Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Biochem J ; 435(3): 689-700, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21309752

RESUMEN

Caenorhabditis elegans harbours several CYP (cytochrome P450) genes that are homologous with mammalian CYP isoforms important to the production of physiologically active AA (arachidonic acid) metabolites. We tested the hypothesis that mammals and C. elegans may share similar basic mechanisms of CYP-dependent eicosanoid formation and action. We focused on CYP33E2, an isoform related to the human AA-epoxygenases CYP2C8 and CYP2J2. Co-expression of CYP33E2 with the human NADPH-CYP reductase in insect cells resulted in the reconstitution of an active microsomal mono-oxygenase system that metabolized EPA (eicosapentaenoic acid) and, with lower activity, also AA to specific sets of regioisomeric epoxy- and hydroxy-derivatives. The main products included 17,18-epoxyeicosatetraenoic acid from EPA and 19-hydroxyeicosatetraenoic acid from AA. Using nematode worms carrying a pCYP33E2::GFP reporter construct, we found that CYP33E2 is exclusively expressed in the pharynx, where it is predominantly localized in the marginal cells. RNAi (RNA interference)-mediated CYP33E2 expression silencing as well as treatments with inhibitors of mammalian AA-metabolizing CYP enzymes, significantly reduced the pharyngeal pumping frequency of adult C. elegans. These results demonstrate that EPA and AA are efficient CYP33E2 substrates and suggest that CYP-eicosanoids, influencing in mammals the contractility of cardiomyocytes and vascular smooth muscle cells, may function in C. elegans as regulators of the pharyngeal pumping activity.


Asunto(s)
Caenorhabditis elegans/enzimología , Sistema Enzimático del Citocromo P-450/clasificación , Sistema Enzimático del Citocromo P-450/metabolismo , Eicosanoides/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Clonación Molecular , Inhibidores Enzimáticos del Citocromo P-450 , Eicosanoides/genética , Silenciador del Gen , Mutación , Faringe/enzimología , Isoformas de Proteínas
2.
J Cell Physiol ; 214(2): 363-70, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17654516

RESUMEN

Transforming growth factor-beta (TGF-beta), a potent inhibitor of normal melanocyte growth, does not significantly suppress growth of melanoma cells. The mechanism of melanocyte desensitization to TGF-beta in the transformation process remains largerly unknown. We investigated whether the tumor promoting phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) may induce melanocyte resistance to TGF-beta. Cell proliferation and DNA synthesis of normal human melanocytes were strongly inhibited by TGF-beta, whereas in the presence of TPA remained largerly unaffected. The inactive phorbol ester 4alpha-phorbol 12,13 didecanoate did not modify the TGF-beta antiproliferative effect, whereas the diacylglycerol analog 1-oleoyl-2-acetyl-sn-glycerol counteracted TGF-beta effects. Protein kinase C (PKC) is the major cellular receptor of tumor promoting phorbol esters. PKC-alpha expression and phosphorylation were almost completely downregulated under combined treatment with TGF-beta + TPA at 24 and 72 h, as shown by immunoblots. Confocal microscopy demonstrated that TGF-beta-induced nuclear accumulation of PKC-alpha was abolished in the presence of TPA at the same time points. The selective PKC inhibitor Ro-31-8220 weakened the TGF-beta antiproliferative effect. Smads are central mediators for TGF-beta signal transduction. Smad-dependent transcriptional activity was suppressed in TGF-beta-treated melanocytes in the presence of TPA, as well as in ALK5 (constitutively active type I TGF-beta receptor)- or Smad3 + Smad4-transfected melanocytes in the presence of Ro-31-8220. In addition, an antisense oligodeoxynucleotide against PKC-alpha abolished TGF-beta-driven Smad-mediated transcription. These findings show that tumor promoting phorbol esters induce melanocyte resistance to TGF-beta, associated with downregulation of PKC-alpha and suppression of Smad-dependent transcription. This may represent an important mechanism for expansion of melanocytes exposed to PKC-targeting tumor promoters.


Asunto(s)
Carcinógenos/farmacología , Melanocitos/citología , Melanocitos/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Reguladoras de la Apoptosis , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Inhibidores Enzimáticos/farmacología , Genes Reporteros , Humanos , Indoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Cinética , Luciferasas/metabolismo , Masculino , Melanocitos/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Piel/citología , Transcripción Genética/efectos de los fármacos , Transfección , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
3.
Diabetes ; 56(2): 346-54, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17259378

RESUMEN

The protein kinase C (PKC)-beta isoform has been implicated to play a pivotal role in the development of diabetic kidney disease. We tested this hypothesis by inducing diabetic nephropathy in PKC-beta-deficient (PKC-beta(-/-)) mice. We studied nondiabetic and streptozotocin-induced diabetic PKC-beta(-/-) mice compared with appropriate 129/SV wild-type mice. After 8 weeks of diabetes, the high-glucose-induced renal and glomerular hypertrophy, as well as the increased expression of extracellular matrix proteins such as collagen and fibronectin, was reduced in PKC-beta(-/-) mice. Furthermore, the high-glucose-induced expression of the profibrotic cytokine transforming growth factor (TGF)-beta1 and connective tissue growth factor were significantly diminished in the diabetic PKC-beta(-/-) mice compared with diabetic wild-type mice, suggesting a role of the PKC-beta isoform in the regulation of renal hypertrophy. Notably, increased urinary albumin-to-creatinine ratio persisted in the diabetic PKC-beta(-/-) mice. The loss of the basement membrane proteoglycan perlecan and the podocyte protein nephrin in the diabetic state was not prevented in the PKC-beta(-/-) mice as previously demonstrated in the nonalbuminuric diabetic PKC-alpha(-/-) mice. In summary, the differential effects of PKC-beta deficiency on diabetes-induced renal hypertrophy and albuminuria suggest that PKC-beta contributes to high-glucose-induced TGF-beta1 expression and renal fibrosis, whereas perlecan, as well as nephrin, expression and albuminuria is regulated by other signaling pathways.


Asunto(s)
Albuminuria/genética , Nefropatías Diabéticas/enzimología , Riñón/patología , Proteína Quinasa C/deficiencia , Albuminuria/prevención & control , Animales , Deleción Cromosómica , Colágeno Tipo IV/metabolismo , Creatinina/metabolismo , Diabetes Mellitus Experimental , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Fibronectinas/metabolismo , Fibrosis , Proteoglicanos de Heparán Sulfato/metabolismo , Hipertrofia , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Tamaño de los Órganos , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Proteína Quinasa C/genética , ARN/metabolismo , Estreptozocina , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
4.
N Engl J Med ; 353(15): 1585-90, 2005 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-16221781

RESUMEN

We report on a patient with long-standing severe autonomic failure that affected his sympathetic and parasympathetic nervous systems. Antibodies against the ganglionic acetylcholine receptors were detected in the serum. Removal of the antibodies by means of plasma exchange resulted in a dramatic clinical improvement.


Asunto(s)
Autoanticuerpos/sangre , Enfermedades Autoinmunes/terapia , Enfermedades del Sistema Nervioso Autónomo/terapia , Intercambio Plasmático , Adulto , Enfermedades del Sistema Nervioso Autónomo/inmunología , Enfermedades del Sistema Nervioso Autónomo/fisiopatología , Terapia Combinada , Ganglios Autónomos/inmunología , Glucocorticoides/uso terapéutico , Humanos , Terapia de Inmunosupresión , Masculino , Prednisolona/uso terapéutico , Receptores Colinérgicos/inmunología , Recurrencia , Síncope/etiología , Síncope/terapia
5.
FASEB J ; 20(7): 994-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16597674

RESUMEN

Long-acting third-generation dihydropyridine calcium channel blockers (CCBs) improve endothelial dysfunction and prevent cardiovascular events in humans, but their cellular and molecular mechanisms of tissue protection are not elucidated in detail. We assessed organ (renal) protection by the highly lipophilic CCB lercanidipine in a double-transgenic rat (dTGR) model with overexpression of human renin and angiotensinogen genes. We randomly treated dTGR with lercanidipine (2.5 mg/kg/day; n=20) or vehicle (n=20) for 3 wk. Furthermore, we explored the influence of lercanidipine on protein kinase C (PKC) signaling in vivo and in vitro using endothelial and vascular smooth muscle cell cultures. Cumulative mortality was 60% in untreated dTGR, whereas none of the lercanidipine-treated animals died (P<0.001). We found significantly less albuminuria and improved renal function in lercanidipine-treated dTGR (both P<0.05). Lercanidipine treatment also significantly (P<0.05) reduced blood levels of the endogenous NOS inhibitor asymmetric dimethylarginine. On histological examination, we observed significantly less tissue inflammation and fibrosis in lercanidipine-treated animals (both P<0.05). Lercanidipine significantly inhibited angiotensin (ANG) I-mediated PKC-alpha and -delta activation in vivo and in vitro, partly due to reduced intracellular calcium flux. As a result, lercanidipine improved endothelial cell permeability in vitro. Lercanidipine prevents tissue injury and improves survival in a model of progressive organ damage. These effects may result, at least in part, from inhibition of tissue inflammation as well as improved NO bioavailability. Modulation of PKC activity may be an important underlying intracellular mechanism.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Dihidropiridinas/farmacología , Hipertensión/metabolismo , Albúminas/metabolismo , Amidohidrolasas/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Animales Modificados Genéticamente , Arginina/análogos & derivados , Arginina/sangre , Arginina/metabolismo , Regulación Enzimológica de la Expresión Génica , Hipertensión/tratamiento farmacológico , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratas , Ratas Sprague-Dawley , Renina/genética , Renina/metabolismo
6.
Cardiovasc Res ; 67(1): 50-9, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15949469

RESUMEN

OBJECTIVE: Angiotensin II (AII) promotes cardiac fibrosis by increased extracellular matrix production and enhanced interaction of matrix proteins with their cellular receptors, including integrins. AII and other growth factors augment beta(1)-integrin-dependent adhesion and spreading by "inside-out signaling" without affecting the total number of integrin receptors. "Inside-out signaling" involves specific signaling pathways, including protein kinase C (PKC), leading to activation of beta1-integrins. In the present study we investigated the mechanisms involved in AII-increased adhesion of adult rat cardiac fibroblasts (CFBs), obtained from Sprague-Dawley rats, to collagen I mediated by beta1-integrin. METHODS AND RESULTS: Treatment of CFBs with AII induced a concentration-dependent increase in adhesion to collagen I (2.2-fold, p<0.01) within 3-6 h of treatment. This effect was mediated by beta1-integrin via the angiotensin AT1 receptor and was significantly reduced by protein kinase C inhibition. AII significantly induced phosphorylation of PKC epsilon (PKCepsilon), which is involved in beta1-integrin-dependent adhesion and motility, and phosphorylation of the cytoplasmatic tail of beta1-integrin at threonine 788/789, required for adhesion. Phosphorylation of beta1-integrin and an increase in adhesion was also induced by l-alpha-phosphatidylinositol-3,4,5-triphosphate (l-alpha-PIP3), an activator of endogenous PKCepsilon. AII failed to increase adhesion in myofibroblasts obtained from PKCepsilon (-/-) mice, but not in cells obtained from control mice. Co-immunoprecipitation and double immunofluorescence demonstrated that AII induced a close association of PKCepsilon with beta1-integrin in CFBs. CONCLUSION: The present study demonstrates that AII increased beta1-integrin-dependent adhesion to collagen I in cardiac fibroblasts by inside-out signaling via PKCepsilon and phosphorylation of the cytoplasmatic tail of the beta1-integrin.


Asunto(s)
Angiotensina II/metabolismo , Insuficiencia Cardíaca/metabolismo , Integrina beta1/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Western Blotting/métodos , Adhesión Celular , Uniones Célula-Matriz/metabolismo , Colágeno/metabolismo , Activación Enzimática , Técnica del Anticuerpo Fluorescente , Insuficiencia Cardíaca/patología , Ratones , Ratones Noqueados , Modelos Animales , Miocitos Cardíacos/patología , Fosforilación , Ratas , Ratas Sprague-Dawley , Remodelación Ventricular
7.
Circulation ; 109(22): 2792-800, 2004 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-15159288

RESUMEN

BACKGROUND: In a double-transgenic human renin and human angiotensinogen rat model, we found that mineralocorticoid receptor (MR) blockade ameliorated angiotensin II (Ang II)-induced renal and cardiac damage. How Ang II and aldosterone (Ald) might interact is ill defined. METHODS AND RESULTS: We investigated the effects of Ang II (10(-7) mol/L) and Ald (10(-7) mol/L) on extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling in vascular smooth muscle cells (VSMCs) with Western blotting and confocal microscopy. Ang II induced ERK 1/2 and JNK phosphorylation by 2 minutes. Ald achieved the same at 10 minutes. Ang II+Ald had a potentiating effect by 2 minutes. Two oxygen radical scavengers and the epidermal growth factor receptor (EGFR) antagonist AG1478 reduced Ang II-, Ald-, and combination-induced ERK1/2 phosphorylation. Preincubating the cells with the MR blocker spironolactone (10(-6) mol/L) abolished Ang II-induced ROS generation, EGFR transactivation, and ERK1/2 phosphorylation. CONCLUSIONS: Ald potentiates Ang II-induced ERK-1/2 and JNK phosphorylation. Oxygen radicals, the MR, and the EGFR play a role in early signaling induced by Ang II and Ald in VSMCs. These in vitro data may help explain the effects of MR blockade on Ang II-induced end-organ damage in vivo.


Asunto(s)
Aldosterona/fisiología , Angiotensina II/fisiología , Músculo Liso Vascular/fisiología , Angiotensina II/antagonistas & inhibidores , Animales , Animales Modificados Genéticamente , Células Cultivadas , Receptores ErbB/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Espironolactona/farmacología
8.
Circulation ; 110(8): 1006-12, 2004 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-15302785

RESUMEN

BACKGROUND: The hematopoietic cytokine erythropoietin has cytoprotective effects in endothelial cells in vitro that are mediated through direct activation of the pro-survival Akt tyrosine kinase signaling pathway. We tested the hypothesis that low-dose therapy with the long-acting recombinant human erythropoietin analogue darbepoetin alpha protects vascular endothelium in vivo in a classic remnant kidney rat model characterized by severe endothelial damage, progressive vascular sclerosis, and ischemia-induced tissue fibrosis. METHODS AND RESULTS: Using a parallel group study design, we randomly assigned animals after 5/6 nephrectomy to treatment with either saline (n=36) or 0.1 microg/kg body wt darbepoetin (n=24) subcutaneously once weekly. We monitored hematocrit, blood pressure, and serum creatinine regularly and obtained renal tissue 6 weeks after nephrectomy for morphological and immunohistochemical analysis. Darbepoetin-treated animals had significantly improved survival compared with saline-treated controls (63% versus 33%; P<0.05), although hematocrit levels were similar in both groups. Darbepoetin treatment ameliorated endothelial damage; attenuated the composite tissue injury score (saline 1.9+/-0.4; darbepoetin 0.4+/-0.2; P<0.001), which included vascular sclerosis, glomerulosclerosis, and tubulointerstitial damage; and preserved renal function. We found persistent activation of the pro-survival Akt signaling pathway in endothelial and epithelial glomerular cells in darbepoetin-treated animals, accompanied by a significant reduction of apoptotic cell death in renal tissue. CONCLUSIONS: Low-dose darbepoetin treatment confers vascular and tissue protection that is associated with persistent stimulation of the pro-survival Akt signaling pathway. The use of recombinant human erythropoietin or analogues may have utility in preventing ischemia-related progressive vascular injury and organ failure.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Insuficiencia Multiorgánica/prevención & control , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Darbepoetina alfa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/patología , Hematócrito , Movilización de Célula Madre Hematopoyética , Hipertensión Renal/etiología , Hipertensión Renal/fisiopatología , Isquemia/prevención & control , Riñón/irrigación sanguínea , Tablas de Vida , Masculino , Insuficiencia Multiorgánica/etiología , Nefrectomía , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , Proteínas Proto-Oncogénicas c-akt , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
9.
Diabetes ; 53(11): 2921-30, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15504973

RESUMEN

This study investigated the role of advanced glycation end products (AGEs) in mediating protein kinase C (PKC) isoform expression in diabetic nephropathy. In vitro, vascular smooth muscle cells incubated in a high-glucose (25-mmol/l) medium demonstrated translocation and increased expression of PKC-alpha as compared with those from a low-glucose (5-mmol/l) environment. Coincubation with the cross-link breaker ALT-711 and, to a lesser extent, with aminoguanidine, an inhibitor of AGE formation, attenuated the increased expression and translocation of PKC-alpha. Streptozotocin-induced diabetic rats were randomized to no treatment, treatment with ALT-711, or treatment with aminoguanidine. Diabetes induced increases in PKC-alpha as well as in the -betaI, -betaII, and -epsilon isoforms. Treatment with ALT-711 and aminoguanidine, which both attenuate renal AGE accumulation, abrogated these increases in PKC expression. However, translocation of phosphorylated PKC-alpha from the cytoplasm to the membrane was reduced only by ALT-711. ALT-711 treatment attenuated expression of vascular endothelial growth factor and the extracellular matrix proteins, fibronectin and laminin, in association with reduced albuminuria. Aminoguanidine had no effect on VEGF expression, although some reduction of fibronectin and laminin was observed. These findings implicate AGEs as important stimuli for the activation of PKC, particularly PKC-alpha, in the diabetic kidney, which can be directly inhibited by ALT-711.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/fisiopatología , Proteínas de la Matriz Extracelular/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Tiazoles/farmacología , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/patología , Guanidinas/farmacología , Corteza Renal/efectos de los fármacos , Corteza Renal/patología , Masculino , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteína Quinasa C-alfa , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
10.
Diabetes ; 53(8): 2101-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15277392

RESUMEN

Activation of protein kinase C (PKC) isoforms has been implicated in the pathogenesis of diabetic nephropathy. We showed earlier that PKC-alpha is activated in the kidneys of hyperglycemic animals. We now used PKC-alpha(-/-) mice to test the hypothesis that this PKC isoform mediates streptozotocin-induced diabetic nephropathy. We observed that renal and glomerular hypertrophy was similar in diabetic wild-type and PKC-alpha(-/-) mice. However, the development of albuminuria was almost absent in the diabetic PKC-alpha(-/-) mice. The hyperglycemia-induced downregulation of the negatively charged basement membrane heparan sulfate proteoglycan perlecan was completely prevented in the PKC-alpha(-/-) mice, compared with controls. We then asked whether transforming growth factor-beta1 (TGF-beta1) and/or vascular endothelial growth factor (VEGF) is implicated in the PKC-alpha-mediated changes in the basement membrane. The hyperglycemia-induced expression of VEGF165 and its receptor VEGF receptor II (flk-1) was ameliorated in PKC-alpha(-/-) mice, whereas expression of TGF-beta1 was not affected by the lack of PKC-alpha. Our findings indicate that two important features of diabetic nephropathy-glomerular hypertrophy and albuminuria-are differentially regulated. The glucose-induced albuminuria seems to be mediated by PKC-alpha via downregulation of proteoglycans in the basement membrane and regulation of VEGF expression. Therefore, PKC-alpha is a possible therapeutic target for the prevention of diabetic albuminuria.


Asunto(s)
Nefropatías Diabéticas/genética , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Proteoglicanos/metabolismo , Albuminuria/orina , Animales , Secuencia de Bases , Glucemia/metabolismo , Peso Corporal , Cartilla de ADN , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/orina , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/orina , Hiperglucemia/genética , Hiperglucemia/fisiopatología , Riñón/anatomía & histología , Riñón/patología , Riñón/ultraestructura , Glomérulos Renales/anatomía & histología , Ratones , Ratones Endogámicos , Ratones Noqueados , Tamaño de los Órganos , Proteína Quinasa C-alfa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
11.
Hypertension ; 66(4): 800-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26283042

RESUMEN

Autosomal-dominant hypertension with brachydactyly is a salt-independent Mendelian syndrome caused by activating mutations in the gene encoding phosphodiesterase 3A. These mutations increase the protein kinase A-mediated phosphorylation of phosphodiesterase 3A resulting in enhanced cAMP-hydrolytic affinity and accelerated cell proliferation. The phosphorylated vasodilator-stimulated phosphoprotein is diminished, and parathyroid hormone-related peptide is dysregulated, potentially accounting for all phenotypic features. Untreated patients die prematurely of stroke; however, hypertension-induced target-organ damage is otherwise hardly apparent. We conducted clinical studies of vascular function, cardiac functional imaging, platelet function in affected and nonaffected persons, and cell-based assays. Large-vessel and cardiac functions indeed seem to be preserved. The platelet studies showed normal platelet function. Cell-based studies demonstrated that available phosphodiesterase 3A inhibitors suppress the mutant isoforms. However, increasing cGMP to indirectly inhibit the enzyme seemed to have particular use. Our results shed more light on phosphodiesterase 3A activation and could be relevant to the treatment of severe hypertension in the general population.


Asunto(s)
Braquidactilia/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , ADN/genética , Hipertensión/congénito , Mutación , Adolescente , Adulto , Presión Sanguínea/fisiología , Braquidactilia/diagnóstico , Braquidactilia/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Análisis Mutacional de ADN , Ecocardiografía Doppler de Pulso , Femenino , Humanos , Hipertensión/diagnóstico , Hipertensión/enzimología , Hipertensión/genética , Immunoblotting , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Adulto Joven
12.
Nat Genet ; 47(6): 647-53, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25961942

RESUMEN

Cardiovascular disease is the most common cause of death worldwide, and hypertension is the major risk factor. Mendelian hypertension elucidates mechanisms of blood pressure regulation. Here we report six missense mutations in PDE3A (encoding phosphodiesterase 3A) in six unrelated families with mendelian hypertension and brachydactyly type E (HTNB). The syndrome features brachydactyly type E (BDE), severe salt-independent but age-dependent hypertension, an increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla, altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated. In vitro analyses of mesenchymal stem cell-derived vascular smooth muscle cells (VSMCs) and chondrocytes provided insights into molecular pathogenesis. The mutations increased protein kinase A-mediated PDE3A phosphorylation and resulted in gain of function, with increased cAMP-hydrolytic activity and enhanced cell proliferation. Levels of phosphorylated VASP were diminished, and PTHrP levels were dysregulated. We suggest that the identified PDE3A mutations cause the syndrome. VSMC-expressed PDE3A deserves scrutiny as a therapeutic target for the treatment of hypertension.


Asunto(s)
Braquidactilia/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Hipertensión/congénito , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Estudios de Casos y Controles , Diferenciación Celular , Niño , Femenino , Estudios de Asociación Genética , Células HeLa , Humanos , Hipertensión/genética , Cinética , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación Missense , Miocitos del Músculo Liso/fisiología , Linaje
13.
Am J Kidney Dis ; 43(2): 286-95, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14750094

RESUMEN

BACKGROUND: Growth arrest-specific gene 6 (Gas6) and its binding partner, the receptor tyrosine kinase Axl, are important mediators in experimental nephritis. The authors tested whether the Gas6/Axl signaling pathway participates in human renal diseases. METHODS: The authors compared 26 human renal specimens from patients with IgA nephritis, acute diffuse immune complex glomerulonephritis, acute lupus nephritis, antineutrophil cytoplasmic antibody--associated glomerulonephritis, acute transplant rejection, and normal renal tissue. Because reactive oxygen species are pivotal in inflammation, the authors tested whether the Axl/Gas6 expression is influenced by NADPH oxidase in vitro. RESULTS: Gas6 and Axl immunofluorescence was barely detectable in normal kidney. However, in disease Axl was copiously expressed in the small vessel media, glomeruli, distal tubules, and collecting ducts. Similarly, Gas6 was upregulated in the small vessel intima and media, all segments of the renal tubules, the brush border, and glomeruli. Gas6 and Axl upregulation was a prominent but nonspecific finding in these renal diseases. Cultured rat vascular smooth muscle cells and immortalized human mesangial cells were stimulated with angiotensin (Ang) II (1 x 10(-7) mol/L) for 6 or 18 hours. Confocal microscopy and Western blot showed Ang II-dependent Gas6 and Axl expression. An antisense probe against the p22 phox unit of NADPH-oxidase suppressed Ang II-induced Gas6 and Axl expression. In addition, in p47 phox knockout cells Ang II-induced Gas6 and Axl expression were blocked. CONCLUSION: GAS6/Axl signaling is involved in human renal disease. The Ang II-induced Gas6 and Axl expression may be dependent on NADPH-oxidase. Gas6 and Axl are important signaling molecules in human renal disease and may be potential therapeutic targets.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedades Renales/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Adolescente , Adulto , Anciano , Angiotensina II/farmacología , Animales , Células Cultivadas , Niño , Femenino , Mesangio Glomerular/metabolismo , Humanos , Inmunohistoquímica , Enfermedades Renales/patología , Masculino , Ratones , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , NADPH Oxidasas/farmacología , Proteínas Proto-Oncogénicas , Ratas , Transducción de Señal , Tirosina Quinasa del Receptor Axl
15.
Hypertension ; 58(3): 471-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21747041

RESUMEN

In humans, dehydroepiandrosterone (DHEA), with its sulfate, is the most abundant adrenal steroid, whereas the rat adrenals are not capable of synthesizing this steroid. Circulating concentrations of DHEA sulfate lie in the millimolar range and those of DHEA in the subnanomolar range. DHEA exerts protective potential during vascular remodeling, although the underlying mechanisms of this protection are imperfectly defined. We hypothesized that physiological doses of DHEA alter signaling pathways that are of central importance for vascular integrity. We exposed human endothelial cells, vascular smooth muscle cells, and fibroblasts to DHEA (10(-6) to 10(-10) mol/L) and observed a dose- and time-dependent increase of extracellular signal-regulated kinases 1 and 2 activation. Similar results were observed in rat vascular smooth muscle cells. In addition, in rat vascular smooth muscle cells, we found altered phosphorylation and cellular translocation of the transcription factor FoxO1. Pharmacological blockade of the mineralocorticoid receptor (MR) with eplerenone or small interfering RNA-mediated MR-silencing prevented DHEA-induced extracellular signal-regulated kinase 1/2 phosphorylation and its effects on FoxO1. Of note, in a cell-based MR transactivation assay, we did not find any agonist effect of DHEA on MR activity. We conclude that DHEA induces early signaling events in vascular cells that might underlie the DHEA-mediated protection against vasculopathies. These effects are dependent on the MR, although the finding that DHEA fails to act as a direct MR agonist suggests that additional signaling proteins are involved. In this regard, DHEA may either interact with coeffectors to modify MR activity or serves as a ligand for a yet unknown receptor that might transactivate the MR.


Asunto(s)
Deshidroepiandrosterona/farmacología , Factores de Transcripción Forkhead/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Mineralocorticoides/metabolismo , Animales , Western Blotting , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Eplerenona , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proteína Forkhead Box O1 , Humanos , Microscopía Confocal , Antagonistas de Receptores de Mineralocorticoides/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Ratas , Receptores de Mineralocorticoides/genética , Espironolactona/análogos & derivados , Espironolactona/farmacología , Factores de Tiempo
16.
Hypertension ; 58(1): 77-84, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21576625

RESUMEN

Pregnant women who subsequently develop preeclampsia are highly sensitive to infused angiotensin (Ang) II; the sensitivity persists postpartum. Activating autoantibodies against the Ang II type 1 (AT(1)) receptor are present in preeclampsia. In vitro and in vivo data suggest that they could be involved in the disease process. We generated and purified activating antibodies against the AT(1) receptor (AT(1)-AB) by immunizing rabbits against the AFHYESQ epitope of the second extracellular loop, which is the binding epitope of endogenous activating autoantibodies against AT(1) from patients with preeclampsia. We then purified AT(1)-AB using affinity chromatography with the AFHYESQ peptide. We were able to detect AT(1)-AB both by ELISA and a functional bioassay. We then passively transferred AT(1)-AB into pregnant rats, alone or combined with Ang II. AT(1)-AB activated protein kinase C-α and extracellular-related kinase 1/2. Passive transfer of AT(1)-AB alone or Ang II (435 ng/kg per minute) infused alone did not induce a preeclampsia-like syndrome in pregnant rats. However, the combination (AT(1)-AB plus Ang II) induced hypertension, proteinuria, intrauterine growth retardation, and arteriolosclerosis in the uteroplacental unit. We next performed gene-array profiling of the uteroplacental unit and found that hypoxia-inducible factor 1α was upregulated by Ang II plus AT(1)-AB, which we then confirmed by Western blotting in villous explants. Furthermore, endothelin 1 was upregulated in endothelial cells by Ang II plus AT(1)-AB. We show that AT(1)-AB induces Ang II sensitivity. Our mechanistic study supports the existence of an "autoimmune-activating receptor" that could contribute to Ang II sensitivity and possible to preeclampsia.


Asunto(s)
Angiotensina II/genética , Autoanticuerpos , Regulación del Desarrollo de la Expresión Génica , Preeclampsia/inmunología , Preñez , ARN/genética , Receptor de Angiotensina Tipo 1/inmunología , Angiotensina II/metabolismo , Animales , Western Blotting , Células Cultivadas , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Femenino , Feto/embriología , Feto/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Conejos , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo
17.
Hypertension ; 54(2): 359-64, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19564549

RESUMEN

Growth arrest-specific protein 6 (Gas 6) is involved in inflammatory kidney diseases, vascular remodeling, cell adhesion, and thrombus formation. We explored a role for Gas 6 in aldosterone-induced target organ damage. We observed that Gas 6 was upregulated in rats with high aldosterone levels. Mineralocorticoid receptor blockade prevented target organ damage and decreased the elevated Gas 6 expression. Vascular smooth muscle cells given aldosterone increased their Gas 6 expression in vitro. To test the pathophysiological relevance, we investigated the effects of deoxycorticosterone acetate (DOCA) on Gas 6 gene-deleted ((-/-)) mice. After 6 weeks DOCA, Gas 6(-/-) mice developed similar telemetric blood pressure elevations compared to wild-type mice but were protected from cardiac hypertrophy. Cardiac expression of interleukin 6 and collagen IV was blunted in Gas 6(-/-) mice, indicating reduced inflammation and fibrosis. Gas 6(-/-) mice also had an improved renal function with reduced albuminuria, compared to wild-type mice. Renal fibrosis and fibronectin deposition in the kidney were also reduced. Gas 6 deficiency reduces the detrimental effects of aldosterone on cardiac and renal remodeling independent of blood pressure reduction. Gas 6 appears to play a role in mineralocorticoid receptor-mediated target organ damage. Furthermore, because warfarin interferes with Gas 6 protein expression, the findings could be of clinical relevance for anticoagulant choices.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Cardiomegalia/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Músculo Liso Vascular/citología , Lesión Renal Aguda/patología , Albuminuria , Aldosterona/farmacología , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/patología , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Desoxicorticosterona/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Probabilidad , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
18.
Hypertension ; 51(5): 1372-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18347231

RESUMEN

Mineralocorticoid receptor blockade protects from angiotensin II-induced target-organ damage. 11beta-Hydroxysteroid dehydrogenase type 2 protects the mineralocorticoid receptor from activation by glucocorticoids; however, high glucocorticoid concentrations and absent 11beta-hydroxysteroid dehydrogenase type 2 in some tissues make glucocorticoids highly relevant mineralocorticoid receptor ligands. We investigated the effects of corticosterone (10(-6) to 10(-12) mol/L) on early vascular mineralocorticoid receptor signaling by Western blotting, confocal microscopy, and myography. Corticosterone initiated extracellular signal-regulated kinase 1/2 phosphorylation in rat vascular smooth muscle cells at > or =10(-11) mol/L doses. Protein synthesis inhibitors had no effect, indicating a nongenomic action. Corticosterone also stimulated c-Jun N-terminal kinase, p38, Src, and Akt phosphorylation at 15 minutes and enhanced angiotensin II-induced signaling at 5 minutes. A specific epidermal growth factor receptor blocker, AG1478, as well as the Src inhibitor PP2, markedly reduced corticosterone-induced extracellular signal-regulated kinase 1/2 phosphorylation, as did preincubation of cells with the mineralocorticoid receptor antagonist spironolactone. Silencing mineralocorticoid receptor with small interfering RNA abolished corticosterone-induced effects. Corticosterone (10(-9) mol/L) enhanced phenylephrine-induced contraction of intact aortic rings. These effects were dependent on the intact endothelium, mineralocorticoid receptor, and mitogen-activated protein kinase kinase 1/extracellular signal-regulated kinase signaling. We conclude that corticosterone induces rapid mineralocorticoid receptor signaling in vascular smooth muscle cells that involves mitogen-activated protein kinase kinase/extracellular signal-regulated kinase-dependent pathways. These new mineralocorticoid receptor-dependent signaling pathways suggest that glucocorticoids may contribute to vascular disease via mineralocorticoid receptor signaling, independent of circulating aldosterone.


Asunto(s)
Aorta/efectos de los fármacos , Corticosterona/farmacología , Músculo Liso Vascular/efectos de los fármacos , Receptores de Mineralocorticoides/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Proteína Tirosina Quinasa CSK , Células Cultivadas , Relación Dosis-Respuesta a Droga , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/fisiología , Vasoconstricción/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas
19.
PLoS One ; 3(11): e3742, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19011682

RESUMEN

BACKGROUND: Agonistic autoantibodies directed at the alpha(1)-adrenergic receptor (alpha(1)-AAB) have been described in patients with hypertension. We implied earlier that alpha(1)-AAB might have a mechanistic role and could represent a therapeutic target. METHODOLOGY/PRINCIPAL FINDINGS: To pursue the issue, we performed clinical and basic studies. We observed that 41 of 81 patients with refractory hypertension had alpha(1)-AAB; after immunoadsorption blood pressure was significantly reduced in these patients. Rabbits were immunized to generate alpha(1)-adrenergic receptor antibodies (alpha(1)-AB). Patient alpha(1)-AAB and rabbit alpha(1)-AB were purified using affinity chromatography and characterized both by epitope mapping and surface plasmon resonance measurements. Neonatal rat cardiomyocytes, rat vascular smooth muscle cells (VSMC), and Chinese hamster ovary cells transfected with the human alpha(1A)-adrenergic receptor were incubated with patient alpha(1)-AAB and rabbit alpha(1)-AB and the activation of signal transduction pathways was investigated by Western blot, confocal laser scanning microscopy, and gene expression. We found that phospholipase A2 group IIA (PLA2-IIA) and L-type calcium channel (Cacna1c) genes were upregulated in cardiomyocytes and VSMC after stimulation with both purified antibodies. We showed that patient alpha(1)-AAB and rabbit alpha(1)-AB result in protein kinase C alpha activation and transient extracellular-related kinase (EKR1/2) phosphorylation. Finally, we showed that the antibodies exert acute effects on intracellular Ca(2+) in cardiomyocytes and induce mesentery artery segment contraction. CONCLUSIONS/SIGNIFICANCE: Patient alpha(1)-AAB and rabbit alpha(1)-AB can induce signaling pathways important for hypertension and cardiac remodeling. Our data provide evidence for a potential clinical relevance for alpha(1)-AAB in hypertensive patients, and the notion of immunity as a possible cause of hypertension.


Asunto(s)
Autoanticuerpos/inmunología , Hipertensión/inmunología , Receptores Adrenérgicos alfa/inmunología , Adsorción/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Animales , Autoanticuerpos/aislamiento & purificación , Autoanticuerpos/farmacología , Presión Sanguínea/efectos de los fármacos , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Activación Enzimática/efectos de los fármacos , Mapeo Epitopo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Hipertensión/fisiopatología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Fosfolipasas A2/metabolismo , Proteína Quinasa C/metabolismo , Estructura Secundaria de Proteína , Ratas , Receptores Adrenérgicos alfa/química
20.
Am J Pathol ; 170(4): 1192-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17392159

RESUMEN

Statins induce heme oxygenase-1 (HO-1) in several cell types, such as vascular smooth muscle cells, endothelial cells, and macrophages. The present study assessed the role of statin-induced HO-1 up-regulation on circulating monocytes/macrophages and their contribution in preventing renal ischemia-reperfusion (IR) injury in a rat model. Cerivastatin was administered via gavage (0.5 mg/kg) for 3 days before IR injury; controls received vehicle. Statin pretreatment reduced renal damage and attenuated renal dysfunction (P < 0.05) after IR injury. The protective statin pretreatment effect was completely abolished by cotreatment with tin protoporphyrin IX (Sn-PP), a competitive HO inhibitor. IR increased HO-1 expression at the transcript and protein level in renal tissue. This effect was significantly more evident (P < 0.05) in the statin-pretreated animals 24 hours after IR injury. We identified infiltrating macrophages as the major source of tissue HO-1 production. Moreover, in ancillary cell culture (monocyte cell line) and in in vivo experiments (isolation of circulating monocytes), we confirmed that statins regulate HO-1 expression in these cells. We conclude that statin treatment up-regulates HO-1 in circulating monocytes/macrophages in vivo and in vitro. We hypothesize that local delivery of HO-1 from infiltrating macrophages exerts anti-inflammatory effects after IR injury and thereby may reduce tissue destruction.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Macrófagos/efectos de los fármacos , Piridinas/farmacología , Daño por Reperfusión/prevención & control , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Riñón/fisiopatología , Macrófagos/enzimología , Macrófagos/patología , Masculino , Microscopía Confocal , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/enzimología , Protoporfirinas/administración & dosificación , Protoporfirinas/farmacología , Piridinas/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA