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.
J Am Soc Nephrol ; 31(1): 85-100, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31690575

RESUMO

BACKGROUND: The matricellular protein periostin has been associated with CKD progression in animal models and human biopsy specimens. Periostin functions by interacting with extracellular matrix components to drive collagen fibrillogenesis and remodeling or by signaling through cell-surface integrin receptors to promote cell adhesion, migration, and proliferation. However, its role in AKI is unknown. METHODS: We used mice with conditional tubule-specific overexpression of periostin or knockout mice lacking periostin expression in the renal ischemia-reperfusion injury model, and primary cultures of isolated tubular cells in a hypoxia-reoxygenation model. RESULTS: Tubular epithelial cells showed strong production of periostin during the repair phase of ischemia reperfusion. Periostin overexpression protected mice from renal injury compared with controls, whereas knockout mice showed increased tubular injury and deteriorated renal function. Periostin interacted with its receptor, integrin-ß1, to inhibit tubular cell cycle arrest and apoptosis in in vivo and in vitro models. After ischemia-reperfusion injury, periostin-overexpressing mice exhibited diminished expression of proinflammatory molecules and had more F4/80+ macrophages compared with knockout mice. Macrophages from periostin-overexpressing mice showed increased proliferation and expression of proregenerative factors after ischemia-reperfusion injury, whereas knockout mice exhibited the opposite. Coculturing a macrophage cell line with hypoxia-treated primary tubules overexpressing periostin, or treating such macrophages with recombinant periostin, directly induced macrophage proliferation and expression of proregenerative molecules. CONCLUSIONS: In contrast to the detrimental role of periostin in CKD, we discovered a protective role of periostin in AKI. Our findings suggest periostin may be a novel and important mediator of mechanisms controlling renal repair after AKI.


Assuntos
Injúria Renal Aguda , Moléculas de Adesão Celular/fisiologia , Proliferação de Células , Macrófagos/fisiologia , Injúria Renal Aguda/etiologia , Animais , Modelos Animais de Doenças , Rim/irrigação sanguínea , Masculino , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia
2.
Kidney Int ; 94(1): 126-138, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29751972

RESUMO

Acute kidney injury is a major risk factor for subsequent chronic renal and/or cardiovascular complications. Previous studies have shown that Notch3 was de novo expressed in the injured renal epithelium in the early phases of chronic kidney disease. Here we examined whether Notch3 is involved in the inflammatory response and the epithelial cell damage that typifies ischemic kidneys using Notch3 knockout mice and mice with short-term activated Notch3 signaling (N3ICD) in renal epithelial cells. After ischemia/reperfusion, N3ICD mice showed exacerbated infiltration of inflammatory cells and severe tubular damage compared to control mice. Inversely, Notch3 knockout mice were protected against ischemia/reperfusion injury. Renal macrophages derived from Notch3 knockout mice failed to activate proinflammatory cytokines. Chromatin immunoprecipitation analysis of the Notch3 promoter identified NF-κB as the principal inducer of Notch3 in ischemia/reperfusion. Thus, Notch3 induced by NF-κB in the injured epithelium sustains a proinflammatory environment attracting activated macrophages to the site of injury leading to a rapid deterioration of renal function and structure. Hence, targeting Notch3 may provide a novel therapeutic strategy against ischemia/reperfusion and acute kidney injury by preservation of epithelial structure and disruption of proinflammatory signaling.


Assuntos
Injúria Renal Aguda/patologia , Túbulos Renais/patologia , Receptor Notch3/metabolismo , Traumatismo por Reperfusão/complicações , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/imunologia , Animais , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/metabolismo , Epitélio/patologia , Humanos , Túbulos Renais/imunologia , Túbulos Renais/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Receptor Notch3/genética
3.
Nephron ; 137(3): 212-220, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28743124

RESUMO

Discoidin domain receptors (DDRs) are a family of 2 non-integrin collagen receptors, DDR1 and DDR2, which display a tyrosine kinase activity. They are mainly expressed during embryonic development and their role during adulthood is very limited. DDR1 has been widely studied in several types of cancers, in atherosclerosis and fibrosis, but also in chronic kidney disease (CKD). This review focuses on the role of DDR1 in chronic nephropathies and on the effect of its deletion in the pathological processes involved in renal disease progression. DDR1 was shown to be de novo expressed in several models of experimental CKD. Its genetic or pharmaco-genetic inhibition led to the preservation of renal structure and function, and to decreased inflammatory influx and fibrosis. Furthermore, delayed pharmaco-genetic inhibition of DDR1 led to significant protection in models of renal disease. These results demonstrate the involvement of DDR1 in inflammatory and fibrotic processes occurring during CKD and the beneficial effect of its inhibition. Thus, DDR1 could be an interesting therapeutic target to treat renal pathologies.


Assuntos
Receptor com Domínio Discoidina 1/fisiologia , Fibrose/fisiopatologia , Inflamação/fisiopatologia , Nefropatias/fisiopatologia , Animais , Receptor com Domínio Discoidina 1/genética , Humanos , Falência Renal Crônica/genética , Falência Renal Crônica/fisiopatologia , Neoplasias/genética , Neoplasias/fisiopatologia
4.
J Am Soc Nephrol ; 28(5): 1475-1490, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27920156

RESUMO

De novo expression in the kidney of periostin, a protein involved in odontogenesis and osteogenesis, has been suggested as a biomarker of renal disease. In this study, we investigated the mechanism(s) of induction and the role of periostin in renal disease. Using a combination of bioinformatics, reporter assay, and chromatin immunoprecipitation analyses, we found that NFκB and other proinflammatory transcription factors induce periostin expression in vitro and that binding of these factors on the periostin promoter is enriched in glomeruli during experimental GN. Mice lacking expression of periostin displayed preserved renal function and structure during GN. Furthermore, delayed administration of periostin antisense oligonucleotides in wild-type animals with GN reversed already established proteinuria, diminished tissue inflammation, and improved renal structure. Lack of periostin expression also blunted the de novo renal expression of integrin-ß3 and phosphorylation of focal adhesion kinase and AKT, known mediators of integrin-ß3 signaling that affect cell motility and survival, observed during GN in wild-type animals. In vitro, recombinant periostin increased the expression of integrin-ß3 and the concomitant phosphorylation of focal adhesion kinase and AKT in podocytes. Notably, periostin and integrin-ß3 were highly colocalized in biopsy specimens from patients with inflammatory GN. These results demonstrate that interplay between periostin and renal inflammation orchestrates inflammatory and fibrotic responses, driving podocyte damage through downstream activation of integrin-ß3 signaling. Targeting periostin may be a novel therapeutic strategy for treating CKD.


Assuntos
Moléculas de Adesão Celular/fisiologia , Integrina beta3/fisiologia , Nefropatias/etiologia , NF-kappa B/fisiologia , Animais , Feminino , Glomerulonefrite/complicações , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
5.
Nephrol Dial Transplant ; 30(12): 1965-71, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25829327

RESUMO

The incidence and prevalence of chronic kidney disease represents an important problem for public health. In renal diseases, the main histologic alterations derive from the development of renal fibrosis which results from the loss of the balance between pro- and anti-fibrotic factors. Tyrosine kinase receptors (RTKs) and matricellular proteins (MPs) are nowadays studied as potential modulators of renal injury. RTKs regulate cell cycle, migration, metabolism and cellular differentiation. Discoidin domain receptor-1 (DDR-1) is an RTK that has been extensively studied in cancer, and lung and renal diseases. It modulates inflammatory recruitment, extracellular matrix deposition and fibrosis; in renal diseases, it appears to act independently of the underlying disease. MPs regulate cell-matrix interactions and matrix accumulation, cellular adhesion and migration, and expression of inflammatory cells. Periostin is an MP, mainly studied in bone, heart, lung and cancer. Several studies demonstrated that it mediates cell-matrix interactions, migration of inflammatory cells and development of fibrosis. Recently, it has been reported in several nephropathies. In this review, we discuss the potential pathological roles of DDR-1 and periostin focussing on the kidney in both experimental models and human diseases.


Assuntos
Moléculas de Adesão Celular/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Insuficiência Renal Crônica/metabolismo , Receptor com Domínio Discoidina 1 , Humanos
6.
J Am Soc Nephrol ; 26(7): 1561-75, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25421557

RESUMO

Notch3 expression is found in the glomerular podocytes of patients with lupus nephritis or focal segmental GN but not in normal kidneys. Here, we show that activation of the Notch3 receptor in the glomeruli is a turning point inducing phenotypic changes in podocytes promoting renal inflammation and fibrosis and leading to disease progression. In a model of rapidly progressive GN, Notch3 expression was induced by several-fold in podocytes concurrently with disease progression. By contrast, mice lacking Notch3 expression were protected because they exhibited less proteinuria, uremia, and inflammatory infiltration. Podocyte outgrowth from glomeruli isolated from wild-type mice during the early phase of the disease was higher than outgrowth from glomeruli of mice lacking Notch3. In vitro studies confirmed that podocytes expressing active Notch3 reorganize their cytoskeleton toward a proliferative/migratory and inflammatory phenotype. We then administered antisense oligodeoxynucleotides targeting Notch3 or scramble control oligodeoxynucleotides in wild-type mice concomitant to disease induction. Both groups developed chronic renal disease, but mice injected with Notch3 antisense had lower values of plasma urea and proteinuria and inflammatory infiltration. The improvement of renal function was accompanied by fewer deposits of fibrin within the glomeruli and by decreased peritubular inflammation. Finally, abnormal Notch3 staining was observed in biopsy samples of patients with crescentic GN. These results demonstrate that abnormal activation of Notch3 may be involved in the progression of renal disease by promoting migratory and proinflammatory pathways. Inhibiting Notch3 activation could be a novel, promising approach to treat GN.


Assuntos
Regulação da Expressão Gênica , Glomerulosclerose Segmentar e Focal/genética , Glomérulos Renais/patologia , Podócitos/patologia , Receptores Notch/genética , Ativação Transcricional/genética , Análise de Variância , Animais , Nitrogênio da Ureia Sanguínea , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Imuno-Histoquímica , Glomérulos Renais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Podócitos/metabolismo , Proteinúria/fisiopatologia , RNA Mensageiro/análise , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/métodos , Receptor Notch3 , Valores de Referência , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Ovinos , Regulação para Cima
7.
J Pathol ; 228(3): 286-99, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22806125

RESUMO

Kidney diseases impart a vast burden on affected individuals and the overall health care system. Progressive loss of renal parenchymal cells and functional decline following injury are often observed. Notch-1 and -2 receptors are crucially involved in nephron development and contribute to inflammatory kidney diseases. We specifically determined the participation of receptor Notch-3 following tubulointerstitial injury and in inflammatory responses. Here we show by heat map analyses that Notch-3 transcripts are up-regulated in human kidney diseases. A similar response was corroborated with kidney cells following TGF-ß exposure in vitro. The murine unilateral ureteral obstruction (UUO) model mirrors hallmarks of tubulointerstitial injury and damage. A subset of tubular and interstitial cells demonstrated up-regulated Notch-3 receptor expression in diseased animals. We hypothesized a relevance of Notch-3 receptors for the chemotactic response. To address this question, animals with genetic ablation of receptor Notch-3 were analysed following UUO. As a result, we found that Notch-3-deficient animals are protected from tubular injury and cell loss with significantly reduced interstitial collagen deposition. Monocytic cell infiltration was significantly reduced and retarded, likely due to abrogated chemokine synthesis. A cell model was set up that mimics enhanced receptor Notch-3 expression and activation. Here a pro-mitogenic response was seen with activated signalling in tubular cells and fibroblasts. In conclusion, Notch-3 receptor fulfils non-redundant roles in the inflamed kidney that may not be replaced by other Notch receptor family members. Thus, specific blockade of this receptor may be suitable as therapeutic option to delay progression of kidney disease.


Assuntos
Inflamação/fisiopatologia , Rim/fisiopatologia , Nefrite Intersticial/fisiopatologia , Receptores Notch/fisiologia , Obstrução Ureteral/fisiopatologia , Animais , Biópsia , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Fibrose , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-2 , Rim/metabolismo , Rim/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite Intersticial/metabolismo , Nefrite Intersticial/patologia , Ratos , Receptor Notch1/fisiologia , Receptor Notch3 , Receptores Notch/deficiência , Receptores Notch/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima/efeitos dos fármacos , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
8.
Kidney Int ; 81(12): 1226-38, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22418977

RESUMO

Thrombospondin-1 (TSP-1) is an endogenous activator of transforming growth factor-ß (TGF-ß), and an anti-angiogenic factor, which may prevent kidney repair. Here we investigated whether TSP-1 is involved in the development of chronic kidney disease using rats with unilateral ureteral obstruction, a well-known model to study renal fibrosis. Obstruction of 10 days duration induced inflammation, tubular cell atrophy, dilation, apoptosis, and proliferation, leading to interstitial fibrosis. TSP-1 expression was increased in parallel to that of collagen III and TGF-ß. Relief of the obstruction at day 10 produced a gradual improvement in renal structure and function, the reappearance of peritubular capillaries, and restoration of renal VEGF content over a 7- to 15-day post-relief period. TSP-1 expression decreased in parallel with that of TGF-ß1 and collagen III. Mice in which the TSP-1 gene was knocked out displayed less inflammation and had better preservation of renal tissue and the peritubular capillary network compared to wild-type mice. Additional studies showed that the inflammatory effect of TSP-1 was mediated, at least in part, by monocyte chemoattractant protein-1 and activation of the Th17 pathway. Thus, TSP-1 is an important profibrotic and inflammatory mediator of renal disease. Blockade of its action may be a treatment against the development of chronic kidney disease.


Assuntos
Mediadores da Inflamação/metabolismo , Nefropatias/etiologia , Rim/metabolismo , Nefrite/etiologia , Trombospondina 1/metabolismo , Obstrução Ureteral/complicações , Animais , Apoptose , Atrofia , Capilares/metabolismo , Capilares/patologia , Proliferação de Células , Quimiocina CCL2/metabolismo , Doença Crônica , Colágeno Tipo III/metabolismo , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Rim/irrigação sanguínea , Rim/patologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Knockout , Nefrectomia , Nefrite/genética , Nefrite/metabolismo , Nefrite/patologia , Nefrite/prevenção & controle , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Trombospondina 1/deficiência , Trombospondina 1/genética , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Nat Med ; 17(10): 1242-50, 2011 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-21946538

RESUMO

Rapidly progressive glomerulonephritis (RPGN) is a life-threatening clinical syndrome and a morphological manifestation of severe glomerular injury that is marked by a proliferative histological pattern ('crescents') with accumulation of T cells and macrophages and proliferation of intrinsic glomerular cells. We show de novo induction of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in intrinsic glomerular epithelial cells (podocytes) from both mice and humans with RPGN. HB-EGF induction increases phosphorylation of the epidermal growth factor receptor (EGFR, also known as ErbB1) in mice with RPGN. In HB-EGF-deficient mice, EGFR activation in glomeruli is absent and the course of RPGN is improved. Autocrine HB-EGF induces a phenotypic switch in podocytes in vitro. Conditional deletion of the Egfr gene from podocytes of mice alleviates the severity of RPGN. Likewise, pharmacological blockade of EGFR also improves the course of RPGN, even when started 4 d after the induction of experimental RPGN. This suggests that targeting the HB-EGF-EGFR pathway could also be beneficial in treatment of human RPGN.


Assuntos
Receptores ErbB/metabolismo , Glomerulonefrite/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glomérulos Renais/lesões , Glomérulos Renais/fisiopatologia , Insuficiência Renal/etiologia , Análise de Variância , Animais , Western Blotting , Transplante de Medula Óssea , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/genética , Citometria de Fluxo , Glomerulonefrite/complicações , Glomerulonefrite/patologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/genética , Glomérulos Renais/citologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Fosforilação , Podócitos/metabolismo , Quinazolinas , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas , Tirfostinas
10.
Am J Pathol ; 179(1): 83-91, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21640971

RESUMO

The interactions between tubulointerstitial infiltrating cells and the extracellular matrix play an important role in regulating renal fibrosis. Discoidin domain receptor 1 (DDR1) is a nonintegrin tyrosine kinase receptor for collagen implicated in cell adhesion, proliferation, and extracellular matrix remodeling. We have previously demonstrated that transgenic mice lacking DDR1 are protected from hypertension-associated renal fibrosis. The purpose of this study was to determine the role of DDR1 in renal inflammation and fibrosis related to primitive tubulointerstitial injury. After 12 days of unilateral ureteral obstruction (UUO), kidney histopathologic and real-time quantitative PCR analyses were performed in DDR1(-/-) and wild-type mice. DDR1 expression was strongly increased in the obstructed kidney. Wild-type mice developed important perivascular and interstitial inflammation and fibrosis. In comparison, DDR1(-/-) mice displayed reduced accumulation of fibrillar collagen and transforming growth factor ß expression. F4/80(+) cell count and proinflammatory cytokines were remarkably blunted in DDR1(-/-) obstructed kidneys. Leukocyte rolling and adhesion evaluated by intravital microscopy were not different between DDR1(-/-) and wild-type mice. Importantly, macrophages isolated from DDR1(-/-) mice presented similar M1/M2 polarization but displayed impaired migration in response to monocyte chemoattractant protein-1. Together, these data suggest that DDR1 plays an important role in the pathogenesis of renal disease via enhanced inflammation. Inhibition of DDR1 expression or activity may represent a novel therapeutic target against the progression of renal diseases.


Assuntos
Movimento Celular , Fibrose/etiologia , Inflamação/etiologia , Nefropatias/complicações , Receptores Proteína Tirosina Quinases/fisiologia , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Células Cultivadas , Receptor com Domínio Discoidina 1 , Fibrose/patologia , Humanos , Inflamação/patologia , Nefropatias/patologia , Leucócitos/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos
11.
Am J Physiol Renal Physiol ; 297(1): F191-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19420116

RESUMO

The present study investigated mechanisms of regression of renal disease after severe proteinuria by focusing on the interaction among EGF receptors, renal hemodynamics, and structural lesions. The nitric oxide (NO) inhibitor N(G)-nitro-l-arginine-methyl ester (l-NAME) was administered chronically in Sprague-Dawley rats. When proteinuria exceeded 2 g/mmol creatinine, animals were divided into three groups for an experimental period of therapy of 2 wk; in one group, l-NAME was removed to allow reactivation of endogenous NO synthesis; in the two other groups, l-NAME removal was combined with EGF or angiotensin receptor type 1 (AT(1)) antagonism. l-NAME removal partially reduced mean arterial pressure and proteinuria and increased renal blood flow (RBF), but not microvascular hypertrophy. Progression of structural damage was stopped, but not reversed. The administration of an EGF receptor antagonist did not have an additional effect on lowering blood pressure or on renal inflammation but did normalize RBF and afferent arteriole hypertrophy; the administration of an AT(1) antagonist normalized all measured functional and structural parameters. Staining with a specific marker of endothelial integrity indicated loss of functional endothelial cells in the l-NAME removal group; in contrast, in the animals treated with an EGF or AT(1) receptor antagonist, functional endothelial cells reappeared at levels equal to control animals. In addition, afferent arterioles freshly isolated from the l-NAME removal group showed an exaggerated constrictor response to endothelin; this response was blunted in the vessels isolated from the EGF or AT(1) receptor antagonist groups. The EGF receptor is an important mediator of endothelial dysfunction and contributes to the decline of RBF in the chronic kidney disease induced by NO deficiency. The EGF receptor antagonist-induced improvement of RBF is important but not sufficient for a complete reversal of renal disease, because it has little effect on renal inflammation. To achieve full recovery, it is necessary to apply AT(1) receptor antagonism.


Assuntos
Receptores ErbB/antagonistas & inibidores , Hipertensão/complicações , Hipertensão/fisiopatologia , Nefropatias/etiologia , Nefropatias/fisiopatologia , Rim/irrigação sanguínea , Fluxo Sanguíneo Regional/fisiologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/patologia , Arteríolas/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Doença Crônica , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Gefitinibe , Nefropatias/patologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Quinazolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
12.
Nephrol Ther ; 3(4): 157-62, 2007 Jul.
Artigo em Francês | MEDLINE | ID: mdl-17658443

RESUMO

The National Institute of Health and Medical Research (Inserm), the Society of Nephrology, and the French Kidney Foundation recognized the need to create a National Research Program for kidney and urinary tract diseases. They organized a conference gathering 80 researchers to discuss the state-of-the art and evaluate the strengths and weaknesses of kidney and urinary tract disease research in France, and to identify research priorities. From these priorities emerged 11 of common interest: 1) conducting epidemiologic studies; 2) conducting large multicenter cohorts of well-phenotyped patients with blood, urine and biopsy biobanks; 3) developing large scale approach: transcriptomics, proteomics, metabolomics; 4) developing human and animal functional imaging techniques; 5) strengthening the expertise in renal pathology and electrophysiology; 6) developing animal models of kidney injury; 7) identifying nontraumatic diagnostic and prognostic biomarkers; 8) increasing research on the fetal programming of adult kidney diseases; 9) encouraging translational research from bench to bedside and to population; 10) creating centers grouping basic and clinical research workforces with critical mass and adequate logistic support; 11) integrating and developing european research programs.


Assuntos
Nefropatias , Pesquisa/tendências , Doenças Urológicas , Fundações , França/epidemiologia , Humanos , Incidência , Nefropatias/classificação , Nefropatias/epidemiologia , Transplante de Rim/estatística & dados numéricos , Doenças Urológicas/classificação , Doenças Urológicas/epidemiologia
13.
FASEB J ; 20(11): 1936-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16877529

RESUMO

Endothelin-1 (ET-1), a powerful vasoconstrictor, is involved in vasospastic diseases such as coronary artery disease and subarachnoidal hemorrhage, as well as in renal and cardiovascular fibrotic remodeling. Transactivation of the epidermal growth factor receptor (EGFR) mediates ET-1 signaling in vascular smooth muscle cells (VSMCs) and isolated arteries. Moreover, EGFR is required for a full constrictive response to ET-1. However, the relevant mechanisms mediating EGFR transactivation in response to ET-1 have not been identified. The present study used isolated arteries and VSMCs to investigate the role of the EGFR ligand heparin binding-epidermal growth factor (HB-EGF) in ET-1-induced transactivation of EGFR, intracellular calcium mobilization, and VSMCs contraction. While baseline blood pressures were similar in HB-EGF-deficient and in wild-type littermate mice, the vasoconstrictor actions of ET-1 were attenuated in HB-EGF-/- animals. In isolated mouse carotid artery segments mounted in an arteriograph, ET-1 caused only a weak increase in isovolumetric tone in HB-EGF-deficient vessels, and this effect was mimicked by inhibition of EGFR tyrosine kinase or phosphoinositide 3-kinase (PI3K) in wild-type arteries with or without endothelium, indicating a specific role in VSMCs. EGFR or PI3K inhibitors had no effect on KCl-induced contraction, which was normal in HB-EGF-deficient mice. To confirm that the abnormal responses in HB-EGF-deficient mice were due to impaired EGFR signaling, we studied VSMCs from waved-2 (wa2) mice; these animals have a mutation causing a partial loss of function of EGFR tyrosine kinase activity. The ET-1-induced calcium peak was reduced by 30% in VSMCs from wa2 mice and from HB-EGF-/- mice. This effect was reproduced by preincubation of wild-type VSMCs with EGFR inhibitor AG1478 and PI3K inhibitors LY294002 and wortmannin. ProHB-EGF is bound to the cell membrane and released after cleavage by metalloproteinases; its action may contribute to effects of GPCR agonists on cell growth. Pretreatment of mouse VSMCs with batimastat, a metalloproteinase inhibitor, significantly attenuated ET-1-induced [Ca(2+)](i) response in wild-type cells. Human proHB-EGF has been shown to be the endogenous receptor for Corynebacterium diphteriae toxin (DT). Mutated DT toxin (CRM197) is devoid of toxicity but it neutralizes HB-EGF binding to EGFR. Pretreatment of human VSMCs from internal mammary arteries with CRM197 significantly blunted ET-1-stimulated calcium transients. In conclusion, these findings suggest that the mechanism of ET-1-induced vasoconstriction involves HB-EGF-mediated transactivation of the EGFR. This functional cascade requires modulation of agonist-induced calcium transient by EGFR and PI3K with extremely fast kinetics, suggesting a novel paradigm for GPCR-mediated calcium signaling, which may offer future therapeutic targets.


Assuntos
Artérias Carótidas/fisiologia , Endotelina-1/farmacologia , Receptores ErbB/fisiologia , Heparina/fisiologia , Animais , Pressão Sanguínea , Artérias Carótidas/efeitos dos fármacos , Receptores ErbB/deficiência , Receptores ErbB/genética , Heparina/deficiência , Heparina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Mutação Puntual , Cloreto de Potássio/farmacologia , Ligação Proteica
14.
Am J Physiol Renal Physiol ; 289(2): F227-34, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16006588

RESUMO

Recent evidence suggests that the progression of renal fibrosis is a reversible process, at least in experimental models. The present review summarizes the new insights concerning the mechanisms of progression and regression of renal disease and examines this novel evidence under the light of feasibility and transfer to human nephropathies. The involved mechanisms are discussed with particular emphasis on the fibrotic role of vasoactive peptides such as angiotensin II and endothelin and growth factors such as transforming growth factor (TGF)-beta. The possibility of regression is introduced by presenting the in vivo efficiency of antihypertensive treatments and of systems that antagonize the fibrogenic action of TGF-beta such as bone morphogenic protein-7 and HGF. Finally, we provide a brief description of the promising future directions and clinical considerations about the applications of the experimental data to humans.


Assuntos
Nefropatias/patologia , Nefropatias/terapia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Progressão da Doença , Fibrose , Humanos , Inflamação/tratamento farmacológico , Inflamação/patologia , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/antagonistas & inibidores , Peptídeos/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/fisiologia
16.
FASEB J ; 18(7): 926-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15033924

RESUMO

Hypertension is frequently associated with the development of renal vascular and glomerular fibrosis. The purpose of the present study was to investigate whether epidermal growth factor receptor (EGFR) activation participates in the development of renal fibrosis and to test if blockade of EGFR activation would have therapeutic effects. Experiments were performed during nitric oxide (NO) deficiency-induced hypertension in rats (L-NAME model). After 4 weeks of L-NAME treatment, animals developed hypertension associated to deterioration of renal structure and function. Over the same period, EGFR was activated twofold within glomeruli. This activation was accompanied by increased activity of the mitogen-activated protein kinase (MAPK) p42/p44 pathway and exaggerated collagen I expression. Gefitinib, an EGFR-tyrosine kinase inhibitor, given concomitantly with L-NAME, normalized MAPK activation and collagen I expression and prevented the decline of renal function and the development of fibrosis. Since endothelin mediates the L-NAME-induced fibrogenesis, the endothelin-EGFR interaction was tested in transgenic mice expressing luciferase under the control of collagen I-alpha2 promoter: In renal cortex of these animals, the endothelin-induced collagen I gene activity was inhibited by an EGFR-phosphorylation inhibitor. These results provide the first evidence that EGFR activation plays an important role in the progression of renal vascular and glomerular fibrosis.


Assuntos
Colágeno/biossíntese , Receptores ErbB/fisiologia , Hipertensão/complicações , Nefropatias/prevenção & controle , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Óxido Nítrico/deficiência , Quinazolinas/uso terapêutico , Animais , Capilares/efeitos dos fármacos , Capilares/patologia , Colágeno/genética , Colágeno Tipo I , Creatinina/sangue , Endotelina-1/farmacologia , Endotelina-1/toxicidade , Receptores ErbB/antagonistas & inibidores , Fibrose , Gefitinibe , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/prevenção & controle , Hipertensão/induzido quimicamente , Isquemia/induzido quimicamente , Isquemia/prevenção & controle , Rim/irrigação sanguínea , Córtex Renal/metabolismo , Córtex Renal/patologia , Nefropatias/sangue , Nefropatias/induzido quimicamente , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Transgênicos , NG-Nitroarginina Metil Éster/toxicidade , Necrose , Óxido Nítrico/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteinúria/induzido quimicamente , Proteinúria/prevenção & controle , Quinazolinas/farmacologia , Ratos , Ratos Sprague-Dawley
17.
Hypertension ; 42(5): 937-44, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14569001

RESUMO

Inhibition of nitric oxide synthases causes systemic hypertension and renal injury in rats. Our objective was to examine whether omapatrilat, a vasopeptidase inhibitor that inhibits both angiotensin-converting enzyme (ACE) and neutral endopeptidase, could induce better regression of renal injury than ACE inhibitor alone. Ten groups of rats were studied. They were fed either a normal (0.8% NaCl) or a high (4% NaCl) sodium diet. Eight of these groups received NG-nitro-L-arginine methyl ester (L-NAME, 20 mg x kg(-1) x d(-1)) in their drinking water. After 4 weeks, 1 group on each diet was killed and considered the L-NAME group, whereas the others received L-NAME alone, captopril (200 mg x kg(-1) x d(-1)) plus L-NAME, or omapatrilat (80 mg x kg(-1) x d(-1)) plus L-NAME for 4 additional weeks. In rats receiving L-NAME alone for 8 weeks, the mortality rate was approximately 90%, irrespective of the diet. In contrast, all rats survived in the captopril and the omapatrilat groups. In rats fed a normal-sodium diet, captopril and omapatrilat normalized systolic blood pressure and induced a complete regression of renal injury. Creatinine clearance and proteinuria were also normalized. In the high-sodium-diet groups, both treatments were less efficient: blood pressure remained elevated, and the regression of renal fibrosis was only partial. Although proteinuria decreased significantly with captopril or omapatrilat, creatinine clearance remained lower than in the controls. These results demonstrate that, in nitric oxide-deficient rats fed a normal-sodium diet, ACE and vasopeptidase inhibitors exhibit a marked renoprotective effect, whereas these treatments are less efficient in rats fed a high-sodium diet.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Captopril/uso terapêutico , Hipertensão Renal/tratamento farmacológico , Neprilisina/antagonistas & inibidores , Inibidores de Proteases/uso terapêutico , Piridinas/uso terapêutico , Tiazepinas/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Vasos Sanguíneos/patologia , GMP Cíclico/sangue , Endotelina-1/análise , Inibidores Enzimáticos/farmacologia , Fibrose , Hipertensão Renal/induzido quimicamente , Hipertensão Renal/patologia , Hipertensão Renal/fisiopatologia , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio , Taxa de Sobrevida
18.
FASEB J ; 17(2): 327-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12475899

RESUMO

Vascular remodeling and rearrangement of the extracellular matrix formation are among the major adaptive mechanisms in response to a chronic blood pressure increase. Vasoactive peptides, such as endothelin, participate in hypertension-associated vascular fibrosis by stimulating collagen I formation and increasing contractility of arterial wall. In the present study, we tested the hypothesis that activation of the epidermal growth factor (EGF) receptor pathway mediates these events. Experiments were performed in transgenic mice harboring the luciferase gene under the control of the collagen I-alpha2 chain promoter. Endothelin induced a rapid phosphorylation of the mitogen-activated protein kinase (MAPK)/ERK and increased collagen I gene activity in freshly isolated aortas. This effect of endothelin was totally inhibited by an endothelin receptor antagonist, an EGF receptor phosphorylation inhibitor, and a blocker of the MAPK/ERK cascade. In parallel experiments, inhibition of EGF receptor phosphorylation decreased the endothelin-induced pressor effect in isolated aortic rings and in anesthetized animals in vivo. In addition, the endothelin-induced increase of blood pressure was blunted in the waved-2 mice, a strain expressing functionally impaired EGF receptors. Our results provide the first evidence that the EGF receptor mediates at least two of the major actions of endothelin in the vascular tissue: contractility and fibrogenesis.


Assuntos
Aorta/efeitos dos fármacos , Endotelina-1/farmacologia , Receptores ErbB/fisiologia , Vasoconstrição/efeitos dos fármacos , Animais , Aorta/patologia , Aorta/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Colágeno Tipo I/genética , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Fibrose , Flavonoides/farmacologia , Técnicas In Vitro , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Regiões Promotoras Genéticas/genética , Quinazolinas/farmacologia , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA