Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Physiol Renal Physiol ; 304(7): F938-47, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364805

RESUMEN

Renal artery stenosis (RAS) is an important cause of chronic renal dysfunction. Recent studies have underscored a critical role for CCL2 (MCP-1)-mediated inflammation in the progression of chronic renal damage in RAS and other chronic renal diseases. In vitro studies have implicated p38 MAPK as a critical intermediate for the production of CCL2. However, a potential role of p38 signaling in the development and progression of chronic renal disease in RAS has not been previously defined. We sought to test the hypothesis that inhibition of p38 MAPK ameliorates chronic renal injury in mice with RAS. We established a murine RAS model by placing a cuff on the right renal artery and treated mice with the p38 inhibitor SB203580 or vehicle for 2 wk. In mice treated with vehicle, the cuffed kidney developed interstitial fibrosis, tubular atrophy, and interstitial inflammation. In mice treated with SB203580, the RAS-induced renal atrophy was reduced (70% vs. 39%, P < 0.05). SB203580 also reduced interstitial inflammation and extracellular matrix deposition but had no effect on the development of hypertension. SB203580 partially blocked the induction of CCL2, CCL7 (MCP-3), CC chemokine receptor 2 (CCR2), and collagen 4 mRNA expression in the cuffed kidneys. In vitro, blockade of p38 hindered both TNF-α and TGF-ß-induced CCL2 upregulation. Based on these observations, we conclude that p38 MAPK plays a critical role in the induction of CCL2/CCL7/CCR2 system and the development of interstitial inflammation in RAS.


Asunto(s)
Quimiocina CCL2/biosíntesis , Riñón/metabolismo , Nefroesclerosis/patología , Obstrucción de la Arteria Renal/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Atrofia/patología , Quimiocina CCL7/biosíntesis , Modelos Animales de Enfermedad , Fibrosis , Imidazoles/farmacología , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Piridinas/farmacología , Receptores CCR2/biosíntesis , Obstrucción de la Arteria Renal/prevención & control , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
Am J Physiol Renal Physiol ; 302(11): F1455-64, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22378822

RESUMEN

Although the two-kidney, one-clip (2K1C) model is widely used as a model of human renovascular hypertension, mechanisms leading to the development of fibrosis and atrophy in the cuffed kidney and compensatory hyperplasia in the contralateral kidney have not been defined. Based on the well-established role of the transforming growth factor (TGF)-ß signaling pathway in renal fibrosis, we tested the hypothesis that abrogation of TGF-ß/Smad3 signaling would prevent fibrosis in the cuffed kidney. Renal artery stenosis (RAS) was established in mice with a targeted disruption of exon 2 of the Smad3 gene (Smad3 KO) and wild-type (WT) controls by placement of a polytetrafluoroethylene cuff on the right renal artery. Serial pulse-wave Doppler ultrasound assessments verified that blood flow through the cuffed renal artery was decreased to a similar extent in Smad3 KO and WT mice. Two weeks after surgery, systolic blood pressure and plasma renin activity were significantly elevated in both the Smad3 KO and WT mice. The cuffed kidney of WT mice developed renal atrophy (50% reduction in weight after 6 wk, P < 0.0001), which was associated with the development of interstitial fibrosis, tubular atrophy, and interstitial inflammation. Remarkably, despite a similar reduction of renal blood flow, the cuffed kidney of the Smad3 KO mice showed minimal atrophy (9% reduction in weight, P = not significant), with no significant histopathological alterations (interstitial fibrosis, tubular atrophy, and interstitial inflammation). We conclude that abrogation of TGF-ß/Smad3 signaling confers protection against the development of fibrosis and atrophy in RAS.


Asunto(s)
Hipertensión Renovascular/genética , Hipertensión Renovascular/patología , Riñón/patología , Proteína smad3/deficiencia , Proteína smad3/genética , Animales , Atrofia , Colágeno/biosíntesis , Constricción Patológica , Fibrosis , Inmunohistoquímica , Pruebas de Función Renal , Ratones , Mutación/genética , Mutación/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Obstrucción de la Arteria Renal/patología , Circulación Renal/genética , Circulación Renal/fisiología , Renina/sangre , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/biosíntesis
3.
Am J Physiol Renal Physiol ; 300(5): F1142-51, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21367920

RESUMEN

Monocyte chemoattractant protein 1 (MCP-1) is a CC cytokine that fundamentally contributes to the pathogenesis of inflammatory renal disease. MCP-1 is highly expressed in cytokine-stimulated mesangial cells in vitro and following glomerular injury in vivo. Interventions to limit MCP-1 expression are commonly effective in assorted experimental models. Fish oil, an abundant source of n-3 fatty acids, has anti-inflammatory properties, the basis of which remains incompletely defined. We examined potential mechanisms whereby fish oil reduces MCP-1 expression and thereby suppresses inflammatory responses to tissue injury. Cultured mesangial cells were treated with TNF-α in the presence of the n-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA); equimolar concentrations of the n-6 fatty acids LA and OA served as controls. MCP-1 mRNA expression was assessed by Northern blotting, and transcriptional activity of the MCP-1 promoter was assessed by transient transfection. The involvement of the ERK and NF-κB pathways was evaluated through transfection analysis and the use of the MEK inhibitor U0126. DHA and EPA decreased TNF-α-stimulated MCP-1 mRNA expression by decreasing transcription of the MCP-1 gene. DHA and EPA decreased p-ERK expression and nuclear translocation of NF-κB, both of which are necessary for TNF-α-stimulated MCP-1 expression. Both NF-κB and AP-1 sites were involved in transcriptional regulation of the MCP-1 gene by DHA and EPA. We conclude that DHA and EPA inhibit TNF-α-stimulated transcription of the MCP-1 gene through interaction of signaling pathways involving ERK and NF-κB. We speculate that such effects may contribute to the salutary effect of fish oil in renal and vascular disease.


Asunto(s)
Antiinflamatorios/farmacología , Quimiocina CCL2/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Células Mesangiales/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Quimiocina CCL2/genética , Regulación hacia Abajo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genes Reporteros , Humanos , Masculino , Células Mesangiales/inmunología , Células Mesangiales/metabolismo , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional/efectos de los fármacos , Transfección
4.
Am J Physiol Renal Physiol ; 297(4): F1055-68, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19625373

RESUMEN

Unilateral renal artery stenosis (RAS) leads to atrophy of the stenotic kidney and compensatory enlargement of the contralateral kidney. Although the two-kidney, one-clip (2K1C) model has been extensively used to model human RAS, the cellular responses in the stenotic and contralateral kidneys, particularly in the murine model, have received relatively little attention. We studied mice 2, 5, and 11 wk after unilateral RAS. These mice became hypertensive within 1 wk. The contralateral kidney increased in size within 2 wk after surgery. This enlargement was associated with a transient increase in expression of phospho-extracellular signal-regulated kinase (p-ERK), the proliferation markers proliferating cell nuclear antigen and Ki-67, the cell cycle inhibitors p21 and p27, and transforming growth factor-beta, with return to baseline levels by 11 wk. The size of the stenotic kidney was unchanged at 2 wk but progressively decreased between 5 and 11 wk. Unlike the contralateral kidney, which showed minimal histopathological alterations, the stenotic kidney developed progressive interstitial fibrosis, tubular atrophy, and interstitial inflammation. Surprisingly, the stenotic kidney showed a proliferative response, which involved largely tubular epithelial cells. The atrophic kidney had little evidence of apoptosis, despite persistent upregulation of p53; expression of cell cycle regulatory proteins in the stenotic kidney was persistently increased through 11 wk. These studies indicate that in the 2K1C model, the stenotic kidney and contralateral, enlarged kidney exhibit a distinct temporal expression of proteins involved in cell growth, cell survival, apoptosis, inflammation, and fibrosis. Notably, an unexpected proliferative response occurs in the stenotic kidney that undergoes atrophy.


Asunto(s)
Hipertensión Renovascular/metabolismo , Transducción de Señal , Actinas/metabolismo , Animales , Apoptosis , Atrofia , Proliferación Celular , Quimiocina CCL2/metabolismo , Colágeno/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Hiperplasia , Hipertensión Renovascular/genética , Hipertensión Renovascular/patología , Hipertrofia , Interfase , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
5.
Exp Biol Med (Maywood) ; 231(3): 288-95, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16514175

RESUMEN

Polycystic kidney diseases (PKD) are characterized by excessive proliferation of renal tubular epithelial cells, development of fluid-filled cysts, and progressive renal insufficiency. cAMP inhibits proliferation of normal renal tubular epithelial cells but stimulates proliferation of renal tubular epithelial cells derived from patients with PKD. Madin-Darby canine kidney (MDCK) epithelial cells, which are widely used as an in vitro model of cystogenesis, also proliferate in response to cAMP. Intracellular cAMP levels are tightly regulated by phosphodiesterases (PDE). Isoform-specific PDE inhibitors have been developed as therapeutic agents to regulate signaling pathways directed by cAMP. In other renal cell types, we have previously demonstrated that cAMP is hydrolyzed by PDE3 and PDE4, but only PDE3 inhibitors suppress proliferation by inhibiting Raf-1 activity (Cheng J, Thompson MA, Walker HJ, Gray CE, Diaz Encarnacion MM, Warner GM, Grande JP. Am J Physiol Renal Physiol 287:F940-F953, 2004.) A potential role for PDE isoform(s) in cAMP-mediated proliferation of MDCK cells has not previously been established. Similar to what we have previously found in several other renal cell types, cAMP hydrolysis in MDCK cells is directed primarily by PDE4 (85% of total activity) and PDE3 (15% of total activity). PDE4 inhibitors are more effective than PDE3 inhibitors in increasing intracellular cAMP levels in MDCK cells. However, only PDE3 inhibitors, and not PDE4 inhibitors, stimulate mitogenesis of MDCK cells. PDE3 but not PDE4 inhibitors activate B-Raf but not Raf-1, as assessed by an in vitro kinase assay. PDE3 but not PDE4 inhibitors activate the ERK pathway and activate cyclins D and E, as assessed by histone H1 kinase assay. We conclude that mitogenesis of MDCK cells is regulated by a functionally compartmentalized intracellular cAMP pool directed by PDE3. Pharmacologic agents that stimulate PDE3 activity may provide the basis for new therapies directed toward reducing cystogenesis in patients with PKD.


Asunto(s)
Riñón/citología , Riñón/efectos de los fármacos , Mitosis/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Quinazolinas/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-AMP Cíclico Fosfodiesterasas/fisiología , Animales , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Línea Celular , Colforsina/farmacología , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Ciclina D , Ciclina E/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/fisiología , Perros , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Riñón/metabolismo , Mitosis/fisiología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Rolipram/farmacología
6.
Exp Biol Med (Maywood) ; 227(3): 171-81, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11856815

RESUMEN

Recent studies in cultured cells have provided evidence that a variety of pathobiologic stimuli, including high glucose, angiotensin II, and thromboxane A(2), trigger a signaling pathway leading to autocrine induction of TGF-beta1. TGF-beta1 production through this pathway may profoundly affect cell growth, matrix synthesis, and response to injury. This study examines the role of autocrine versus exogenously added TGF-beta1 in cellular proliferation and collagen IV production, critical targets of TGF-beta1 signaling, using renal cells derived from TGF-beta1 knockout (KO) animals or wild-type (WT) controls. Growth of WT and KO cells was assessed by cell counting and [(3)H]thymidine uptake. Basal and TGF-beta1-stimulated collagen production was assessed by Northern and Western blotting; transcriptional activity of the alpha1(IV) collagen gene was assessed by transient transfection analysis. KO cells grew at a faster rate than WT cells carefully matched for plating density and passage number. This increased growth rate was paralleled by increases in [(3)H]thymidine uptake. KO cells expressed lower levels of the cell cycle inhibitors p21 and p27 than WT cells. KO cells failed to express TGF-beta1, as expected. Basal TGF-beta3 mRNA levels were higher in KO cells than in WT cells. WT cells expressed higher basal levels of TGF-beta2 mRNA than KO cells. Basal alpha1(IV) and alpha2(IV) collagen mRNA and protein expression were significantly lower in KO cells than WT cells. Administration of exogenous TGF-beta1 induced collagen IV production in both KO and WT cells. Although basal transcriptional activity of an alpha1(IV) collagen-CAT construct was lower in KO cells than WT cells, administration of exogenous TGF-beta1 was associated with significant increases in transcriptional activity of this construct in both KO and WT cells. These studies provide evidence that autocrine production of TGF-beta1 may play a critical role in regulation of growth and basal collagen IV production by renal tubular epithelial cells.


Asunto(s)
Comunicación Autocrina/fisiología , Colágeno Tipo IV/biosíntesis , Túbulos Renales/citología , Túbulos Renales/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , División Celular/fisiología , Células Cultivadas , Colágeno Tipo IV/genética , Ratones , Ratones Noqueados , Activación Transcripcional , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1
7.
Am J Physiol Renal Physiol ; 294(6): F1323-35, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18385269

RESUMEN

Although many studies have indicated that fish oil (FO) improves cardiovascular risk factors and reduces histopathological manifestations of injury in experimental renal injury models, potential mechanisms underlying this protective effect have not been adequately defined. The objective of this study was to identify potential signaling pathways that confer protection in the Dahl rat model of salt-sensitive hypertension. Male Dahl salt-sensitive rats (n = 10/group) were provided with formulated diets containing 8% NaCl, 20% protein, and 25% FO or 25% corn oil (CO) for 28 days. FO reduced blood pressure (-11% at 4 wk; P < 0.05), urine protein excretion (-45% at 4 wk; P < 0.05), plasma cholesterol and triglyceride levels (-54%, P < 0.001; and -58%, P < 0.05), and histopathological manifestations of renal injury, including vascular hypertrophy, segmental and global glomerular sclerosis, interstitial fibrosis, and tubular atrophy. Interstitial inflammation was significantly reduced by FO (-32%; P < 0.001), as assessed by quantitative analysis of ED1-positive cells in sections of the renal cortex. FO reduced tubulointerstitial proliferative activity, as assessed by Western blot analysis of cortical homogenates for PCNA (-51%; P < 0.01) and quantitative analysis of Mib-1-stained sections of the renal cortex (-42%; P < 0.001). Decreased proliferative activity was associated with reduced phospho-ERK expression (-37%; P < 0.005) and NF-kappaB activation (-42%; P < 0.05). FO reduced cyclooxygenase (COX)-2 expression (-63%; P < 0.01) and membrane translocation of the NADPH oxidase subunits p47(phox) and p67(phox) (-26 and -34%; P < 0.05). We propose that FO ameliorates renal injury in Dahl salt-sensitive rats through the inhibition of ERK, decreased NF-kappaB activation, inhibition of COX-2 expression, and decreased NADPH oxidase activation.


Asunto(s)
Aceites de Pescado/farmacología , Hipertensión Renal/tratamiento farmacológico , Hipertensión Renal/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , División Celular/efectos de los fármacos , Aceite de Maíz/farmacología , Grasas de la Dieta/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Hipertensión Renal/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Miocardio/patología , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Tamaño de los Órganos , Fosfoproteínas/metabolismo , Proteinuria/tratamiento farmacológico , Proteinuria/metabolismo , Proteinuria/patología , Ratas , Ratas Endogámicas Dahl , Transducción de Señal/fisiología
8.
Nephrol Dial Transplant ; 22(4): 1136-43, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17255128

RESUMEN

BACKGROUND: Preeclampsia is a pregnancy-specific disorder characterized by hypertension and proteinuria. In other disease states, proteinuria has been linked to altered expressions of podocyte foot-process proteins, but this has not been studied in women with preeclampsia. We sought to test the hypothesis that proteinuria in preeclampsia is associated with dysregulated expression of the podocyte cytoskeleton and/or tight junction proteins. METHODS: Renal tissue was obtained from autopsy material from seven women who had severe preeclampsia during the second half of their pregnancies up to 48 h after delivery, and who subsequently died. As controls, we used autopsy material from two women who died accidentally during the second half of their otherwise normal pregnancies. Immunohistochemical stains for nephrin, synaptopodin and podocin were performed on representative sections prepared from paraffin-embedded material. RESULTS: Expression of both nephrin and synaptopodin was markedly decreased in preeclamptic compared with control kidney sections. By contrast, both cases and controls demonstrated strong staining for podocin. CONCLUSIONS: We conclude that down-regulation of nephrin and synaptopodin is associated with proteinuria in women with preeclampsia. Recent studies have demonstrated that soluble vascular endothelial growth factor receptor 1 (sFlt-1) levels are elevated in preeclampsia compared with normal pregnancy. Studies in mice have shown that sFlt-1 may play a role in inducing proteinuria by neutralizing vascular endothelial growth factor (VEGF) and suppressing nephrin. Proteinuria and elevations of sFlt-1 in preeclampsia are temporally related, further supporting a possible role of sFlt-1 in the dysregulation of podocyte foot-process proteins.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glomérulos Renales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Preeclampsia/metabolismo , Adolescente , Adulto , Animales , Anticuerpos/efectos adversos , Biopsia , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Glomérulos Renales/patología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos , Proteínas de Microfilamentos/genética , Preeclampsia/genética , Embarazo , Proteinuria/inducido químicamente , Proteinuria/metabolismo , Factor A de Crecimiento Endotelial Vascular/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular
9.
Am J Physiol Cell Physiol ; 289(4): C959-70, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15930146

RESUMEN

Monocyte chemoattractant protein-1 (MCP-1) and transforming growth factor (TGF)-beta1 are critical mediators of renal injury by promoting excessive inflammation and extracellular matrix deposition, thereby contributing to progressive renal disease. In renal disease models, MCP-1 stimulates the production of TGF-beta1. However, a potential role for TGF-beta1 in the regulation of MCP-1 production by mesangial cells (MCs) has not previously been evaluated. The objectives of this study were to define the role of TGF-beta1 in regulation of MCP-1 expression in cultured MCs and to define mechanisms through which rolipram (Rp), a phosphodiesterase isoenzyme 4 (PDE4) inhibitor with anti-inflammatory properties, alters MCP-1 expression. TGF-beta1 induced MCP-1 in a time- and dose-dependent manner without increasing transcription of the MCP-1 gene. TGF-beta1-mediated induction of MCP-1 occurred without activation of the NF-kappaB pathway. Rp blocked TGF-beta1-stimulated MCP-1 expression via a protein kinase A-dependent process, at least in part, by decreasing MCP-1 message stability. Rp exerted no effect on activation of the Smad pathway by TGF-beta1. TGF-beta1-mediated induction of MCP-1 required activation of ERK and p38, both of which were suppressed by a PDE4 inhibitor. TGF-beta1-stimulated reactive oxygen species (ROS) generation by MCs, and Rp inhibited ROS generation in TGF-beta1-stimulated MCs; in addition, both Rp and ROS scavengers blocked TGF-beta1-stimulated MCP-1 expression. We conclude that TGF-beta1 stimulates MCP-1 expression through pathways involving activation of ERK, p38, and ROS generation. Positive cross-talk between TGF-beta1 and MCP-1 signaling in MCs may underlie the development of progressive renal disease. Rp, by preventing TGF-beta1-stimulated MCP-1 production, may offer a therapeutic approach in retarding the progression of renal disease.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Quimiocina CCL2/biosíntesis , Mesangio Glomerular/metabolismo , Factor de Crecimiento Transformador beta/fisiología , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Animales , Línea Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Mesangio Glomerular/citología , FN-kappa B/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Am J Physiol Renal Physiol ; 287(5): F940-53, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15280158

RESUMEN

Mesangial cell (MC) mitogenesis is regulated through "negative cross talk" between cAMP-PKA and ERK signaling. Although it is widely accepted that cAMP inhibits mitogenesis through PKA-mediated phosphorylation of Raf-1, recent studies have indicated that cAMP-mediated inhibition of mitogenesis may occur independently of Raf-1 phosphorylation or without inhibiting ERK activity. We previously showed that MCs possess functionally compartmentalized intracellular pools of cAMP that are differentially regulated by cAMP phosphodiesterases (PDE); an intracellular pool directed by PDE3 but not by PDE4 suppresses mitogenesis. We therefore sought to determine whether there was a differential effect of PDE3 vs. PDE4 inhibitors on the Ras-Raf-MEK-ERK pathway in cultured MC. Although PDE3 and PDE4 inhibitors activated PKA and modestly elevated cAMP levels to a similar extent, only PDE3 inhibitors suppressed MC mitogenesis (-57%) and suppressed Raf-1 kinase and ERK activity (-33 and -68%, respectively). Both PDE3 and PDE4 inhibitors suppressed B-Raf kinase activity. PDE3 inhibitors increased phosphorylation of Raf-1 on serine 43 and serine 259 and decreased phosphorylation on serine 338; PDE4 inhibitors were without effect. Overexpression of a constitutively active MEK-1 construct reversed the antiproliferative effect of PDE3 inhibitors. PDE3 inhibitors also reduced cyclin A levels (-27%), cyclin D and cyclin E kinase activity (-30 and -50%, respectively), and induced expression of the cell cycle inhibitor p21 (+90%). We conclude that the antiproliferative effects of PDE3 inhibitors are mechanistically related to inhibition of the Ras-Raf-MEK-ERK pathway. Additional cell cycle targets of PDE3 inhibitors include cyclin A, cyclin D, cyclin E, and p21.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/fisiología , Mesangio Glomerular/citología , Mesangio Glomerular/fisiología , Mitosis/fisiología , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3 , Caspasas/metabolismo , División Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ciclina E/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Masculino , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
J Lab Clin Med ; 141(5): 318-29, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12761475

RESUMEN

Although dietary fish oil supplementation has been used to prevent the progression of kidney disease in patients with IgA nephropathy, relatively few studies provide a mechanistic rationale for its use. Using an antithymocyte (ATS) model of mesangial proliferative glomerulonephritis, we recently demonstrated that fish oil inhibits mesangial cell (MC) activation and proliferation, reduces proteinuria, and decreases histologic evidence of glomerular damage. We therefore sought to define potential mechanisms underlying the antiproliferative effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), the predominant omega-3 polyunsaturated fatty acids found in fish oil, in cultured MC. DHA and EPA were administered to MC as bovine serum albumin fatty-acid complexes. Low-dose (10-50 micromol/L) DHA, but not EPA, inhibited basal and epidermal growth factor (EGF)-stimulated [(3)H]-thymidine incorporation in MCs. At higher doses (100 micromol/L), EPA and DHA were equally effective in suppressing basal and EGF-stimulated MC mitogenesis. Low-dose DHA, but not EPA, decreased ERK activation by 30% (P <.01), as assessed with Western-blot analysis using phosphospecific antibodies. JNK activity was increased by low-dose DHA but not by EPA. p38 activity was not significantly altered by DHA or EPA. Cyclin E activity, as assessed with a histone H1 kinase assay, was inhibited by low-dose DHA but not by EPA. DHA increased expression of the cell cycle inhibitor p21 but not p27; EPA had no effect on p21 or p27. We propose that the differential effect of low-dose DHA vs EPA in suppressing MC mitogenesis is related to down-regulation of ERK and cyclin E activity and to induction of p21.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Aceites de Pescado/farmacología , Mesangio Glomerular/efectos de los fármacos , Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Mesangio Glomerular/citología , Mesangio Glomerular/enzimología , Mesangio Glomerular/metabolismo , Glomerulonefritis por IGA/patología , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA