although this latter function remains unclear. The goal of this study was to determine whether EBP50 was implicated in the ductular reaction that occurs in liver disease. EBP50 expression was examined in normal human liver, in human cholangiopathies (ie, cystic fibrosis, primary biliary cirrhosis, and primary sclerosing cholangitis), and in rats subjected to bile-duct ligation. The regulation of EBP50 by estrogens and its impact on proliferation were assessed in both bile duct-ligated rats and Mz-Cha-1 human biliary epithelial cells. Analyses of cell isolates and immunohistochemical studies showed that in normal human liver, EBP50 is expressed in the canalicular membranes of hepatocytes and, together with ezrin and cystic fibrosis transmembrane conductance regulator, in the apical domains of cholangiocytes. In both human cholangiopathies and bile duct-ligated rats, EBP50 was redistributed to the cytoplasmic and nuclear compartments. EBP50 underwent a transient increase in rat cholangiocytes after bile-duct ligation, whereas such expression was down-regulated in ovariectomized rats. In addition, in Mz-Cha-1 cells, EBP50 underwent up-regulation and intracellular redistribution in response to 17beta-estradiol, whereas its proliferation was inhibited by siRNA-mediated EBP50 knockdown. These results indicate that both the expression and distribution of EBP50 are regulated by estrogens and contribute to the proliferative response in biliary epithelial cells.


Assuntos
Divisão Celular/fisiologia , Células Epiteliais/citologia , Vesícula Biliar/citologia , Fosfoproteínas/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Adolescente , Adulto , Idoso , Animais , Ductos Biliares/fisiologia , Criança , Pré-Escolar , Colangite Esclerosante/patologia , Fibrose Cística/patologia , Células Epiteliais/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/fisiologia , Feminino , Vesícula Biliar/efeitos dos fármacos , Humanos , Cirrose Hepática Biliar/patologia , Masculino , Pessoa de Meia-Idade , Ovariectomia , Fosfoproteínas/genética , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Trocadores de Sódio-Hidrogênio/genética , Adulto Jovem
10.
Pediatr Res ; 62(5): 528-32, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17805210

RESUMO

To elucidate the impact of nutrition in cystic fibrosis (CF), we compared the phenotypic traits of Cftr -/- mice fed either a lipid-enriched liquid diet (Peptamen) or a standard chow combined with polyethylenglycol osmotic laxative (PEG), two strategies commonly used to prevent intestinal obstruction in CF mice. Survival, growth, liver, and ventilatory status were determined in Cftr -/- and Cftr +/+ mice, followed-up until 120 d. Ventilation was recorded in conscious animals using whole-body plethysmography. We found that the survival rate was similar in Peptamen and PEG Cftr -/- mice. Cftr -/- mice had lower minute ventilation than Cftr +/+ mice, whatever the diet. Both Cftr -/- and Cftr +/+ mice fed Peptamen displayed preadult growth delay compared with PEG-treated animals. Despite subsequent growth catch-up, Cftr -/- mice remained smaller than Cftr +/+ mice, whatever the diet. All Peptamen fed Cftr -/- mice showed hepatomegaly and liver steatosis, which also occurred but to a lesser extent in Peptamen fed Cftr +/+ animals. Therefore, while both treatment strategies are similarly efficient to avoid high mortality at weaning, Peptamen induces preadult growth delay and liver steatosis. These effects of diet are important to consider in future animal studies and also prompt to evaluate high-energy diets in CF patients.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/efeitos dos fármacos , Fibrose Cística/tratamento farmacológico , Obstrução Intestinal/prevenção & controle , Laxantes/efeitos adversos , Oligopeptídeos/efeitos adversos , Polietilenoglicóis/efeitos adversos , Animais , Peso Corporal/efeitos dos fármacos , Fibrose Cística/complicações , Fibrose Cística/genética , Fibrose Cística/patologia , Fibrose Cística/fisiopatologia , Modelos Animais de Doenças , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/patologia , Transtornos do Crescimento/induzido quimicamente , Transtornos do Crescimento/fisiopatologia , Hepatomegalia/induzido quimicamente , Hepatomegalia/patologia , Obstrução Intestinal/etiologia , Obstrução Intestinal/fisiopatologia , Laxantes/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/patologia , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos CFTR , Oligopeptídeos/administração & dosagem , Fenótipo , Polietilenoglicóis/administração & dosagem , Ventilação Pulmonar/efeitos dos fármacos , Fatores de Tempo
11.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G25-35, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17615179

RESUMO

Cholestatic disorders may arise from liver ischemia (e.g., in liver transplantation) through various mechanisms. We have examined the potential of hypoxia to induce changes in the expression of hepatobiliary transporter genes. In a model of arterial liver ischemia subsequent to complete arterial deprivation of the rat liver, the mRNA levels of VEGF, a hypoxia-inducible gene, were increased fivefold after 24 h. The pattern of VEGF-induced expression and ultrastructural changes, including swelling of the endoplasmic reticulum, indicated that hypoxia affected primarily cholangiocytes, but also hepatocytes, predominantly in the periportal area. Serum and bile analyses demonstrated liver dysfunction of cholestatic type with reduced bile acid biliary excretion. Fluorescence-labeled ursodeoxycholic acid used as a tracer displayed no regurgitation, eliminating bile leakage as a significant mechanism of cholestasis in this model. In liver tissue, a marked reduction in the mRNA levels of Na(+)-taurocholate-cotransporting polypeptide (Ntcp), bile salt export protein (Bsep), and multidrug resistance-associated protein 2 (Mrp2) and an increase in those of Cftr were detected before bile duct proliferation occurred. In cultured hepatocytes, a nontoxic hypoxic treatment caused a decrease in the mRNA and protein expression of Ntcp, Bsep, and Mrp2 and in the mRNA levels of nuclear factors involved in the transactivation of these genes, i.e., HNF4alpha, RXRalpha, and FXR. In bile duct preparations, hypoxic treatment elicited an increase in Cftr transcripts, along with a rise in cAMP, a major regulator of Cftr expression and function. In conclusion, hypoxia triggers a downregulation of hepatocellular transporters, which may contribute to cholestasis, whereas Cftr, which drives secretion in cholangiocytes, is upregulated.


Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Regulação da Expressão Gênica/fisiologia , Hipóxia/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/biossíntese , Transportadores de Ânions Orgânicos/biossíntese , Simportadores/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Animais , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Células Cultivadas , Colestase/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/biossíntese , Isquemia/fisiopatologia , Fígado/irrigação sanguínea , Masculino , Ratos , Ratos Wistar , Ácido Ursodesoxicólico/metabolismo
12.
Lab Invest ; 87(3): 292-303, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17260005

RESUMO

Liver fibrosis is produced by myofibroblasts of different origins. In culture models, rat myofibroblasts derived from hepatic stellate cells (HSCs) and from periductal portal mesenchymal cells, show distinct proliferative and immunophenotypic evolutive profiles, in particular regarding desmin microfilament (overexpressed vs shut-down, respectively). Here, we examined the contributions of both cell types, in two rat models of cholestatic injury, arterial liver ischemia and bile duct ligation (BDL). Serum and (immuno)histochemical hepatic analyses were performed at different time points (2 days, 1, 2 and 6 weeks) after injury induction. Cholestatic liver injury, as attested by serum biochemical tests, was moderate/resolutive in ischemia vs severe and sustained in BDL. Spatio-temporal and morphometric analyses of cytokeratin-19 and Sirius red stainings showed that in both models, fibrosis accumulated around reactive bile ductules, with a significant correlation between the progression rates of fibrosis and of the ductular reaction (both higher in BDL). After 6 weeks, fibrosis was stabilized and did not exceed F2 (METAVIR) in arterial ischemia, whereas micronodular cirrhosis (F4) was established in BDL. Immuno-analyses of alpha-smooth muscle actin and desmin expression profiles showed that intralobular HSCs underwent early phenotypic changes marked by desmin overexpression in both models and that the accumulation of fibrosis coincided with that of alpha-SMA-labeled myofibroblasts around portal/septal ductular structures. With the exception of desmin-positive myofibroblasts located at the portal/septal-lobular interface at early stages, and of myofibroblastic HSCs detected together with fine lobular septa in BDL cirrhotic liver, the vast majority of myofibroblasts were desmin-negative. These findings suggest that both in resolutive and sustained cholestatic injury, fibrosis is produced by myofibroblasts that derive predominantly from portal/periportal mesenchymal cells. While HSCs massively undergo phenotypic changes marked by desmin overexpression, a minority fully converts into matrix-producing myofibroblasts, at sites, which however may be important in the healing process that circumscribes wounded hepatocytes.


Assuntos
Colestase/patologia , Isquemia/patologia , Cirrose Hepática/patologia , Mesoderma/citologia , Actinas/metabolismo , Animais , Desmina/metabolismo , Imuno-Histoquímica , Masculino , Ratos , Ratos Wistar
13.
FASEB J ; 20(6): 773-5, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16484330

RESUMO

Recent evidence indicates that growth hormone (GH) is involved in liver regeneration. To test whether insulin-like growth factor I (IGF-I) mediates this effect, we studied liver regeneration induced by partial hepatectomy in liver-specific IGF type 1 receptor knockout (LIGFREKO) mice. The absence of IGF-1R caused a significant decrease in hepatocyte proliferation in males (-52%), but not in females, as assessed by Ki67 immunohistochemistry. Cyclin D1 and cyclin A protein levels in the livers of LIGFREKO males were only half those in controls, indicating that cyclin induction during liver regeneration is dependent on IGF-1R signaling. Analyzing the signaling cascade initiated by IGF-1R, we observed a lack of IRS-1 induction in LIGFREKO livers. In contrast, the induction of IRS-2 synthesis was similar in LIGFREKO and control groups, suggesting the existence of differential regulation of IRS synthesis during liver regeneration. Regenerating livers from LIGFREKO animals also showed significantly less activated ERKs than controls. Our findings demonstrate that IGF-1R makes a significant contribution to liver regeneration. Using the LIGFREKO model, we provide new evidence that IGF-1R/IRS-1/ERK signaling may be the intracellular pathway controlling the cell cycle via cyclin D1 and cyclin A in the regenerating liver.


Assuntos
Hepatócitos/citologia , Hepatócitos/metabolismo , Regeneração Hepática , Fígado/metabolismo , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/metabolismo , Animais , Proliferação de Células , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica , Genótipo , Proteínas Substratos do Receptor de Insulina , Fígado/citologia , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptor IGF Tipo 1/genética , Caracteres Sexuais
14.
Am J Pathol ; 167(3): 869-77, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127164

RESUMO

De-regulations in insulin and insulin-like growth factor (IGF) pathways may contribute to hepatocellular carcinoma. Although intracellular insulin receptor substrate-2 (IRS-2) is the main effector of insulin signaling in the liver, its role in hepatocarcinogenesis is unknown. Here, we show that IRS-2 was overexpressed in two murine models of hepatocarcinogenesis: administration of diethylnitrosamine and hepatic overexpression of SV40 large T antigen. In both models, IRS-2 overexpression was detected in preneoplastic lesions and at higher levels in tumoral nodules. IRS-2 overexpression associated with IGF-2 and IRS-1 overexpression and with GSK-3beta inhibition. Increased expression of IRS-2 was also detected in human hepatocellular carcinoma specimens and hepatoma cell lines. In murine and human hepatoma cells, IRS-2 protein induction associated with increased IRS-2 mRNA levels. The functionality of IRS-2 was demonstrated in Hep 3 B cells, in which IRS-2 tyrosine phosphorylation and its association with phosphatidylinositol-3 kinase were induced by IGF-2. Moreover, down-regulation of IRS-2 expression increased apoptosis in these cells. In conclusion, we demonstrate that IRS-2 is overexpressed in human and murine hepatocellular carcinoma. The emergence of IRS-2 overexpression at preneoplastic stages during experimental hepatocarcinogenesis and its protective effect against apoptosis suggest that IRS-2 contributes to liver tumor progression.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Fosfoproteínas/metabolismo , Alquilantes , Animais , Antígenos Transformantes de Poliomavirus/imunologia , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Dietilnitrosamina , Humanos , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Fatores de Tempo
15.
Hepatology ; 41(2): 307-14, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15660382

RESUMO

Epidermal growth factor receptor (EGFR) binds transforming growth factor alpha (TGF-alpha) which is mitogenic for hepatocytes. Diverse lines of evidence suggest that activation of the TGF-alpha /EGFR pathway contributes to hepatocellular carcinoma (HCC) formation. Herein, we developed an experimental model of cirrhosis giving rise to HCC and tested the antitumoral effect of gefitinib, a selective EGFR tyrosine kinase inhibitor, in this model. Rats received weekly intraperitoneal injections of diethylnitrosamine (DEN) followed by a 2-week wash-out period that caused cirrhosis in 14 weeks and multifocal HCC in 18 weeks. Hepatocyte proliferation was increased in diseased tissue at 14 weeks compared with control liver and at even higher levels in HCC nodules compared with surrounding diseased tissues at 18 weeks. Increased proliferation was paralleled by upregulation of TGF-alpha messenger RNA expression. A group of DEN-treated rats received daily intraperitoneal injections of gefitinib between weeks 12 and 18. In rats treated with gefitinib, the number of HCC nodules was significantly lower than in untreated rats (18.1 +/- 2.4 vs. 3.7 +/- 0.45; P < .05), while EGFR was activated to a lesser extent in the diseased and tumoral tissues of these animals compared with untreated rats. HCC nodules from both untreated and gefitinib-treated animals displayed insulin-like growth factor 2 overexpression that contributed to tumor formation in treated animals. In conclusion, the blockade of EGFR activity by gefitinib has an antitumoral effect on the development of HCC in DEN-exposed rats, suggesting that it may provide benefit for the chemoprevention of HCC.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/prevenção & controle , Receptores ErbB/antagonistas & inibidores , Cirrose Hepática/complicações , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/prevenção & controle , Quinazolinas/farmacologia , Alquilantes , Animais , Dietilnitrosamina , Receptores ErbB/metabolismo , Gefitinibe , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Cirrose Hepática/fisiopatologia , Masculino , Ratos , Ratos Wistar , Transdução de Sinais , Fator de Crescimento Transformador alfa/metabolismo
16.
J Hepatol ; 42(2): 257-65, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15664253

RESUMO

BACKGROUND/AIMS: A rat model of bile duct ischemia was established and used to examine the potential of bile duct proliferation to provide an adaptative response in cholestatic disorders. METHODS: Rats underwent partial or complete arterial deprivation of the liver. Serum biochemical tests, histological analyses and bile secretion measurements were performed at different time points up to 6 weeks after surgery. RESULTS: Rats developed biochemical signs of cholestasis exclusively after complete arterial deprivation. Within 4h, cholangiocytes in these rats showed morphological signs of cell damage. After 48h, they displayed VEGF expression and became proliferative. The proportion of Ki67-labeled cholangiocytes ( approximately 30%) was similar in interlobular bile ducts and periportal ductules. A ductular reaction made of well-formed bile ducts confined to portal tracts developed within 1 week. Bile flow which was initially decreased, was restored at 3 weeks, while the biochemical signs of cholestasis completely resolved at 6 weeks. At this time, the number of bile duct sections was maximal. Fibrosis intensity was also maximal, although moderate (

Assuntos
Ductos Biliares/irrigação sanguínea , Ductos Biliares/patologia , Artéria Hepática/cirurgia , Isquemia/patologia , Animais , Divisão Celular , Colestase/patologia , Modelos Animais de Doenças , Hipóxia , Circulação Hepática , Testes de Função Hepática , Masculino , Sistema Porta , Ratos , Ratos Wistar , Fatores de Tempo
17.
Lab Invest ; 83(2): 163-73, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12594232

RESUMO

The origin of myofibroblasts and the factors promoting their differentiation during liver fibrogenesis remain uncertain. During biliary-type fibrogenesis, the proliferation and chemoattraction of hepatic stellate cells (HSC) toward bile ducts is mediated by platelet-derived growth factor (PDGF), while myofibroblastic conversion of peribiliary cells distinct from HSC also occurs. We herein examined the phenotype of these peribiliary myofibroblasts as compared with myofibroblastic HSC and tested whether their differentiation was affected by PDGF. Biliary-type liver fibrogenesis was induced by common bile duct ligation in rats. After 48 hours, periductular fibrosis in portal tracts colocalized with smooth muscle alpha-actin-immunoreactive myofibroblasts, the majority of which were desmin negative. Simultaneously, in sinusoids, desmin immunoreactivity was induced in a large number of HSC, which were smooth muscle alpha-actin negative. Cultures of peribiliary myofibroblasts were expanded from isolated bile duct segments and compared with myofibroblastic HSC. Peribiliary myofibroblasts outgrowing from bile duct segments expressed smooth muscle alpha-actin, alpha1 (I) collagen mRNA, and PDGF receptor-beta subunit. Desmin immunoreactivity gradually decreased in cultured peribiliary myofibroblasts, contrasting with constant labeling of all myofibroblastic HSC. In addition, IL-6 expression in peribiliary myofibroblasts was up to 100-fold lower than in myofibroblastic HSC, whereas the expression of the complement-activating protease P100 in both cell types showed little difference and that of the extracellular matrix component fibulin 2 was similar. The expression of smooth muscle alpha-actin protein in cultured peribiliary myofibroblasts was stimulated by PDGF-BB and inhibited by STI571, a PDGF receptor tyrosine kinase inhibitor, whereas in bile duct-ligated rats, the administration of STI571 caused a significant decrease in peribiliary smooth muscle alpha-actin immunoreactivity, and to a lesser extent, a decrease in peribiliary fibrosis. These results indicate that peribiliary cells distinct from HSC undergo a PDGF-mediated conversion into myofibroblasts expressing IL-6 at lower levels than myofibroblastic HSC and contribute to the initial formation of biliary-type liver fibrosis.


Assuntos
Ductos Biliares/patologia , Fibroblastos/patologia , Células de Kupffer/patologia , Cirrose Hepática Biliar/patologia , Cirrose Hepática Experimental/patologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Actinas/análise , Animais , Benzamidas , Ductos Biliares/efeitos dos fármacos , Diferenciação Celular , Linhagem da Célula , Colágeno/genética , Colágeno/metabolismo , Desmina/análise , Modelos Animais de Doenças , Fibroblastos/metabolismo , Mesilato de Imatinib , Células de Kupffer/efeitos dos fármacos , Cirrose Hepática Biliar/etiologia , Cirrose Hepática Biliar/metabolismo , Cirrose Hepática Experimental/etiologia , Cirrose Hepática Experimental/metabolismo , Masculino , Fenótipo , Piperazinas , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Hepatol ; 37(2): 184-91, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12127422

RESUMO

BACKGROUND/AIMS: Bosentan, a dual endothelin ET(A/B) receptor antagonist, may cause dose-dependent reversible cholestatic liver injury. We herein tested whether bosentan or metabolites, both eliminated in bile, induce alterations in bile secretion. METHODS: Bile flow and output of bile constituents were monitored in pentobarbital-anesthetized rats with biliary fistulas. Normal and TR(-) rats with a genetic defect in mrp2, received bosentan intravenous injections. RESULTS: Bosentan bolus intravenous injections of 0.1-10mg/kg triggered a dose-dependent increase in biliary bilirubin excretion. In addition, doses (> or =10mg/kg) caused a sustained increase in canalicular bile salt-independent bile flow, combined with significant increases in the concentration and output of glutathione and of bicarbonate in bile. In rats receiving bosentan (> or =10mg/kg), both under basal conditions and under intravenous taurocholate perfusion (2micromol/min/kg), phospholipid and cholesterol secretions were profoundly inhibited and uncoupled from bile salt secretion. In TR(-) rats, the choleretic effect of bosentan was reduced to non-significant levels. The stimulation of bilirubin secretion and the uncoupling of phospholipid from bile salt secretion were absent, whereas that of cholesterol was maintained. CONCLUSIONS: Bosentan alters canalicular bile formation in major part via mrp2-mediated mechanisms. Intermittent uncoupling of lipid from bile salt secretion may contribute to bosentan hepatic adverse reaction.


Assuntos
Anti-Hipertensivos/farmacologia , Canalículos Biliares/metabolismo , Proteínas de Membrana Transportadoras , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Sulfonamidas/farmacologia , Animais , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/efeitos dos fármacos , Bilirrubina/metabolismo , Bosentana , Colesterol/metabolismo , Antagonistas dos Receptores de Endotelina , Masculino , Proteína 2 Associada à Farmacorresistência Múltipla , Fosfolipídeos/metabolismo , Ratos , Ratos Mutantes , Ratos Sprague-Dawley , Ratos Wistar , Receptor de Endotelina A , Receptor de Endotelina B
19.
Hepatology ; 35(5): 1010-21, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11981751

RESUMO

Cirrhosis consists of hepatocyte nodules surrounded by a highly vascularized fibrous tissue. We previously showed that the development of biliary cirrhosis in the rat is associated with the occurrence of hepatocellular hypoxia and the induction of hepatic angiogenesis. We herein examined the occurrence of hypoxia in an experimental model of diethylnitrosamine (DEN)-induced cirrhosis. We also determined whether hypoxia directly affects the expression of vascular endothelial growth factor (VEGF), of VEGF receptors (Flt-1, Flk-1), and of type I and type IV collagens in activated hepatic stellate cells (HSCs) and the expression of VEGF in hepatocytes. Our results show that in DEN-treated rats, although the progression of liver fibrosis is associated with hepatocellular hypoxia and angiogenesis, VEGF and Flt-1 expressions in the liver are increased and correlated with the density of microvessels. In vitro, hypoxia induces the expression of VEGF, Flt-1, and type I collagen in activated HSCs and that of VEGF in hepatocytes. In addition, we show that hypoxia-induced type I collagen expression in HSCs may occur independently of transforming growth factor beta1 (TGF-beta1) overexpression. In conclusion, the present study provides further evidence that hepatocellular hypoxia and angiogenesis progress together with fibrogenesis after liver injury and that hypoxia directly contributes to the progression of liver fibrosis.


Assuntos
Colágeno Tipo I/genética , Fatores de Crescimento Endotelial/genética , Hipóxia/fisiopatologia , Cirrose Hepática Experimental/fisiopatologia , Linfocinas/genética , Neovascularização Patológica/fisiopatologia , Alquilantes , Animais , Células Cultivadas , Colágeno Tipo IV/genética , Dietilnitrosamina , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/genética , Fibrose , Expressão Gênica , Hepatócitos/citologia , Hepatócitos/fisiologia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/patologia , Masculino , Ratos , Ratos Wistar , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...