Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Exp Pathol ; 94(1): 1-16, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23110747

RESUMO

Connective tissue growth factor (CTGF, CCN2) is a member of the CCN family of matricellular proteins. It interacts with many other proteins, including plasma membrane proteins, modulating cell function. It is expressed at low levels in normal adult kidney cells but is increased in kidney diseases, playing important roles in inflammation and in the development of glomerular and interstitial fibrosis in chronic disease. This review reports the evidence for its expression in human and animal models of chronic kidney disease and summarizes data showing that anti-CTGF therapy can successfully attenuate fibrotic changes in several such models, suggesting that therapies targeting CTGF and events downstream of it in renal cells may be useful for the treatment of human kidney fibrosis. Connective tissue growth factor stimulates the development of fibrosis in the kidney in many ways including activating cells to increase extracellular matrix synthesis, inducing cell cycle arrest and hypertrophy, and prolonging survival of activated cells. The relationship between CTGF and the pro-fibrotic factor TGFß is examined and mechanisms by which CTGF promotes signalling by the latter are discussed. No specific cellular receptors for CTGF have been discovered but it interacts with and activates several plasma membrane proteins including low-density lipoprotein receptor-related protein (LRP)-1, LRP-6, tropomyosin-related kinase A, integrins and heparan sulphate proteoglycans. Intracellular signalling and downstream events triggered by such interactions are reviewed. Finally, the relationships between CTGF and several anti-fibrotic factors, such as bone morphogenetic factor-4 (BMP4), BMP7, hepatocyte growth factor, CCN3 and Oncostatin M, are discussed. These may determine whether injured tissue heals or progresses to fibrosis.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Animais , Fator de Crescimento do Tecido Conjuntivo/genética , Citocinas/metabolismo , Progressão da Doença , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Nefropatias/terapia , Prognóstico , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
2.
Diabetes ; 61(9): 2280-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22586581

RESUMO

CCN2, a secreted profibrotic protein, is highly expressed in diabetic nephropathy (DN) and implicated in its pathogenesis; however, the actions of CCN2 in DN remain elusive. We previously demonstrated that CCN2 triggers signaling via tropomyosin receptor kinase A (TrkA). Trace expression of TrkA is found in normal kidneys, but its expression is elevated in several nephropathies; yet its role in DN is unexplored. In this study we show de novo expression of TrkA in human and murine DN. We go on to study the molecular mechanisms leading to TrkA activation and show that it involves hypoxia, as demonstrated by ischemia-reperfusion injury and in vitro experiments mimicking hypoxia, implicating hypoxia as a common pathway leading to disease. We also expose renal cells to hyperglycemia, which led to TrkA phosphorylation in mesangial cells, tubular epithelial cells, and podocytes but not in glomerular endothelial cells and renal fibroblasts. In addition, we report that hyperglycemia caused an induction of phosphorylated extracellular signal-related kinase 1/2 and Snail1 that was abrogated by silencing of TrkA or CCN2 using small interfering RNA. In conclusion, we provide novel evidence that TrkA is activated in diabetic kidneys and suggest that anti-TrkA therapy may prove beneficial in DN.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/fisiologia , Nefropatias Diabéticas/etiologia , Hiperglicemia/complicações , Animais , Fator de Crescimento do Tecido Conjuntivo/genética , Nefropatias Diabéticas/fisiopatologia , Humanos , Hiperglicemia/fisiopatologia , Hipóxia/complicações , Hipóxia/fisiopatologia , Rim/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , Células Mesangiais/metabolismo , Camundongos , Fosforilação , RNA Interferente Pequeno/farmacologia , Receptor trkA/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese
3.
Matrix Biol ; 30(7-8): 396-403, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21864682

RESUMO

Accumulation of type I collagen is a key event in renal interstitial fibrosis. As there is no effective treatment, understanding the site where collagen is transcribed and the factors driving it in response to disease in vivo is critical for designing future therapies. The present research investigated the transcriptional activity of the COL1A2 gene in a mouse model of progressive fibrosis induced by aristolochic acid (aristolochic acid nephropathy, AAN). To achieve this we genetically modified mice to express a reporter gene (LacZ) and CCN2 (connective tissue growth factor) under the transcriptional control of the COL1A2 promoter /enhancer sequences. Using these mice we asked where is collagen actively transcribed and secondly, what is the role of CCN2 in AAN. Here, we report that de-novo transcription of the COL1A2 gene occurred predominantly in damaged tubular epithelial cells during progressive interstitial fibrosis in vivo. The activation of COL1A2 was studied by detection of the reporter gene LacZ and COL1A2 mRNA in interstitial, glomerular, vascular, and tubular epithelial tissue from laser capture microscopy. We also demonstrated that LacZ-positive cells co-express E-Cadherin a marker of epithelial origin which is consistent with an epithelial phenotype which is capable of collagen expression during injury. There was no evidence of detachment of these cells from tubules to become myofibroblasts. Moreover, we showed that the transgenic mice show a modest enhancement of CCN2 expression; however fibrosis induced by AA is the same in transgenics and controls suggesting that CCN2, at this level of expression, is not sufficient to enhance fibrogenesis. Overall our study provides a better understanding into the expression patterns and roles of two major extracellular matrix proteins: type I collagen and CCN2.


Assuntos
Ácidos Aristolóquicos/efeitos adversos , Colágeno Tipo I/metabolismo , Células Epiteliais/metabolismo , Fibrose/patologia , Rim/patologia , Animais , Ácidos Aristolóquicos/administração & dosagem , Ácidos Aristolóquicos/farmacologia , Biomarcadores/análise , Nitrogênio da Ureia Sanguínea , Caderinas/metabolismo , Colágeno Tipo I/genética , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Fibrose/etiologia , Fibrose/metabolismo , Túbulos Renais/citologia , Microdissecção e Captura a Laser , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ativação Transcricional , Transgenes
4.
Int J Exp Pathol ; 92(3): 143-50, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21554437

RESUMO

Epithelial to mesenchymal transition (EMT) is a well established biological process in metazoan embryological development. Over the past 15 years, investigators have sought to establish whether EMT also occurs in renal epithelial cells, following kidney injury, and to show that the mesenchymal cells formed could give rise to myofibroblasts which populate the renal interstitium, causing fibrosis within it. There is no doubt that proximal tubular epithelial cells (PTECs) can undergo EMT in vitro in response to TGFß-1 and other inflammatory stimuli. Moreover, the results of experiments with animal models of renal fibrosis and examination of biopsies from patients with chronic kidney disease have lent support to the hypothesis that EMT occurs in vivo. This review discusses some of the key evidence underlying that idea and summarises recent advances in understanding the molecular mechanism underlying the process. Early experiments using mice which were genetically engineered to mark PTECs with the LacZ gene to trace their fate following kidney injury provided evidence supporting the occurrence of EMT. Recently, however, cell lineage tracking experiments using the red fluorescent protein (RFP) as a high-resolution marker for cells of renal epithelial origin did not replicate this result; the interstitial space following kidney injury was devoid of RFP expressing cells, leading the investigators to reject the renal EMT hypothesis.


Assuntos
Diferenciação Celular , Células Epiteliais/patologia , Rim/patologia , Mesoderma/patologia , Animais , Adesão Celular , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Humanos , Camundongos
5.
Mol Biol Cell ; 22(11): 1836-44, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21471005

RESUMO

Transforming growth factor-ß (TGF-ß) is an inducer of type I collagen, and uncontrolled collagen production leads to tissue scarring and organ failure. Here we hypothesize that uncovering a molecular mechanism that enables us to switch off type I collagen may prove beneficial in treating fibrosis. For the first time, to our knowledge, we provide evidence that CUX1 acts as a negative regulator of TGF-ß and potent inhibitor of type I collagen transcription. We show that CUX1, a CCAAT displacement protein, is associated with reduced expression of type I collagen both in vivo and in vitro. We show that enhancing the expression of CUX1 results in effective suppression of type I collagen. We demonstrate that the mechanism by which CUX1 suppresses type I collagen is through interfering with gene transcription. In addition, using an in vivo murine model of aristolochic acid (AA)-induced interstitial fibrosis and human AA nephropathy, we observe that CUX1 expression was significantly reduced in fibrotic tissue when compared to control samples. Moreover, silencing of CUX1 in fibroblasts from kidneys of patients with renal fibrosis resulted in increased type I collagen expression. Furthermore, the abnormal CUX1 expression was restored by addition of TGF-ß via the p38 mitogen-activated protein kinase pathway. Collectively, our study demonstrates that modifications of CUX1 expression lead to aberrant expression of type I collagen, which may provide a molecular basis for fibrogenesis.


Assuntos
Colágeno Tipo I/antagonistas & inibidores , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Ácidos Aristolóquicos , Células Cultivadas , Colágeno Tipo I/genética , Relação Dose-Resposta a Droga , Retroalimentação Fisiológica , Fibrose/tratamento farmacológico , Fibrose/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Rim/patologia , Nefropatias/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Proteína Smad3/metabolismo , Proteína Smad7 , Fatores de Transcrição , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
J Cell Commun Signal ; 3(2): 95-104, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19214781

RESUMO

Connective tissue growth factor (CTGF/CCN2) is a member of the CCN family of matricellular proteins. Its expression is induced by a number of factors including TGF-beta. It has been associated with fibrosis in various tissues including the kidney. Diabetic nephropathy (DN) develops in about 30% of patients with diabetes and is characterized by thickening of renal basement membranes, fibrosis in the glomerulus (glomerulosclerosis), tubular atrophy and interstitial fibrosis, all of which compromise kidney function. This review examines changes in CTGF expression in the kidney in DN, the effects they have on glomerular mesangial and podocyte cells and the tubulointerstitium, and how these contribute to driving fibrotic changes in the disease. CTGF can bind to several other growth factors modifying their function. CTGF is also able to interact with receptors on cells, including integrins, tyrosine receptor kinase A (TrkA), low density lipoprotein receptor-related protein (LRP) and heparan sulphate proteoglycans. These interactions, the intracellular signalling pathways they activate, and the cellular responses evoked are reviewed. CTGF also induces the expression of chemokines which themselves have pharmacological actions on cells. CTGF may prompt some responses by acting through several different mechanisms, possibly simultaneously. For example, CTGF is often described as an effector of TGF-beta. It can promote TGF-beta signalling by binding directly to the growth factor, promoting its interaction with the TGF-beta receptor; by triggering intracellular signalling on binding the TrkA receptor, which leads to the transcriptional repression of Smad7, an inhibitor of the TGF-beta signalling pathway; and by binding to BMP-7 whose own signalling pathway opposing TGF-beta is inhibited, leading to enhanced TGF-beta signalling.

7.
Biochem J ; 406(1): 131-8, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17489738

RESUMO

Activated mesangial cells are thought to play a pivotal role in the development of kidney fibrosis under chronic pathological conditions, including DN (diabetic nephropathy). Their prolonged survival may enhance the development of the disease since they express increased amounts of growth factors and extracellular matrix proteins. CTGF (connective tissue growth factor) is one of the growth factors produced by activated mesangial cells and is reported to play a key role in the pathogenesis of DN. Previous studies have shown that addition of exogenous CTGF to HMCs (human mesangial cells) rapidly activates ERK1/2 (extracellular-signal-regulated kinase 1/2) MAPK (mitogen-activated protein kinase) and JNK (c-Jun N-terminal kinase) MAPK, but not the p38 MAPK, despite the activation of the upstream kinases, MKK3/6 (MAPK kinase 3/6). The aim of the present study was to investigate whether the lack of phosphorylated p38 MAPK by CTGF has an anti-apoptotic effect on activated HMCs. We show that in HMC CTGF induces the rapid transcriptional activation and synthesis of MKP-1 (MAPK phosphatase-1), a dual specificity phosphatase that dephosphorylates p38 MAPK. This in turn prevents the anti-apoptotic protein, Bcl-2, from being phosphorylated and losing its function, leading to the survival of the cells. Knockout of MKP-1 protein in mesangial cells treated with CTGF, using siRNA (small interfering RNA) or antisense oligonucleotides, allows p38 MAPK activation and induces mesangial cell death.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Células Mesangiais/citologia , Células Mesangiais/enzimologia , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Adulto , Proteínas de Ciclo Celular/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo , Fosfatase 1 de Especificidade Dupla , Ativação Enzimática/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células Mesangiais/efeitos dos fármacos , Modelos Biológicos , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Nephron Exp Nephrol ; 104(4): e129-34, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16902316

RESUMO

In the adult kidney, the cellular phenotypes are maintained by a strict balance of growth factors. Epithelial-to-mesenchymal transition (EMT) is a program whereby injured epithelial cells that function as ion and fluid transporters become matrix remodelling mesenchymal cells. This process requires either transcriptional repression of genes that maintain the epithelial phenotype and transcriptional activation, or relieved repression of genes needed for functional myofibroblasts. The transcriptional regulators are controlled by several integrated signalling pathways which are triggered by growth factors. Emerging evidence indicates that the growth factors TGFbeta/CTGF and BMP-7/HGF are the main determinants that maintain the two cellular phenotypes. Both TGFbeta and BMP-7 counteract the activity of each other by cross-inducing their respective inhibitory Smads. Both growth factors may also induce the expression of other factors that can change the cellular environment and enhance their function. Chronic kidney diseases (regardless of the aetiology of the disease) are associated with increased TGFbeta and CTGF expression levels which, in turn, have an inverse effect on the activity level of BMP-7 and HGF, leading to an EMT of injured tubular epithelial cells and a progression of the disease. A detailed understanding of the complex interrelationship between these growth factors may lead to the development of novel drugs.


Assuntos
Células Epiteliais/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Mesoderma/fisiologia , Animais , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo , Fator de Crescimento de Hepatócito/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Proteína Smad6/fisiologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/fisiologia
9.
Am J Nephrol ; 26(4): 381-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16873994

RESUMO

BACKGROUND/AIM: Recent studies have suggested that statins may play a role in the protection against renal failure which is independent of cholesterol reduction. Activation of RhoGTPases is a key step in renal tubular cells' epithelial-to-mesenchymal transition (EMT) which contributes to renal interstitial fibrosis. We hypothesized that statins could act by inhibiting the synthesis of the isoprenoids, such as geranylgeranyl pyrophosphate, which is essential for membrane attachment and biological activity of RhoGTPases, RhoA and Rac1. METHODS: Human proximal tubular epithelial cells (HK2) were used to examine the inhibitory effect of statins on EMT induced with medium conditioned by activated peripheral blood mononuclear cells. RESULTS: Our study demonstrates that the statins lovastatin, simvastatin, and pravastatin inhibit HK2 cells to undergo EMT. Inhibition of EMT in HK2 cells with these statins resulted in a reduction of RhoA and Rac1 activation in both the cytoplasmic and membrane-bound forms, in preservation of the expression of the epithelial cell markers E-cadherin and cytokeratin-19, and in a decrease in Fn-EDA expression, a marker for the myofibroblast phenotype. The decreased levels of activated RhoA and Rac1 in both the cytoplasmic and membrane fractions of the cells were reversed by geranylgeranyl pyrophosphate and mevalonate, and thus attributable to the inhibition of isoprenylation of RhoGTPases by statins. CONCLUSION: This phenomenon could explain the beneficial effect of statins on EMT and on renal fibrosis prevention.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos
10.
Respir Res ; 6: 56, 2005 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-15946381

RESUMO

BACKGROUND: Fibroblastic foci are characteristic features in lung parenchyma of patients with idiopathic pulmonary fibrosis (IPF). They comprise aggregates of mesenchymal cells which underlie sites of unresolved epithelial injury and are associated with progression of fibrosis. However, the cellular origins of these mesenchymal phenotypes remain unclear. We examined whether the potent fibrogenic cytokine TGF-beta1 could induce epithelial mesenchymal transition (EMT) in the human alveolar epithelial cell line, A549, and investigated the signaling pathway of TGF-beta1-mediated EMT. METHODS: A549 cells were examined for evidence of EMT after treatment with TGF-beta1. EMT was assessed by: morphology under phase-contrast microscopy; Western analysis of cell lysates for expression of mesenchymal phenotypic markers including fibronectin EDA (Fn-EDA), and expression of epithelial phenotypic markers including E-cadherin (E-cad). Markers of fibrogenesis, including collagens and connective tissue growth factor (CTGF) were also evaluated by measuring mRNA level using RT-PCR, and protein by immunofluorescence or Western blotting. Signaling pathways for EMT were characterized by Western analysis of cell lysates using monoclonal antibodies to detect phosphorylated Erk1/2 and Smad2 after TGF-beta1 treatment in the presence or absence of MEK inhibitors. The role of Smad2 in TGF-beta1-mediated EMT was investigated using siRNA. RESULTS: The data showed that TGF-beta1, but not TNF-alpha or IL-1beta, induced A549 cells with an alveolar epithelial type II cell phenotype to undergo EMT in a time-and concentration-dependent manner. The process of EMT was accompanied by morphological alteration and expression of the fibroblast phenotypic markers Fn-EDA and vimentin, concomitant with a downregulation of the epithelial phenotype marker E-cad. Furthermore, cells that had undergone EMT showed enhanced expression of markers of fibrogenesis including collagens type I and III and CTGF. MMP-2 expression was also evidenced. TGF-beta1-induced EMT occurred through phosphorylation of Smad2 and was inhibited by Smad2 gene silencing; MEK inhibitors failed to attenuate either EMT-associated Smad2 phosphorylation or the observed phenotypic changes. CONCLUSION: Our study shows that TGF-beta1 induces A549 alveolar epithelial cells to undergo EMT via Smad2 activation. Our data support the concept of EMT in lung epithelial cells, and suggest the need for further studies to investigate the phenomenon.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Fator de Crescimento Transformador beta/administração & dosagem , Caderinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibronectinas/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Fator de Crescimento Transformador beta1
11.
Exp Cell Res ; 307(2): 305-14, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15950619

RESUMO

Transforming growth factor-beta (TGFbeta) drives fibrosis in diseases such as diabetic nephropathy (DN). Connective tissue growth factor (CTGF; CCN2) has also been implicated in this, but the molecular mechanism is unknown. We show that CTGF enhances the TGFbeta/Smad signaling pathway by transcriptional suppression of Smad 7 following rapid and sustained induction of the transcription factor TIEG-1. Smad 7 is a known antagonist of TGFbeta signaling and TIEG-1 is a known repressor of Smad 7 transcription. CTGF enhanced TGFbeta-induced phosphorylation and nuclear translocation of Smad 2 and Smad 3 in mesangial cells. Antisense oligonucleotides directed against TIEG-1 prevented CTGF-induced downregulation of Smad 7. CTGF enhanced TGFbeta-stimulated transcription of the SBE4-Luc reporter gene and this was markedly reduced by TIEG-1 antisense oligonucleotides. Expression of the TGFbeta-responsive genes PAI-1 and Col III over 48 h was maximally stimulated by TGFbeta+CTGF compared to TGFbeta alone, while CTGF alone had no significant effect. TGFbeta-stimulated expression of these genes was markedly reduced by both CTGF and TIEG-1 antisense oligonucleotides, consistent with the endogenous induction of CTGF by TGFbeta. We propose that under pathological conditions, where CTGF expression is elevated, CTGF blocks the negative feedback loop provided by Smad 7, allowing continued activation of the TGFbeta signaling pathway.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Mesângio Glomerular/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Colágeno Tipo III/genética , Fator de Crescimento do Tecido Conjuntivo , Inibidor de Quinase Dependente de Ciclina p15 , Proteínas de Ligação a DNA/genética , Fatores de Transcrição de Resposta de Crescimento Precoce , Expressão Gênica/efeitos dos fármacos , Mesângio Glomerular/citologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Fatores de Transcrição Kruppel-Like , Oligorribonucleotídeos Antissenso/farmacologia , Fosforilação/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/genética , Proteínas Recombinantes/farmacologia , Proteínas Smad , Proteína Smad2 , Proteína Smad3 , Proteína Smad7 , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Proteínas Supressoras de Tumor/genética
12.
J Am Soc Nephrol ; 16(7): 1977-84, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15901767

RESUMO

ESRD is characterized by an interstitial infiltrate of inflammatory cells in association with tubular atrophy, epithelial mesenchymal transdifferentiation (EMT), and interstitial fibrosis. Human proximal tubular epithelial cells (HK2 cells) undergo EMT in response to activated PBMC conditioned medium (aPBMC-CM), showing acquisition of a fibroblastoid morphology, increased fibronectin-EDA (EDA) expression, loss of junctional E-cadherin localization, and cytokeratin 19 (K19) expression. The signaling pathway(s) that regulates EMT in response to aPBMC-CM is not well understood. This study shows that aPBMC-CM induces a rapid activation of RhoA, Rac1, and Cdc42 activity in HK2 cells from 15 min to 48 h. Moreover, infection with adenovirus expressing constitutively active RhoA, Rac1, and Cdc42 significantly increased the expression of EDA and downregulated expression of E-cadherin and K19. Dominant negative RhoA expression suppressed aPBMC-CM-induced upregulation of EDA but did not restore the expression of E-cadherin and K19. Constitutively active RhoA activated the Rho kinase and its downstream effectors, whereas constitutively active Rac1 and Cdc42 activated the P21-activated protein kinase in epithelial cells. In further experiments, HK2 cells were treated with toxin B, exoenzyme C3, Y-27632, and HA1077. These strategies, inhibiting the Rho/Rho kinase pathway, as well as the Rac1/Cdc42/P21-activated protein kinase pathway, blocked transdifferentiation of HK2 cells in response to aPBMC-CM. To conclude, these results indicate that aPBMC-CM activates RhoA, Rac1, and Cdc42 and their downstream effectors, leading to HK2 cells undergoing transdifferentiation. Therefore, activation of small RhoGTPases is a key step in the mechanism of EMT and likely to be a contributor to tubulointerstitial fibrosis.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Túbulos Renais Proximais/fisiologia , Mesoderma/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados , Fibrose , Humanos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/patologia , Leucócitos Mononucleares , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
13.
J Am Soc Nephrol ; 16(2): 340-51, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15601748

RESUMO

Connective tissue growth factor (CTGF) is implicated as a factor promoting tissue fibrosis in several disorders, including diabetic nephropathy. However, the molecular mechanism(s) by which it functions is not known. CTGF rapidly activates several intracellular signaling molecules in human mesangial cells (HMC), including extracellular signal-related kinase 1/2, Jun NH(2)-terminal kinase, protein kinase B, CaMK II, protein kinase Calpha, and protein kinase Cdelta, suggesting that it functions via a signaling receptor. Treating HMC with CTGF stimulated tyrosine phosphorylation of proteins 75 to 80 and 140 to 180 kD within 10 min, and Western blot analysis of anti-phosphotyrosine immunoprecipitates identified the neurotrophin receptor TrkA (molecular weight approximately 140 kD). Cross-linking rCTGF to cell surface proteins with 3,3'-dithiobis(sulfosuccinimidylpropionate) revealed that complexes formed with TrkA and with the general neurotrophin co-receptor p75(NTR). rCTGF stimulated phosphorylation of TrkA (tyr 490, 674/675). K252a, a known selective inhibitor of Trk, blocked this phosphorylation, CTGF-induced activation of signaling proteins, and CTGF-dependent induction of the transcription factor TGF-beta-inducible early gene in HMC. It is concluded that TrkA serves as a tyrosine kinase receptor for CTGF.


Assuntos
Mesângio Glomerular/citologia , Proteínas Imediatamente Precoces/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Receptor trkA/efeitos dos fármacos , Receptor trkA/fisiologia , Transdução de Sinais/fisiologia , Adulto , Sequência de Bases , Sítios de Ligação , Western Blotting , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo , Reagentes de Ligações Cruzadas , Imunofluorescência , Humanos , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade
14.
Curr Opin Nephrol Hypertens ; 13(1): 53-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15090860

RESUMO

PURPOSE OF REVIEW: Connective tissue growth factor (CCN2) has recently received much attention as a possible key determinant of progressive renal fibrosis. However, the mechanism(s) by which this growth factor functions is not known. The purpose of this review is to summarize and discuss the recent findings regarding the possible mechanisms involved. RECENT FINDINGS: Emerging evidence from in-vitro studies of renal cells indicates that connective tissue growth factor is a crucial mediator for transforming growth factor-beta-induced cellular dysfunction, manifest by increased cellular hypertrophy, synthesis of extracellular matrix proteins and their deposition and assembly around the cells. Indeed, recent evidence suggests that the interrelationship between connective tissue growth factor and transforming growth factor-beta is stronger than first thought. While transforming growth factor-beta induces the expression of connective tissue growth factor, the latter plays a key role in both bioactivation of latent transforming growth factor-beta and the promotion of its Smad signalling activity. SUMMARY: Connective tissue growth factor is clearly implicated in the pathogenesis of progressive renal disease. Although there is much to learn about the production, function, and mechanism of action of connective tissue growth factor, some progress has been made in understanding the molecular basis of its relationship with transforming growth factor-beta. Elucidating the signal transduction pathways activated by connective tissue growth factor will also definitely help to clarify other actions of connective tissue growth factor which may be independent of transforming growth factor-beta. Because of the inflammatory and immunosuppressive properties of transforming growth factor-beta, connective tissue growth factor seems to be an attractive alternative therapeutic target for combating renal fibrosis.


Assuntos
Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Nefropatias/fisiopatologia , Animais , Fator de Crescimento do Tecido Conjuntivo , Humanos , Proteínas Imediatamente Precoces/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Rim/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
J Am Soc Nephrol ; 15(1): 21-32, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14694154

RESUMO

Interactions between inflammatory infiltrates and resident tubular epithelial cells may play important roles in the development of tubulointerstitial fibrosis, by promoting epithelial cell-myofibroblast transdifferentiation (EMT). Human proximal tubular epithelial cells transdifferentiated to myofibroblasts after treatment with activated PBMC conditioned medium. mRNA and protein levels for alpha-smooth muscle actin, collagen I, and fibronectin EDA(+) (markers for the myofibroblastic phenotype) were increased, whereas those for E-cadherin and cytokeratin 19 (markers for the epithelial phenotype) were decreased. cDNA microarray analysis was used to identify other changes in gene expression that might point to novel molecular mechanisms driving EMT. Of 1176 array genes, 61 demonstrated at least a twofold change at at least two consecutive time points, of the five time points examined (0.5, 4, 8, 16, and 48 h). Of these genes, 59% were upregulated and 41% were downregulated. The array indicated upregulation of expression of the oncostatin M (OSM)-specific receptor beta subunit from 4 to 48 h after exposure of kidney epithelial cells to activated PBMC conditioned medium, which contained high levels of OSM. In additional experiments, it was demonstrated that OSM induced EMT. OSM activated the Jak/Stat signaling pathway in epithelial cells, and a specific inhibitor of Jak2 blocked both its phosphorylation after exposure to OSM and the induction of alpha-actin and loss of cytokeratin 19 expression. Therefore, OSM is a novel inducer of EMT and is likely to be one of several cytokines produced by inflammatory infiltrates that contribute to this and subsequent tubulointerstitial fibrosis.


Assuntos
Diferenciação Celular , Citocinas/fisiologia , Células Epiteliais/citologia , Fibroblastos/citologia , Leucócitos Mononucleares/metabolismo , Mioblastos/citologia , Peptídeos/fisiologia , Transdução de Sinais , Diferenciação Celular/genética , Células Cultivadas , Expressão Gênica , Humanos , Túbulos Renais/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Oncostatina M , Urotélio/citologia
16.
Arthritis Rheum ; 48(12): 3452-63, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14673996

RESUMO

OBJECTIVE: To investigate the development of osteoarthritis (OA) after transection of the medial collateral ligament and partial medial meniscectomy in mice in which genes encoding either interleukin-1beta (IL-1beta), IL-1beta-converting enzyme (ICE), stromelysin 1, or inducible nitric oxide synthase (iNOS) were deleted. METHODS: Sectioning of the medial collateral ligament and partial medial meniscectomy were performed on right knee joints of wild-type and knockout mice. Left joints served as unoperated controls. Serial histologic sections were obtained from throughout the whole joint of both knees 4 days or 1, 2, 3, or 4 weeks after surgery. Sections were graded for OA lesions on a scale of 0-6 and were assessed for breakdown of tibial cartilage matrix proteoglycan (aggrecan) and type II collagen by matrix metalloproteinases (MMPs) and aggrecanases with immunohistochemistry studies using anti-VDIPEN, anti-NITEGE, and Col2-3/4C(short) neoepitope antibodies. Proteoglycan depletion was assessed by Alcian blue staining and chondrocyte cell death, with the TUNEL technique. RESULTS: All knockout mice showed accelerated development of OA lesions in the medial tibial cartilage after surgery, compared with wild-type mice. ICE-, iNOS-, and particularly IL-1beta-knockout mice developed OA lesions in the lateral cartilage of unoperated limbs. Development of focal histopathologic lesions was accompanied by increased levels of MMP-, aggrecanase-, and collagenase-generated cleavage neoepitopes in areas around lesions, while nonlesional areas showed no change in immunostaining. Extensive cell death was also detected by TUNEL staining in focal areas around lesions. CONCLUSION: We postulate that deletion of each of these genes, which encode molecules capable of producing degenerative changes in cartilage, leads to changes in the homeostatic controls regulating the balance between anabolism and catabolism, favoring accelerated cartilage degeneration. These observations suggest that these genes may play important regulatory roles in maintaining normal homeostasis in articular cartilage matrix turnover.


Assuntos
Caspase 1/genética , Proteínas da Matriz Extracelular , Interleucina-1/genética , Metaloproteinase 3 da Matriz/genética , Óxido Nítrico Sintase/genética , Osteoartrite do Joelho/fisiopatologia , Osteoartrite do Joelho/cirurgia , Proteínas ADAM , Proteína ADAMTS4 , Proteína ADAMTS5 , Agrecanas , Azul Alciano , Animais , Anticorpos , Colágeno Tipo II/imunologia , Colágeno Tipo II/metabolismo , Corantes , Fragmentação do DNA , Deleção de Genes , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Lectinas Tipo C , Masculino , Ligamento Colateral Médio do Joelho/cirurgia , Meniscos Tibiais/cirurgia , Metaloendopeptidases/imunologia , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II , Oligopeptídeos , Osteoartrite do Joelho/patologia , Fragmentos de Peptídeos , Complicações Pós-Operatórias/patologia , Pró-Colágeno N-Endopeptidase , Proteoglicanas/metabolismo , Coloração e Rotulagem
17.
J Am Soc Nephrol ; 14(5): 1358-73, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12707406

RESUMO

Diabetic nephropathy is characterized by excessive deposition of extracellular matrix proteins in the mesangium and basement membrane of the glomerulus and in the renal tubulointerstitium. This review summarizes the main changes in protein composition of the glomerular mesangium and basement membrane and the evidence that, in the mesangium, these are initiated by changes in glucose metabolism and the formation of advanced glycation end products. Both processes generate reactive oxygen species (ROS). The review includes discussion of how ROS may activate intracellular signaling pathways leading to the activation of redox-sensitive transcription factors. This in turn leads to change in the expression of genes encoding extracellular matrix proteins and the protease systems responsible for their turnover.


Assuntos
Nefropatias Diabéticas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Mesângio Glomerular/metabolismo , Animais , Humanos
18.
J Am Soc Nephrol ; 14(3): 601-10, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12595495

RESUMO

Excessive deposition of fibronectin in the glomerular mesangium in diabetic nephropathy (DN) is partly due to the induction of transforming growth factor-beta (TGF-beta) by high glucose. TGF-beta induces its downstream mediator connective tissue growth factor (CTGF), which stimulates fibronectin matrix synthesis, a process that requires the presence of alpha5beta1 integrin. Although TGF-beta has been shown to upregulate alpha5beta1 integrin expression in human mesangial cells (HMC), little is known about the effect of CTGF on levels of this receptor. This study tested whether CTGF modulates alpha5beta1 expression by HMC in culture and whether changes induced by TGF-beta are mediated through the induction of CTGF. FACS analysis showed that both TGF-beta and CTGF significantly increased cell-surface alpha5beta1 levels compared with basal conditions. RT-PCR indicated that the changes were at the level of transcription. Treatment of cells with TGF-beta and antisense CTGF oligonucleotides significantly reduced the TGF-beta-induced increases in alpha5beta1 levels. CTGF and TGF-beta also significantly increased levels of ligand-occupied cell-surface beta1 integrins and cell adhesion to fibronectin, the main alpha5beta1 substrate. Antisense CTGF significantly reduced the number of adherent cells from TGF-beta-stimulated cultures. Finally, alpha5beta1 blocking antibodies inhibited HMC fibronectin matrix deposition, confirming the importance of this receptor for this process. Taken together, these data provide evidence that CTGF controls alpha5beta1 expression by HMC in vitro. Alterations in alpha5beta1 levels induced by TGF-beta are mediated at least in part through the induction of CTGF, and specific targeting of either alpha5beta1 or CTGF could be useful in controlling excessive fibronectin matrix production in DN.


Assuntos
Fibronectinas/metabolismo , Mesângio Glomerular/metabolismo , Proteínas Imediatamente Precoces/farmacologia , Integrina alfa5beta1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Mitógenos/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Anticorpos/farmacologia , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo , Ácido Desoxicólico , Detergentes , Matriz Extracelular/metabolismo , Mesângio Glomerular/citologia , Humanos , Técnicas In Vitro , Integrina alfa5beta1/imunologia , Ligantes , Solubilidade , Regulação para Cima/efeitos dos fármacos
19.
J Am Soc Nephrol ; 13(10): 2437-45, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12239232

RESUMO

Connective tissue growth factor (CTGF) is now considered to be one of the important driver molecules for the pathogenesis of diabetic nephropathy (DN) and possibly many other fibrotic disorders. However, the molecular mechanisms by which CTGF functions remain to be established. In an attempt to define these mechanisms, this study was designed to investigate whether CTGF has any effect on the cell cycle of human mesangial cells (HMC), which are known to undergo hypertrophy in DN. This report provides the first evidence that CTGF is a hypertrophic factor for HMC. CTGF stimulates HMC to actively enter the G(1) phase from G(0), but they do not then progress further through the cell cycle. The molecular mechanisms underlying this G(1) phase arrest appear to be due to the induction of the cyclin-dependent kinase inhibitors (CDKI) p15(INK4), p21(Cip1), and p27(Kip1), which are known to bind and inactivate cyclinD/CDK4/6 and the cyclin E/CDK2 kinase complexes. This could account for the maintenance of pRb protein in a non- or very low-phosphorylated state, preventing cell cycle progression. Using CTGF antisense oligonucleotides, the results also indicate that the previously identified transforming growth factor-beta (TGF-beta)-induced hypertrophy in mesangial cells is CTGF-dependent. Mesangial cell hypertrophy is one of the earliest abnormalities of diabetic nephropathy; therefore, therapeutic strategies targeting CTGF may be beneficial in controlling DN.


Assuntos
Ciclo Celular/fisiologia , Mesângio Glomerular/patologia , Proteínas Imediatamente Precoces/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Mitógenos/fisiologia , Sequência de Aminoácidos/genética , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Ciclinas/metabolismo , Indução Enzimática , Inibidores Enzimáticos/metabolismo , Fase G1 , Humanos , Hipertrofia , Proteínas Imediatamente Precoces/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitógenos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
20.
Biochem J ; 362(Pt 3): 643-9, 2002 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11879191

RESUMO

Transforming growth factor-beta (TGFbeta) is a key mediator of extracellular matrix (ECM) accumulation in sclerotic kidney diseases such as diabetic nephropathy. One of the main target cells for TGFbeta in the kidney are glomerular mesangial cells, which respond by increasing expression of ECM proteins, such as collagens, laminin and fibronectin, while suppressing the expression of ECM-degrading proteases and increasing the synthesis of ECM protease inhibitors, including plasminogen activator inhibitor-1. Previous studies have shown that exposure of mesangial cells to chronic high-glucose conditions, such as those seen in diabetes, increases ECM deposition in a mechanism involving glucose-mediated up-regulation of TGFbeta expression. Naturally occurring inhibitors of this TGFbeta-dependent fibrotic response include decorin, a small leucine-rich proteoglycan. While the mechanism by which TGFbeta stimulates gene expression via the Smad signal-transduction pathway is becoming clear, the precise mechanism by which decorin may impinge upon TGFbeta activity remains to be established. In this study, for the first time we provide evidence that decorin can disrupt glucose- and TGFbeta/Smad-dependent transcriptional events in human mesangial cells through a mechanism that involves an increase in Ca(2+) signalling, the activation of Ca(2+)/calmodulin-dependent protein kinase II and ensuing phosphorylation of Smad2 at Ser-240. We show that decorin also induces Ser-240 phospho-Smad hetero-oligomerization with Smad4 and the nuclear localization of this complex independently of TGFbeta receptor activation. Thus, in human mesangial cells, the mechanism of decorin-mediated inhibition of TGFbeta signalling may involve activation of Ca(2+) signalling, the subsequent phosphorylation of Smad2 at a key regulatory site, and the sequestration of Smad4 in the nucleus.


Assuntos
Cálcio/fisiologia , Proteínas de Ligação a DNA/metabolismo , Mesângio Glomerular/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Proteoglicanas/farmacologia , Serina , Transativadores/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Benzilaminas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Linhagem Celular Transformada , Primers do DNA , Proteínas de Ligação a DNA/química , Decorina , Inibidores Enzimáticos/farmacologia , Proteínas da Matriz Extracelular , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Mesângio Glomerular/efeitos dos fármacos , Humanos , Luciferases/genética , Fosforilação , Inibidores de Proteínas Quinases , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad2 , Sulfonamidas/farmacologia , Transativadores/química , Transfecção , Fator de Crescimento Transformador beta/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...