Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Intervalo de año de publicación
2.
Arterioscler Thromb Vasc Biol ; 38(10): 2484-2497, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354220

RESUMEN

Objective- EMILIN-1 (elastin microfibrils interface located protein-1) protein inhibits pro-TGF-ß (transforming growth factor-ß) proteolysis and limits TGF-ß bioavailability in vascular extracellular matrix. Emilin1-/- null mice display increased vascular TGF-ß signaling and are hypertensive. Because EMILIN-1 is expressed in vessels from embryonic life to adulthood, we aimed at unravelling whether the hypertensive phenotype of Emilin1-/- null mice results from a developmental defect or lack of homeostatic role in the adult. Approach and Results- By using a conditional gene targeting inactivating EMILIN-1 in smooth muscle cells of adult mice, we show that increased blood pressure in mice with selective smooth muscle cell ablation of EMILIN-1 depends on enhanced myogenic tone. Mechanistically, we unveil that higher TGF-ß signaling in smooth muscle cells stimulates HB-EGF (heparin-binding epidermal growth factor) expression and subsequent transactivation of EGFR (epidermal growth factor receptor). With increasing intraluminal pressure in resistance arteries, the cross talk established by TGF-ß and EGFR signals recruits TRPC6 (TRP [transient receptor potential] classical type 6) and TRPM4 (TRP melastatin type 4) channels, lastly stimulating voltage-dependent calcium channels and potentiating myogenic tone. We found reduced EMILIN-1 and enhanced myogenic tone, dependent on increased TGF-ß-EGFR signaling, in resistance arteries from hypertensive patients. Conclusions- Taken together, our findings implicate an unexpected role of the TGF-ß-EGFR pathway in hypertension with current translational perspectives.


Asunto(s)
Receptores ErbB/metabolismo , Hipertensión/metabolismo , Glicoproteínas de Membrana/metabolismo , Arterias Mesentéricas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Vasoconstricción , Animales , Presión Sanguínea , Canales de Calcio/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Canales Catiónicos TRPM/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Vasoconstricción/efectos de los fármacos
3.
Cardiovasc Res ; 114(3): 456-467, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29324984

RESUMEN

AIMS: Chronic increase of mineralocorticoids obtained by administration of deoxycorticosterone acetate (DOCA) results in salt-dependent hypertension in animals. Despite the lack of a generalized sympathoexcitation, DOCA-salt hypertension has been also associated to overdrive of peripheral nervous system in organs typically targeted by blood pressure (BP), as kidneys and vasculature. Aim of this study was to explore whether DOCA-salt recruits immune system by overactivating sympathetic nervous system in lymphoid organs and whether this is relevant for hypertension. METHODS AND RESULTS: To evaluate the role of the neurosplenic sympathetic drive in DOCA-salt hypertension, we challenged splenectomized mice or mice with left coeliac ganglionectomy with DOCA-salt, observing that they were both unable to increase BP. Then, we evaluated by immunofluorescence and ELISA levels of the placental growth factor (PlGF) upon DOCA-salt challenge, which significantly increased the growth factor expression, but only in the presence of an intact neurosplenic sympathetic drive. When PlGF KO mice were subjected to DOCA-salt, they were significantly protected from the increased BP observed in WT mice under same experimental conditions. In addition, absence of PlGF hampered DOCA-salt mediated T cells co-stimulation and their consequent deployment towards kidneys where they infiltrated tissue and provoked end-organ damage. CONCLUSION: Overall, our study demonstrates that DOCA-salt requires an intact sympathetic drive to the spleen for priming of immunity and consequent BP increase. The coupling of nervous system and immune cells activation in the splenic marginal zone is established through a sympathetic-mediated PlGF release, suggesting that this pathway could be a valid therapeutic target for hypertension.


Asunto(s)
Presión Sanguínea , Acetato de Desoxicorticosterona , Ganglios Simpáticos/fisiopatología , Hipertensión/metabolismo , Activación de Linfocitos , Neuroinmunomodulación , Factor de Crecimiento Placentario/metabolismo , Bazo/inervación , Bazo/metabolismo , Linfocitos T/metabolismo , Animales , Modelos Animales de Enfermedad , Ganglios Simpáticos/cirugía , Ganglionectomía , Hipertensión/inducido químicamente , Hipertensión/inmunología , Hipertensión/fisiopatología , Riñón/inmunología , Riñón/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Crecimiento Placentario/deficiencia , Factor de Crecimiento Placentario/genética , Bazo/inmunología , Esplenectomía , Linfocitos T/inmunología
4.
Immunity ; 47(5): 959-973.e9, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29150241

RESUMEN

Aortic aneurysms are life-threatening conditions with effective treatments mainly limited to emergency surgery or trans-arterial endovascular stent grafts, thus calling for the identification of specific molecular targets. Genetic studies have highlighted controversial roles of transforming growth factor ß (TGF-ß) signaling in aneurysm development. Here, we report on aneurysms developing in adult mice after smooth muscle cell (SMC)-specific inactivation of Smad4, an intracellular transducer of TGF-ß. The results revealed that Smad4 inhibition activated interleukin-1ß (IL-1ß) in SMCs. This danger signal later recruited innate immunity in the adventitia through chemokine (C-C motif) ligand 2 (CCL2) and modified the mechanical properties of the aortic wall, thus favoring vessel dilation. SMC-specific Smad4 deletion in Il1r1- or Ccr2-null mice resulted in milder aortic pathology. A chronic treatment with anti-IL-1ß antibody effectively hampered aneurysm development. These findings identify a mechanistic target for controlling the progression of aneurysms with compromised TGF-ß signaling, such as those driven by SMAD4 mutations.


Asunto(s)
Aneurisma de la Aorta/prevención & control , Interleucina-1beta/antagonistas & inhibidores , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Ratones , Miocitos del Músculo Liso/inmunología , FN-kappa B/fisiología , Receptores CCR2/antagonistas & inhibidores , Proteína Smad4/fisiología , Tamoxifeno/farmacología
5.
Rev. esp. cardiol. (Ed. impr.) ; 70(1): 16-24, ene. 2017. graf, tab, ilus
Artículo en Español | IBECS | ID: ibc-159299

RESUMEN

Introducción y objetivos: Las enfermedades cardiovasculares, incluida la miocardiopatía, son las principales complicaciones de la diabetes mellitus. Un conocimiento más profundo de los mecanismos moleculares que conducen a la miocardiopatía es crucial para el desarrollo de nuevos tratamientos. Se propuso la fosfoinosítido 3-cinasa gamma (PI3Kγ) como objetivo molecular contra la miocardiopatía diabética, dado el papel que desempeña la PI3Kγ en el remodelado cardiaco frente a la sobrecarga de presión. Dada la disponibilidad de un inhibidor farmacológico de este objetivo molecular, el GE21, se ha investigado la validez de nuestra hipótesis induciendo la diabetes a ratones con ablación genética de la PI3Kγ, o knock-in, para una PI3Kγ catalíticamente inactiva. Métodos: Se provocó la diabetes a los ratones empleando estreptozotocina. Se evaluó la función cardiaca mediante exámenes ecocardiográficos secuenciales, mientras que la fibrosis y la inflamación se evaluaron mediante examen histológico. Resultados: La diabetes mellitus indujo disfunción cardiaca en los ratones genéticamente intactos (wild-type). La disfunción sistólica se previno completamente y la diastólica parcialmente, tanto en ratones con PI3Kγ inactivada como ratones sin actividad de cinasa. La disfunción cardiaca se recuperó en la misma medida administrando el inhibidor de la PI3Kγ GE21, de manera dependiente de la dosis. Estas acciones de inhibición genética o farmacológica de la PI3Kγ se asociaron con una reducción de la fibrosis y la inflamación en el corazón diabético. Conclusiones: Nuestro estudio demuestra un papel fundamental de la PI3Kγ en la miocardiopatía diabética en ratones y el efecto beneficioso de la inhibición farmacológica de la PI3Kγ; destaca su potencial como estrategia prometedora para el tratamiento clínico de las complicaciones cardiacas de los pacientes diabéticos (AU)


Introduction and objectives: Cardiovascular diseases, including cardiomyopathy, are the major complications in diabetes. A deeper understanding of the molecular mechanisms leading to cardiomyopathy is critical for developing novel therapies. We proposed phosphoinositide3-kinase gamma (PI3Kγ) as a molecular target against diabetic cardiomyopathy, given the role of PI3Kγ in cardiac remodeling to pressure overload. Given the availability of a pharmacological inhibitor of this molecular target GE21, we tested the validity of our hypothesis by inducing diabetes in mice with genetic ablation of PI3Kγ or knock-in for a catalytically inactive PI3Kγ. Methods: Mice were made diabetic by streptozotocin. Cardiac function was assessed by serial echocardiographic analyses, while fibrosis and inflammation were evaluated by histological analysis. Results: Diabetes induced cardiac dysfunction in wild-type mice. Systolic dysfunction was completely prevented, and diastolic dysfunction was partially blocked, in both PI3Kγ knock-out and kinase-dead mice. Cardiac dysfunction was similarly rescued by administration of the PI3Kγ inhibitor GE21 in a dose-dependent manner. These actions of genetic and pharmacological PI3Kγ inhibition were associated with a decrease in inflammation and fibrosis in diabetic hearts. Conclusions: Our study demonstrates a fundamental role of PI3Kγ in diabetic cardiomyopathy in mice and the beneficial effect of pharmacological PI3Kγ inhibition, highlighting its potential as a promising strategy for clinical treatment of cardiac complications of diabetic patients (AU)


Asunto(s)
Animales , Ratones , Cardiomiopatías Diabéticas/prevención & control , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/farmacocinética , Modelos Animales de Enfermedad , Drogas en Investigación/farmacocinética , Pruebas de Función Cardíaca , Inflamación/patología , Fibrosis/patología
6.
Rev Esp Cardiol (Engl Ed) ; 70(1): 16-24, 2017 Jan.
Artículo en Inglés, Español | MEDLINE | ID: mdl-27422446

RESUMEN

INTRODUCTION AND OBJECTIVES: Cardiovascular diseases, including cardiomyopathy, are the major complications in diabetes. A deeper understanding of the molecular mechanisms leading to cardiomyopathy is critical for developing novel therapies. We proposed phosphoinositide3-kinase gamma (PI3Kγ) as a molecular target against diabetic cardiomyopathy, given the role of PI3Kγ in cardiac remodeling to pressure overload. Given the availability of a pharmacological inhibitor of this molecular target GE21, we tested the validity of our hypothesis by inducing diabetes in mice with genetic ablation of PI3Kγ or knock-in for a catalytically inactive PI3Kγ. METHODS: Mice were made diabetic by streptozotocin. Cardiac function was assessed by serial echocardiographic analyses, while fibrosis and inflammation were evaluated by histological analysis. RESULTS: Diabetes induced cardiac dysfunction in wild-type mice. Systolic dysfunction was completely prevented, and diastolic dysfunction was partially blocked, in both PI3Kγ knock-out and kinase-dead mice. Cardiac dysfunction was similarly rescued by administration of the PI3Kγ inhibitor GE21 in a dose-dependent manner. These actions of genetic and pharmacological PI3Kγ inhibition were associated with a decrease in inflammation and fibrosis in diabetic hearts. CONCLUSIONS: Our study demonstrates a fundamental role of PI3Kγ in diabetic cardiomyopathy in mice and the beneficial effect of pharmacological PI3Kγ inhibition, highlighting its potential as a promising strategy for clinical treatment of cardiac complications of diabetic patients.


Asunto(s)
Cardiomiopatías Diabéticas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Cardiomiopatías Diabéticas/diagnóstico , Cardiomiopatías Diabéticas/enzimología , Cardiomiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Ecocardiografía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/enzimología , Miocardio/patología
7.
Nat Commun ; 7: 13035, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27676657

RESUMEN

The crucial role of the immune system in hypertension is now widely recognized. We previously reported that hypertensive challenges couple the nervous drive with immune system activation, but the physiological and molecular mechanisms of this connection are unknown. Here, we show that hypertensive challenges activate splenic sympathetic nerve discharge to prime immune response. More specifically, a vagus-splenic nerve drive, mediated by nicotinic cholinergic receptors, links the brain and spleen. The sympathetic discharge induced by hypertensive stimuli was absent in both coeliac vagotomized mice and in mice lacking α7nAChR, a receptor typically expressed by peripheral ganglionic neurons. This cholinergic-sympathetic pathway is necessary for T cell activation and egression on hypertensive challenges. In addition, we show that selectively thermoablating the splenic nerve prevents T cell egression and protects against hypertension. This novel experimental procedure for selective splenic denervation suggests new clinical strategies for resistant hypertension.

8.
Mol Metab ; 4(10): 741-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26500845

RESUMEN

OBJECTIVE: Tissue inhibitor of metalloproteinase 3 (TIMP3) is an extracellular matrix (ECM) bound protein, which has been shown to be downregulated in human subjects and experimental models with cardiometabolic disorders, including type 2 diabetes mellitus, hypertension and atherosclerosis. The aim of this study was to investigate the effects of TIMP3 on cardiac energy homeostasis during increased metabolic stress conditions. METHODS: ApoE(-/-)TIMP3(-/-) and ApoE(-/-) mice on a C57BL/6 background were subjected to telemetric ECG analysis and experimental myocardial infarction as models of cardiac stress induction. We used Western blot, qRT-PCR, histology, metabolomics, RNA-sequencing and in vivo phenotypical analysis to investigate the molecular mechanisms of altered cardiac energy metabolism. RESULTS: ApoE(-/-)TIMP3(-/-) revealed decreased lifespan. Telemetric ECG analysis showed increased arrhythmic episodes, and experimental myocardial infarction by left anterior descending artery (LAD) ligation resulted in increased peri-operative mortality together with increased scar formation, ventricular dilatation and a reduction of cardiac function after 4 weeks in the few survivors. Hearts of ApoE(-/-)TIMP3(-/-) exhibited accumulation of neutral lipids when fed a chow diet, which was exacerbated by a high fat, high cholesterol diet. Metabolomics analysis revealed an increase in circulating markers of oxidative stress with a reduction in long chain fatty acids. Using whole heart mRNA sequencing, we identified apelin as a putative modulator of these metabolic defects. Apelin is a regulator of fatty acid oxidation, and we found a reduction in the levels of enzymes involved in fatty acid oxidation in the left ventricle of ApoE(-/-)TIMP3(-/-) mice. Injection of apelin restored the hitherto identified metabolic defects of lipid oxidation. CONCLUSION: TIMP3 regulates lipid metabolism as well as oxidative stress response via apelin. These findings therefore suggest that TIMP3 maintains metabolic flexibility in the heart, particularly during episodes of increased cardiac stress.

9.
Immunity ; 41(5): 737-52, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25517614

RESUMEN

Hypertension is a health problem affecting over 1 billion people worldwide. How the immune system gets activated under hypertensive stimuli to contribute to blood pressure elevation is a fascinating enigma. Here we showed a splenic role for placental growth factor (PlGF), which accounts for the onset of hypertension, through immune system modulation. PlGF repressed the expression of the protein Timp3 (tissue inhibitor of metalloproteinases 3), through the transcriptional Sirt1-p53 axis. Timp3 repression allowed costimulation of T cells and their deployment toward classical organs involved in hypertension. We showed that the spleen is an essential organ for the development of hypertension through a noradrenergic drive mediated by the celiac ganglion efferent. Overall, we demonstrate that PlGF mediates the neuroimmune interaction in the spleen, organizing a unique and nonredundant response that allows the onset of hypertension.


Asunto(s)
Presión Sanguínea/inmunología , Hipertensión/inmunología , Proteínas Gestacionales/inmunología , Bazo/inmunología , Angiotensina II/inmunología , Animales , Presión Sanguínea/genética , Ganglios Simpáticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Interferencia de ARN , ARN Interferente Pequeño , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Linfocitos T/inmunología , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteína p53 Supresora de Tumor/genética
10.
Arterioscler Thromb Vasc Biol ; 32(9): 2178-84, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22814752

RESUMEN

OBJECTIVE: Emilin-1 is a protein of elastic extracellular matrix involved in blood pressure (BP) control by negatively affecting transforming growth factor (TGF)-ß processing. Emilin1 null mice are hypertensive. This study investigates how Emilin-1 deals with vascular mechanisms regulating BP. METHODS AND RESULTS: This study uses a phenotype rescue approach in which Emilin-1 is expressed in either endothelial cells or vascular smooth muscle cells of transgenic animals with the Emilin1(-/-) background. We found that normalization of BP required Emilin-1 expression in smooth muscle cells, whereas expression of the protein in endothelial cells did not modify the hypertensive phenotype of Emilin1(-/-) mice. We also explored the effect of treatment with anti-TGF-ß antibodies on the hypertensive phenotype of Emilin1(-/-) mice, finding that neutralization of TGF-ß in Emilin1 null mice normalized BP quite rapidly (2 weeks). Finally, we evaluated the vasoconstriction response of resistance arteries to perfusion pressure and neurohumoral agents in different transgenic mouse lines. Interestingly, we found that the hypertensive phenotype was coupled with an increased arteriolar myogenic response to perfusion pressure, while the vasoconstriction induced by neurohumoral agents remained unaffected. We further elucidate that, as for the hypertensive phenotype, the increased myogenic response was attributable to increased TGF-ß activity. CONCLUSIONS: Our findings clarify that Emilin-1 produced by vascular smooth muscle cells acts as a main regulator of resting BP levels by controlling the myogenic response in resistance arteries through TGF-ß.


Asunto(s)
Presión Sanguínea , Hipertensión/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Vasoconstricción , Animales , Anticuerpos Neutralizantes/administración & dosificación , Arteriolas/metabolismo , Arteriolas/fisiopatología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Monitoreo Ambulatorio de la Presión Arterial/métodos , Relación Dosis-Respuesta a Droga , Ecocardiografía Doppler , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Genotipo , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Telemetría , Factores de Tiempo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstricción/genética , Vasoconstrictores/farmacología
11.
Neurobiol Aging ; 33(1): 205.e19-29, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20961666

RESUMEN

Hypertension and sporadic Alzheimer's disease (AD) have been associated but clear pathophysiological links have not yet been demonstrated. Hypertension and AD share inflammation as a pathophysiological trait. Thus, we explored if modulating neuroinflammation could influence hypertension-induced ß-amyloid (Aß) deposition. Possible interactions among hypertension, inflammation and Aß-deposition were studied in hypertensive mice with transverse aortic coarctation (TAC). Given that brain Aß deposits are detectable as early as 4 weeks after TAC, brain pathology was analyzed in 3-week TAC mice, before Aß deposition, and at a later time (8-week TAC mice). Microglial activation and interleukin (IL)-1ß upregulation were already found in 3-week TAC mice. At a later time, along with evident Aß deposition, microglia was still activated. Finally, immune system stimulation (LPS) or inhibition (ibuprofen), strategies described to positively or negatively modulate neuroinflammation, differently affected Aß deposition. We demonstrate that hypertension per se triggers neuroinflammation before Aß deposition. The finding that only immune system activation, but not its inhibition, strongly reduced amyloid burden suggests that stimulating inflammation in the appropriate time window may represent a promising strategy to limit vascular-triggered AD-pathology.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Hipertensión/metabolismo , Inflamación Neurogénica/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Animales , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad , Hipertensión/complicaciones , Hipertensión/fisiopatología , Ibuprofeno/administración & dosificación , Sistema Inmunológico/inmunología , Interleucina-1beta/metabolismo , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Inflamación Neurogénica/complicaciones , Inflamación Neurogénica/fisiopatología , Regulación hacia Arriba
12.
Cardiovasc Res ; 93(1): 200-9, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22038741

RESUMEN

AIMS: The lipid and protein kinase phosphoinositide 3-kinase γ (PI3Kγ) is abundantly expressed in inflammatory cells and in the cardiovascular tissue. In recent years, its role in inflammation and in cardiac function and remodelling has been unravelled, highlighting the beneficial effects of its pharmacological inhibition. Furthermore, a role for PI3Kγ in the regulation of vascular tone has been emphasized. However, the impact of this signalling in the control of blood pressure is still poorly understood. Our study investigated the effect of a selective inhibition of PI3Kγ, obtained by using two independent small molecules, on blood pressure. Moreover, we dissected the molecular mechanisms involved in control of contraction of resistance arteries by PI3Kγ. METHODS AND RESULTS: We showed that inhibition of PI3Kγ reduced blood pressure in normotensive and hypertensive mice in a concentration-dependent fashion. This effect was dependent on enhanced vasodilatation, documented in vivo by decreased peripheral vascular resistance, and ex vivo by vasorelaxing effects on isolated resistance vessels. The vasorelaxation induced by PI3Kγ inhibition relied on blunted pressure-induced Akt phosphorylation and a myogenic contractile response. Molecular insights revealed that PI3Kγ inhibition affected smooth muscle L-type calcium channel current density and calcium influx by impairing plasma membrane translocation of the α1C L-type calcium channel subunit responsible for channel open-state probability. CONCLUSION: Overall our findings suggest that PI3Kγ inhibition could be a novel tool to modulate calcium influx in vascular smooth muscle cells, thus relaxing resistance arteries and lowering blood pressure.


Asunto(s)
Presión Sanguínea/fisiología , Canales de Calcio Tipo L/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Inhibidores Enzimáticos/farmacología , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Quinoxalinas/farmacología , Tiazolidinedionas/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
13.
Circulation ; 124(12): 1337-50, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21900081

RESUMEN

BACKGROUND: Heart failure is one of the leading causes of mortality and is primarily the final stage of several overload cardiomyopathies, preceded by an early adaptive hypertrophic response and characterized by coordinated cardiomyocyte growth, angiogenesis, and inflammation. Therefore, growth factors and cytokines have to be critically regulated during cardiac response to transverse aortic constriction. Interestingly, the dual properties of placental growth factor as an angiogenic factor and cytokine make it a candidate to participate in cardiac remodeling in response to hemodynamic overload. METHODS AND RESULTS: After transverse aortic constriction, placental growth factor knockout mice displayed a dysregulation of cardiac remodeling, negatively affecting muscle growth. Molecular insights underscored that this effect was ascribable mainly to a failure in the establishment of adequate inflammatory response owing to an impaired activity of tumor necrosis factor-α-converting enzyme. Interestingly, after transverse aortic constriction, placental growth factor knockout mice had strongly increased levels of tissue inhibitor of metalloproteinases-3, the main natural TACE inhibitor, thus indicating an unbalance of the tissue inhibitor of metalloproteinases-3/tumor necrosis factor-α-converting enzyme axis. Strikingly, when we used an in vivo RNA interference approach to reduce tissue inhibitor of metalloproteinases-3 levels in placental growth factor knockout mice during transverse aortic constriction, we obtained a complete phenotype rescue of early dilated cardiomyopathy. CONCLUSIONS: Our results demonstrate that placental growth factor finely tunes a balanced regulation of the tissue inhibitor of metalloproteinases-3/tumor necrosis factor-α-converting enzyme axis and the consequent TNF-α activation in response to transverse aortic constriction, thus allowing the establishment of an inflammatory response necessary for adaptive cardiac remodeling.


Asunto(s)
Proteínas ADAM/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Miocarditis/fisiopatología , Proteínas Gestacionales/fisiología , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Remodelación Ventricular/fisiología , Proteínas ADAM/fisiología , Proteína ADAM17 , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/fisiología , Animales , Aorta/fisiopatología , Cardiomiopatía Dilatada/tratamiento farmacológico , Cardiomiopatía Dilatada/fisiopatología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Masculino , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Proteínas Gestacionales/farmacología , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Presión Ventricular/efectos de los fármacos , Presión Ventricular/fisiología , Remodelación Ventricular/efectos de los fármacos
14.
Cardiovasc Res ; 91(3): 456-64, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21493702

RESUMEN

AIMS: The Raf-MEK1/2-ERK1/2 (ERK1/2-extracellular signal-regulated kinases 1/2) signalling cascade is crucial in triggering cardiac responses to different stress stimuli. Scaffold proteins are key elements in coordinating signalling molecules for their appropriate spatiotemporal activation. Here, we investigated the role of IQ motif-containing GTPase-activating protein 1 (IQGAP1), a scaffold for the ERK1/2 cascade, in heart function and remodelling in response to pressure overload. METHODS AND RESULTS: IQGAP1-null mice have unaltered basal heart function. When subjected to pressure overload, IQGAP1-null mice initially develop a compensatory hypertrophy indistinguishable from that of wild-type (WT) mice. However, upon a prolonged stimulus, the hypertrophic response develops towards a thinning of left ventricular walls, chamber dilation, and a decrease in contractility, in an accelerated fashion compared with WT mice. This unfavourable cardiac remodelling is characterized by blunted reactivation of the foetal gene programme, impaired cardiomyocyte hypertrophy, and increased cardiomyocyte apoptosis. Analysis of signalling pathways revealed two temporally distinct waves of both ERK1/2 and AKT phosphorylation peaking, respectively, at 10 min and 4 days after aortic banding in WT hearts. IQGAP1-null mice show strongly impaired phosphorylation of MEK1/2-ERK1/2 and AKT following 4 days of pressure overload, but normal activation of these kinases after 10 min. Pull-down experiments indicated that IQGAP1 is able to bind the three components of the ERK cascade, namely c-Raf, MEK1/2, and ERK1/2, as well as AKT in the heart. CONCLUSION: These data demonstrate, for the first time, a key role for the scaffold protein IQGAP1 in integrating hypertrophy and survival signals in the heart and regulating long-term left ventricle remodelling upon pressure overload.


Asunto(s)
Presión Sanguínea , Hipertensión/complicaciones , Hipertrofia Ventricular Izquierda/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocardio/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Remodelación Ventricular , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Aorta/fisiopatología , Aorta/cirugía , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Ligadura , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Miocardio/patología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Factores de Tiempo , Ultrasonografía , Proteínas Activadoras de ras GTPasa/deficiencia , Proteínas Activadoras de ras GTPasa/genética
15.
Circulation ; 123(4): 391-9, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21242482

RESUMEN

BACKGROUND: Signaling from phosphoinositide 3-kinase γ (PI3Kγ) is crucial for leukocyte recruitment and inflammation but also contributes to cardiac maladaptive remodeling. To better understand the translational potential of these findings, this study investigates the role of PI3Kγ activity in pressure overload-induced heart failure, addressing the distinct contributions of bone marrow-derived and cardiac cells. METHODS AND RESULTS: After transverse aortic constriction, mice knock-in for a catalytically inactive PI3Kγ (PI3Kγ KD) showed reduced fibrosis and normalized cardiac function up to 16 weeks. Accordingly, treatment with a selective PI3Kγ inhibitor prevented transverse aortic constriction-induced fibrosis. To define the cell types involved in this protection, bone marrow chimeras, lacking kinase activity in the immune system or the heart, were studied after transverse aortic constriction. Bone marrow-derived cells from PI3Kγ KD mice were not recruited to wild-type hearts, thus preventing fibrosis and preserving diastolic function. After prolonged pressure overload, chimeras with PI3Kγ KD bone marrow-derived cells showed slower development of left ventricular dilation and higher fractional shortening than controls. Conversely, in the presence of a wild-type immune system, KD hearts displayed bone marrow-derived cell infiltration and fibrosis at early stages but reduced left ventricular dilation and preserved contractile function at later time points. CONCLUSIONS: Together, these data demonstrate that, in response to transverse aortic constriction, PI3Kγ contributes to maladaptive remodeling at multiple levels by modulating both cardiac and immune cell functions.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Insuficiencia Cardíaca/enzimología , Leucocitos/enzimología , Miocardio/enzimología , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Fibrosis/genética , Técnicas de Sustitución del Gen , Corazón/fisiopatología , Insuficiencia Cardíaca/genética , Ratones , Ratones Endogámicos C57BL , Remodelación Ventricular/genética
16.
Hypertension ; 54(1): 150-6, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19470883

RESUMEN

Hypertension can lead to subarachnoid hemorrhage and eventually to cerebral vasospasm. It has been suggested that the latter could be the result of oxidative stress and an inflammatory response evoked by subarachnoid hemorrhage. Because an unavoidable consequence of hemorrhage is lysis of red blood cells, we first tested the hypothesis on carotid arteries that the proinflammatory cytokine tumor necrosis factor-alpha contributes to vascular oxidative stress evoked by hemolysis. We observed that hemolysis induces a significant increase in tumor necrosis factor-alpha both in blood and in vascular tissues, where it provokes Rac-1/NADPH oxidase-mediated oxidative stress and vasoconstriction. Furthermore, we extended our observations to cerebral vessels, demonstrating that tumor necrosis factor-alpha triggered this mechanism on the basilar artery. Finally, in an in vivo model of subarachnoid hemorrhage obtained by the administration of hemolyzed blood in the cisterna magna, we demonstrated, by high-resolution ultrasound analysis, that tumor necrosis factor-alpha inhibition prevented and resolved acute cerebral vasoconstriction. Moreover, tumor necrosis factor-alpha inhibition rescued the hemolysis-induced brain injury, evaluated with the method of 2,3,5-triphenyltetrazolium chloride and by the histological analysis of pyknotic nuclei. In conclusion, our results demonstrate that tumor necrosis factor-alpha plays a crucial role in the onset of hemolysis-induced vascular injury and can be used as a novel target of the therapeutic strategy against cerebral vasospasm.


Asunto(s)
Hemólisis , Hemorragia Subaracnoidea/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo , Vasoconstricción/fisiología , Vasoespasmo Intracraneal/fisiopatología , Animales , Anticuerpos Monoclonales/farmacología , Arteria Basilar/efectos de los fármacos , Arteria Basilar/patología , Arteria Basilar/fisiopatología , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades Neurodegenerativas/prevención & control , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/sangre , Hemorragia Subaracnoidea/complicaciones , Factor de Necrosis Tumoral alfa/inmunología , Vasoconstricción/efectos de los fármacos , Vasoespasmo Intracraneal/sangre , Proteínas de Unión al GTP rac/metabolismo
17.
Epilepsia ; 50 Suppl 1: 59-64, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19125850

RESUMEN

PURPOSE: The role of alpha 1b-adrenergic receptor (alpha 1b-AR) in relation with neuronal degeneration, drug addiction, and seizure susceptibility has recently emerged. In particular, mice that overexpress alpha 1b-AR undergo spontaneous epileptic seizures and progressive neuronal loss in a variety of brain areas. Therefore, one should expect that the blockade of alpha 1b-AR leads to anticonvulsant and neuroprotective effects. However, the lack of alpha 1b-AR antagonists does not allow testing of this hypothesis. METHODS: The development of alpha 1b-AR knockout (KO) mice led us to measure seizure susceptibility and neurodegeneration following systemic excitotoxins in these mice. RESULTS: We found that alpha 1b-AR KO mice are markedly resistant to kainate- and pilocarpine-induced seizures. Moreover, when marked seizure duration and severity are obtained by doubling the dose of chemoconvulsants in alpha 1b-AR KO, neuronal degeneration never occurs. CONCLUSIONS: These data indicate that alpha 1b-AR per se plays a fundamental role in the mechanisms responsible for seizure onset, severity, and duration, whereas the brain damage observed in alpha 1b-AR-overexpressing mice is likely to be a secondary phenomenon. In fact, the absence of alpha 1b-AR confers resistance to neurotoxicity induced by seizures/chemoconvulsants. These data, although confirming a pivotal role of alpha 1b-AR in modulating seizure threshold and neuronal death, offer a novel target, which may be used to develop novel anticonvulsants and neuroprotective agents.


Asunto(s)
Epilepsia/genética , Epilepsia/prevención & control , Predisposición Genética a la Enfermedad , Receptores Adrenérgicos alfa 1/deficiencia , Receptores Adrenérgicos alfa 1/genética , Animales , Encéfalo/patología , Epilepsia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Receptores Adrenérgicos alfa 1/biosíntesis , Receptores Adrenérgicos alfa 1/fisiología , Índice de Severidad de la Enfermedad
18.
Neurobiol Aging ; 30(2): 222-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17673335

RESUMEN

There are conflicting evidence regarding the association of hypertension with Alzheimer's disease (AD), and so far it is still unexplored whether increased blood pressure levels can be mechanistically related to the pathophysiology of AD. Since the deposition of beta-amyloid (A beta) in brain represents the first pathogenetic event in the onset of AD, in this study we investigated the role of hypertension in the brain deposition of A beta. We analyzed two independent mouse models of hypertension. In both models we observed an increased permeability of blood-brain barrier in cortex and hippocampus. More interestingly, in the same areas hypertensive mice showed a marked positivity to anti-A beta antibodies and the presence of A beta-like fragments. Finally, we analyzed mice after passive immunotherapy with anti-A beta IgG. We observed that this latter approach determined a markedly reduced A beta immunopositivity in both cortex and hippocampus. Our study demonstrates that chronic hypertension determines an impairment of the blood-brain barrier permeability with deposition of A beta in brain tissue and that passive immunotherapy prevents this latter phenomenon.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Hipertensión/terapia , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Tisular , Resultado del Tratamiento
19.
Circ Res ; 102(7): 813-22, 2008 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-18292603

RESUMEN

The Pitx2 gene regulates left-right (L/R) asymmetrical cardiac morphogenesis. Constitutive Pitx2 knock out (ko) mice die before birth and display, among other defects, right atrial isomerism, atrial and ventricular septal defects, and double outlet right ventricle. The myocardial role of the gene has not been dissected. In particular, how Pitx2 regulates the differential L/R cardiac identity program is not clear. Additionally, the relation between Pitx2 ko ventricular defects and the gene expression pattern is not understood. In this article we analyze Pitx2 myocardial function during mouse heart development. By in situ hybridization analysis we show that myocardial Pitx2 expression delineates the remodeling of the left atrioventricular canal, the inner curvature, the ventral part of the interventricular ring, and the ventral portion of the right and left ventricle. By genetic analysis using an allelic series of Pitx2 mutants, among which a myocardial specific ko (ko(myo)) we show it has a crucial role in this process. Pitx2 ko(myo) mutants survive to adulthood, when they present strong cardiac morphological and functional defects. Confocal analysis of embryonic Pitx2 ko(myo) hearts reveals delayed cardiomyocyte development in the ventricular but not in the atrial Pitx2 null areas. Conversely, selective left atrial BMP10 mRNA downregulation which normally occurs at fetal stages is not found in the Pitx2 ko(myo) mice. This is the first evidence for distinct Pitx2 action in mediating L/R atrial identity and asymmetrical ventricular remodeling.


Asunto(s)
Atrios Cardíacos/embriología , Ventrículos Cardíacos/embriología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Remodelación Ventricular/fisiología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Atrios Cardíacos/metabolismo , Cardiopatías Congénitas/patología , Ventrículos Cardíacos/metabolismo , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , ARN Mensajero/metabolismo , Nodo Sinoatrial/embriología , Nodo Sinoatrial/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Proteína del Homeodomínio PITX2
20.
Diabetes ; 57(3): 577-83, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18065520

RESUMEN

OBJECTIVE: Since vascular dysfunction is a main trait of obese subjects, in the present study we evaluated the vascular impact of resistin, a recently discovered hormone markedly increased in obesity. RESEARCH DESIGN AND METHODS: We performed our analysis on aortic and mesenteric segments from young and old C57BL/6 mice and on cultured endothelial cells. Resistin-induced vascular effect was evaluated in vitro and in vivo. Molecular analyses were performed by immunoprecipitation and Western blotting. RESULTS: Recombinant murine resistin did not induce changes in either basal vascular tone or phenylephrine-induced vascular contraction. In contrast, both in vivo and in vitro administration of resistin significantly impaired dose-dependent insulin-evoked vasodilation by reducing endothelial nitric oxide synthase (eNOS) enzymatic activity. This effect of resistin was selective for insulin vascular action, since vasodilatation induced by increasing doses of acetylcholine or nitroglycerin was not influenced by the hormone. Molecular analysis of endothelial cells further detailed resistin-induced vascular resistance by showing impairment of insulin-evoked AKT and eNOS phosphorylations after exposure to resistin. Even this latter abnormality is selective of insulin signaling since AKT/eNOS phosphorylations are normally activated during acetylcholine stimulation. More important, the resistin-induced endothelial dysfunction depends on resistin's ability to alter insulin receptor substrate (IRS)-1 tyrosine/serine phosphorylation and its consequent interaction with phosphatidylinositol 3-kinase. CONCLUSIONS: Our results demonstrate that resistin is able to induce a selective vascular insulin resistance-impairing endothelial IRS-1 signaling pathway that leads to eNOS activation and vasodilation.


Asunto(s)
Insulina/farmacología , Resistina/farmacología , Vasodilatación/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Envejecimiento , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Insulina/sangre , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Resistina/sangre , Resistina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...