Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nutrients ; 15(23)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38068729

RESUMO

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is associated with visceral adiposity. We assessed the effectiveness of time-restricted fasting (TRF) for 16 h daily without calorie restrictions compared to standard care (SC; diet and lifestyle advice) in improving visceral adiposity and steatosis via controlled attenuation parameter (CAP). METHODS: In a prospective single-blind randomized controlled trial, 32 participants with NAFLD were randomly assigned to TRF or SC for 12 weeks. The secondary endpoints were changes in liver stiffness, anthropometry, blood pressure, and other metabolic factors. RESULTS: Twenty-eight participants completed the first arm of the study (TRF = 14, SC = 14), with 23 completing the crossover arm (TRF = 10, SC = 13). The baseline demographics were similar between the groups. Intermittent fasting caused a significant decrease in hepatic steatosis (p = 0.038), weight (p = 0.005), waist circumference (p = 0.001), and BMI (p = 0.005) compared to standard care. Intermittent fasting also resulted in additional within-group changes that were not seen in the standard care intervention. CONCLUSION: TRF offers superior improvements in patients with NAFLD, improving steatosis, weight, and waist circumference despite a lack of change in overall caloric intake. Time-restricted fasting should be considered as a primary weight loss intervention in the context of NAFLD. TRIAL REGISTRATION: ACTRN12613000935730.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Jejum Intermitente , Estudos Cross-Over , Estudos Prospectivos , Método Simples-Cego , Fígado/metabolismo
2.
World J Gastroenterol ; 23(31): 5692-5699, 2017 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-28883694

RESUMO

AIM: To evaluate the role of tissue factor (TF) and protease activated receptor (PAR)-2 in liver fibrosis. METHODS: Using CCl4 administration for eight weeks, we induced hepatic fibrosis in wild-type C57BL/6 mice and in mice with deletion of the cytoplasmic signalling domain of TF (TF§CT/§CT), deletion of PAR-2 (PAR-2-/-) and combined deletion of TF signalling domain and PAR-2 (TF§CT/§CT/PAR-2-/-). Hepatic fibrosis area was assessed by quantitative imaging of picrosirius red staining. Hepatic collagen content was assessed by hydroxyproline levels. Hepatic stellate cells (αSMA positive) and hepatic macrophages (CD68 positive) were identified by immunohistochemistry. Hepatic gene expression was determined by PCR and liver TGFß1 content by ELISA. RESULTS: CCl4 treated mice with deletion of the PAR-2 gene (PAR-2-/-) and the cytoplasmic domain of TF (TF§CT/§CT) developed significantly less hepatic fibrosis, characterised by reduced liver fibrosis area and hydroxyproline content, compared to control wildtype mice treated with CCl4. The observed reduction in histological fibrosis was accompanied by a significant decrease in the hepatic content of TGFß, the prototypic fibrogenic cytokine, as well as fewer activated hepatic stellate cells and hepatic macrophages. Deletion of the TF cytoplasmic signalling domain reduced hepatic fibrosis to levels similar to those observed in mice lacking PAR-2 signalling but combined deletion provided no added protection against fibrosis indicating a lack of mutual modulating effects that have been observed in other contexts such as angiogenic responses. CONCLUSION: Tissue factor cytoplasmic domain is involved in TF-PAR-2 signalling initiating hepatic fibrosis and is a potential therapeutic target, as its deletion would not impact coagulation.


Assuntos
Cirrose Hepática Experimental/patologia , Fígado/patologia , Domínios Proteicos/genética , Receptor PAR-2/metabolismo , Tromboplastina/metabolismo , Sequência de Aminoácidos/genética , Animais , Coagulação Sanguínea , Tetracloreto de Carbono/toxicidade , Colágeno/análise , Células Estreladas do Fígado/imunologia , Humanos , Hidroxiprolina/análise , Fígado/citologia , Fígado/imunologia , Cirrose Hepática Experimental/sangue , Cirrose Hepática Experimental/induzido quimicamente , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-2/genética , Deleção de Sequência , Transdução de Sinais , Tromboplastina/genética , Fator de Crescimento Transformador beta/análise
3.
Cytotherapy ; 16(8): 1132-44, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24642017

RESUMO

BACKGROUND: Intravenous infusion of human amniotic epithelial cells (hAECs) has been shown to ameliorate hepatic fibrosis in murine models. Hepatic stellate cells (HSCs) are the principal collagen-secreting cells in the liver. The aim of this study was to investigate whether factors secreted by hAECs and present in hAEC-conditioned medium (CM) have anti-fibrotic effects on activated human HSCs. METHODS: Human AECs were isolated from the placenta and cultured. Human hepatic stellate cells were exposed to hAEC CM to determine potential anti-fibrotic effects. RESULTS: HSCs treated for 48 h with hAEC CM displayed a significant reduction in the expression of the myofibroblast markers α-smooth muscle actin and platelet-derived growth factor. Expression of the pro-fibrotic cytokine transforming growth factor-ß1 (TGF-ß1) and intracellular collagen were reduced by 45% and 46%, respectively. Human AEC CM induced HSC apoptosis in 11.8% of treated cells and reduced HSC proliferation. Soluble human leukocyte antigen-G1, a hAEC-derived factor, significantly decreased TGF-ß1 and collagen production in activated HSCs, although the effect on collagen production was less than that of hAEC CM. The reduction in collagen and TGF-B1 could not be attributed to PGE2, relaxin, IL-10, TGF-B3, FasL or TRAIL. CONCLUSIONS: Human AEC CM treatment suppresses markers of activation, proliferation and fibrosis in human HSCs as well as inducing apoptosis and reducing proliferation. Human AEC CM treatment may be effective in ameliorating liver fibrosis and warrants further study.


Assuntos
Líquido Amniótico/citologia , Colágeno/biossíntese , Meios de Cultivo Condicionados/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Líquido Amniótico/metabolismo , Animais , Apoptose/efeitos dos fármacos , Colágeno/metabolismo , Modelos Animais de Doenças , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Humanos , Fator de Crescimento Transformador beta/biossíntese
4.
PLoS One ; 8(8): e69299, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936322

RESUMO

Lung diseases are a major cause of global morbidity and mortality that are treated with limited efficacy. Recently stem cell therapies have been shown to effectively treat animal models of lung disease. However, there are limitations to the translation of these cell therapies to clinical disease. Studies have shown that delayed treatment of animal models does not improve outcomes and that the models do not reflect the repeated injury that is present in most lung diseases. We tested the efficacy of amnion mesenchymal stem cells (AM-MSC), bone marrow MSC (BM-MSC) and human amniotic epithelial cells (hAEC) in C57BL/6 mice using a repeat dose bleomycin-induced model of lung injury that better reflects the repeat injury seen in lung diseases. The dual bleomycin dose led to significantly higher levels of inflammation and fibrosis in the mouse lung compared to a single bleomycin dose. Intravenously infused stem cells were present in the lung in similar numbers at days 7 and 21 post cell injection. In addition, stem cell injection resulted in a significant decrease in inflammatory cell infiltrate and a reduction in IL-1 (AM-MSC), IL-6 (AM-MSC, BM-MSC, hAEC) and TNF-α (AM-MSC). The only trophic factor tested that increased following stem cell injection was IL-1RA (AM-MSC). IL-1RA levels may be modulated by GM-CSF produced by AM-MSC. Furthermore, only AM-MSC reduced collagen deposition and increased MMP-9 activity in the lung although there was a reduction of the pro-fibrogenic cytokine TGF-ß following BM-MSC, AM-MSC and hAEC treatment. Therefore, AM-MSC may be more effective in reducing injury following delayed injection in the setting of repeated lung injury.


Assuntos
Células-Tronco Adultas/citologia , Lesão Pulmonar/terapia , Células-Tronco Adultas/transplante , Âmnio/citologia , Animais , Bleomicina/farmacologia , Células da Medula Óssea/citologia , Colágeno/metabolismo , Células Epiteliais/citologia , Células Epiteliais/transplante , Feminino , Fibrose , Humanos , Inflamação/metabolismo , Inflamação/terapia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Receptores de Interleucina-1/antagonistas & inibidores
5.
PLoS One ; 7(6): e38631, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22719909

RESUMO

Chronic hepatic inflammation from multiple etiologies leads to a fibrogenic response that can progress to cirrhosis and liver failure. Transplantation of human amniotic epithelial cells (hAEC) from term delivered placenta has been shown to decrease mild to moderate hepatic fibrosis in a murine model. To model advanced human liver disease and assess the efficacy of hAEC therapy, we transplanted hAEC in mice with advanced hepatic fibrosis. Immunocompetent C57BL/6 mice were administered carbon tetrachloride (CCl(4)) twice weekly resulting in bridging fibrosis by 12 weeks. hAEC (2 × 10(6)) were infused via the tail vein at week 8 or weeks 8 and 10 (single and double dose, respectively). Human cells were detected in mouse liver four weeks after transplantation showing hAEC engraftment. CCl(4) treated mice receiving single or double hAEC doses showed a significant but similar decrease in liver fibrosis area associated with decreased activation of collagen-producing hepatic stellate cells and decreased hepatic protein levels of the pro-fibrogenic cytokine, transforming growth factor-beta1. CCl(4) administration caused hepatic T cell infiltration that decreased significantly following hAEC transplantation. Hepatic macrophages play a crucial role in both fibrogenesis and fibrosis resolution. Mice exposed to CCl(4) demonstrated increased numbers of hepatic macrophages compared to normal mice; the number of macrophages decreased significantly in CCl(4) treated mice given hAEC. These mice had significantly lower hepatic protein levels of the chemokine monocyte chemoattractant protein-1 than mice given CCl(4) alone. Alternatively activated M2 macrophages are associated with fibrosis resolution. CCl(4) treated mice given hAEC showed increased expression of genes associated with M2 macrophages including YM-1, IL-10 and CD206. We provide novel data showing that hAEC transplantation induces a wound healing M2 macrophage phenotype associated with reduction of established hepatic fibrosis that justifies further investigation of this potential cell-based therapy for advanced hepatic fibrosis.


Assuntos
Âmnio/transplante , Transplante de Células , Cirrose Hepática/prevenção & controle , Ativação de Macrófagos , Âmnio/citologia , Animais , Formação de Anticorpos , Tetracloreto de Carbono/administração & dosagem , Tetracloreto de Carbono/toxicidade , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/transplante , Humanos , Imuno-Histoquímica , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
6.
Hepatology ; 55(3): 879-87, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22095855

RESUMO

UNLABELLED: Protease-activated receptor (PAR) 2 is a G-protein-coupled receptor that is activated after proteolytic cleavage by serine proteases, including mast cell tryptase and activated coagulation factors. PAR-2 activation augments inflammatory and profibrotic pathways through the induction of genes encoding proinflammatory cytokines and extracellular matrix proteins. Thus, PAR-2 represents an important interface linking coagulation and inflammation. PAR-2 is widely expressed in cells of the gastrointestinal tract, including hepatic stellate cells (HSCs), endothelial cells, and hepatic macrophages; however, its role in liver fibrosis has not been previously examined. We studied the development of CCl(4) -induced liver fibrosis in PAR-2 knockout mice, and showed that PAR-2 deficiency reduced the progression of liver fibrosis, hepatic collagen gene expression, and hydroxyproline content. Reduced fibrosis was associated with decreased transforming growth factor beta (TGFß) gene and protein expression and decreased matrix metalloproteinase 2 and tissue inhibitor of matrix metalloproteinase 1 gene expression. In addition, PAR-2 stimulated activation, proliferation, collagen production, and TGFß protein production by human stellate cells, indicating that hepatic PAR-2 activation increases profibrogenic cytokines and collagen production both in vivo and in vitro. CONCLUSION: Our findings demonstrate the capacity of PAR-2 activation to augment TGFß production and promote hepatic fibrosis in mice and to induce a profibrogenic phenotype in human HSCs. PAR-2 antagonists have recently been developed and may represent a novel therapeutic approach in preventing fibrosis in patients with chronic liver disease.


Assuntos
Tetracloreto de Carbono/efeitos adversos , Progressão da Doença , Células Estreladas do Fígado/fisiologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/fisiopatologia , Receptor PAR-2/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Células Estreladas do Fígado/citologia , Humanos , Hidroxiprolina/metabolismo , Técnicas In Vitro , Fígado/metabolismo , Fígado/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligopeptídeos/farmacologia , Receptor PAR-2/agonistas , Receptor PAR-2/deficiência , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo
7.
Cell Transplant ; 19(9): 1157-68, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20447339

RESUMO

Chronic liver injury and inflammation lead to hepatic fibrosis, cirrhosis, and liver failure. Embryonic and mesenchymal stem cells have been shown to reduce experimental liver fibrosis but have potential limitations, including the formation of dysplastic precursors, tumors, and profibrogenic cells. Other stem-like cells may reduce hepatic inflammation and fibrosis without tumor and profibrogenic cell formation. To test this hypothesis we transplanted human amnion epithelial cells (hAEC), isolated from term delivered placenta, into immunocompetent C57/BL6 mice at week 2 of a 4-week regimen of carbon tetrachloride (CCl4) exposure to induce liver fibrosis. Two weeks following hAEC infusion, intact cells expressing the human-specific markers inner mitochondrial membrane protein and human leukocyte antigen-G were found in mouse liver without evidence of host rejection of the transplanted cells. Human albumin, known to be produced by hAEC, was detected in sera of hAEC-treated mice. Human DNA was detected in mouse liver and also spleen, lungs, and heart of some animals. Following hAEC transplantation, CCl4-treated animals showed decreased serum ALT levels and reduced hepatocyte apoptosis, compared to controls. hAEC-treated mouse liver had lower TNF-α and IL-6 protein levels and higher IL-10 compared to animals given CCl4 alone. Compared to CCl4 controls, hAEC-treated mice showed fewer activated collagen-producing hepatic stellate cells and less fibrosis area and collagen content. Reduced hepatic TGF-ß levels in conjunction with a twofold increase in the active form of the collagen-degrading enzyme matrix metalloproteinase-2 in hAEC-treated mice compared to CCl4 controls may account for the reduction in fibrosis. hAEC transplantation into immunocompetent mice leads to cell engraftment, reduced hepatocyte apoptosis, and decreased hepatic inflammation and fibrosis.


Assuntos
Âmnio/citologia , Transplante de Células/métodos , Cirrose Hepática/cirurgia , Animais , Apoptose/fisiologia , Tetracloreto de Carbono , Intoxicação por Tetracloreto de Carbono/imunologia , Intoxicação por Tetracloreto de Carbono/patologia , Intoxicação por Tetracloreto de Carbono/cirurgia , Separação Celular/métodos , Células Epiteliais/citologia , Feminino , Humanos , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Fator de Crescimento Transformador beta/metabolismo , Transplante Heterólogo
8.
Am J Pathol ; 175(1): 303-13, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19497992

RESUMO

Acute respiratory distress syndrome is characterized by loss of lung tissue as a result of inflammation and fibrosis. Augmenting tissue repair by the use of mesenchymal stem cells may be an important advance in treating this condition. We evaluated the role of term human umbilical cord cells derived from Wharton's jelly with a phenotype consistent with mesenchymal stem cells (uMSCs) in the treatment of a bleomycin-induced mouse model of lung injury. uMSCs were administered systemically, and lungs were harvested at 7, 14, and 28 days post-bleomycin. Injected uMSCs were located in the lung 2 weeks later only in areas of inflammation and fibrosis but not in healthy lung tissue. The administration of uMSCs reduced inflammation and inhibited the expression of transforming growth factor-beta, interferon-gamma, and the proinflammatory cytokines macrophage migratory inhibitory factor and tumor necrosis factor-alpha. Collagen concentration in the lung was significantly reduced by uMSC treatment, which may have been a consequence of the simultaneous reduction in Smad2 phosphorylation (transforming growth factor-beta activity). uMSCs also increased matrix metalloproteinase-2 levels and reduced their endogenous inhibitors, tissue inhibitors of matrix metalloproteinases, favoring a pro-degradative milieu following collagen deposition. Notably, injected human lung fibroblasts did not influence either collagen or matrix metalloproteinase levels in the lung. The results of this study suggest that uMSCs have antifibrotic properties and may augment lung repair if used to treat acute respiratory distress syndrome.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Lesão Pulmonar/terapia , Transplante de Células-Tronco Mesenquimais , Síndrome do Desconforto Respiratório/terapia , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Western Blotting , Colágeno/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Sangue Fetal , Humanos , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/patologia , Inflamação/terapia , Lesão Pulmonar/induzido quimicamente , Fatores Inibidores da Migração de Macrófagos/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Células-Tronco Mesenquimais , Camundongos , Camundongos SCID , Reação em Cadeia da Polimerase , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Fibrose Pulmonar/terapia , RNA Mensageiro/análise , Síndrome do Desconforto Respiratório/patologia , Inibidores Teciduais de Metaloproteinases/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Clin Sci (Lond) ; 117(11): 375-86, 2009 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-19371232

RESUMO

Ang-(1-7) (angiotensin-1-7), a peptide product of the recently described ACE (angiotensin-converting enzyme) homologue ACE2, opposes the harmful actions of AngII (angiotensin II) in cardiovascular tissues, but its role in liver disease is unknown. The aim of the present study was to assess plasma levels of Ang-(1-7) in human liver disease and determine its effects in experimental liver fibrosis. Angiotensin peptide levels were measured in cirrhotic and non-cirrhotic patients with hepatitis C. The effects of Ang-(1-7) on experimental fibrosis were determined using the rat BDL (bile-duct ligation) model. Liver histology, hydroxyproline quantification and expression of fibrosis-related genes were assessed. Expression of RAS (renin-angiotensin system) components and the effects of Ang-(1-7) were examined in rat HSCs (hepatic stellate cells). In human patients with cirrhosis, both plasma Ang-(1-7) and AngII concentrations were markedly elevated (P<0.001). Non-cirrhotic patients with hepatitis C had elevated Ang-(1-7) levels compared with controls (P<0.05), but AngII concentrations were not increased. In BDL rats, Ang-(1-7) improved fibrosis stage and collagen Picrosirius Red staining, and reduced hydroxyproline content, together with decreased gene expression of collagen 1A1, alpha-SMA (smooth muscle actin), VEGF (vascular endothelial growth factor), CTGF (connective tissue growth factor), ACE and mas [the Ang-(1-7) receptor]. Cultured HSCs expressed AT1Rs (AngII type 1 receptors) and mas receptors and, when treated with Ang-(1-7) or the mas receptor agonist AVE 0991, produced less alpha-SMA and hydroxyproline, an effect reversed by the mas receptor antagonist A779. In conclusion, Ang-(1-7) is up-regulated in human liver disease and has antifibrotic actions in a rat model of cirrhosis. The ACE2/Ang-(1-7)/mas receptor axis represents a potential target for antifibrotic therapy in humans.


Assuntos
Angiotensina I/sangue , Hepatite C Crônica/sangue , Cirrose Hepática/sangue , Fragmentos de Peptídeos/sangue , Sistema Renina-Angiotensina/fisiologia , Regulação para Cima , Actinas/metabolismo , Adulto , Angiotensina I/genética , Angiotensina I/uso terapêutico , Angiotensina II/sangue , Animais , Ductos Biliares/patologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Feminino , Hepatite C Crônica/complicações , Humanos , Hidroxiprolina/metabolismo , Fígado/metabolismo , Cirrose Hepática/virologia , Cirrose Hepática Experimental/tratamento farmacológico , Cirrose Hepática Experimental/etiologia , Cirrose Hepática Experimental/patologia , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/uso terapêutico , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Renina/sangue
10.
Am J Pathol ; 171(3): 800-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17640968

RESUMO

Protease-activated receptor-2 (PAR-2) is a cellular receptor expressed prominently on epithelial, mesangial, and endothelial cells in the kidney and on macrophages. PAR-2 is activated by serine proteases such as trypsin, tryptase, and coagulation factors VIIa and Xa. It induces pleiotropic effects including vasodilatation, increasing plasminogen activator inhibitor (PAI-1) expression, mesangial cell proliferation, and cytokine production by macrophages. The role of PAR-2 in renal inflammation was studied in antiglomerular basement membrane antibody-induced crescentic glomerulonephritis (CGN) using PAR-2-deficient (PAR-2(-/-)) mice and wild-type littermate controls. PAR-2(-/-) mice had reduced crescent formation, proteinuria, and serum creatinine compared with wild-type mice 21 days after initiation of CGN. Glomerular accumulation of CD4(+) T cells and macrophages and the number of proliferating cells in glomeruli were similar in both groups. Glomerular fibrin deposition was significantly reduced in PAR-2(-/-) mice, and this was associated with reduced renal plasminogen activator inhibitor expression and increased renal matrix-metalloprotinase-9 activity. These results demonstrate a proinflammatory role for PAR-2 in CGN that is independent of effects on glomerular leukocyte recruitment and mesangial cell proliferation. PAR-2-mediated augmentation of renal plasminogen activator inhibitor expression and inhibition of matrix-metalloprotinase-9 activity may contribute to increased glomerular fibrin accumulation and glomerular injury in CGN.


Assuntos
Glomerulonefrite , Glomérulos Renais/patologia , Receptor PAR-2/metabolismo , Animais , Fibrina/metabolismo , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Receptor PAR-2/genética , Serpina E2 , Serpinas/genética , Serpinas/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
11.
Am J Physiol Gastrointest Liver Physiol ; 290(1): G137-44, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16123203

RESUMO

Activin A, a member of the transforming growth factor-beta superfamily, is constitutively expressed in hepatocytes and regulates liver mass through tonic inhibition of hepatocyte DNA synthesis. Follistatin is the main biological inhibitor of activin bioactivity. These molecules may be involved in hepatic fibrogenesis, although defined roles remain unclear. We studied activin and follistatin gene and protein expression in cultured rat hepatic stellate cells (HSCs) and in rats given CCl4 for 8 wk and examined the effect of follistatin administration on the development of hepatic fibrosis. In activated HSCs, activin mRNA was upregulated with high expression levels, whereas follistatin mRNA expression was unchanged from baseline. Activin A expression in normal lobular hepatocytes redistributed to periseptal hepatocytes and smooth muscle actin-positive HSCs in the fibrotic liver. A 32% reduction in fibrosis, maximal at week 4, occurred in CCl4-exposed rats treated with follistatin. Hepatocyte apoptosis decreased by 87% and was maximal at week 4 during follistatin treatment. In conclusion, activin is produced by activated HSCs in vitro and in vivo. Absence of simultaneous upregulation of follistatin gene expression in HSCs suggests that HSC-derived activin is biologically active and unopposed by follistatin. Our in vivo and in vitro results demonstrate that activin-mediated events contribute to hepatic fibrogenesis and that follistatin attenuates early events in fibrogenesis by constraining HSC proliferation and inhibiting hepatocyte apoptosis.


Assuntos
Apoptose , Folistatina/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Ativinas/genética , Ativinas/metabolismo , Animais , Folistatina/genética , Regulação da Expressão Gênica , Hepatócitos/patologia , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Cirrose Hepática/genética , Masculino , Subunidades Proteicas/metabolismo , Transporte Proteico , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...