Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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.

6.
EMBO Mol Med ; 7(7): 904-17, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25882071

RESUMEN

Although PI3Kγ has been extensively investigated in inflammatory and cardiovascular diseases, the exploration of its functions in the brain is just at dawning. It is known that PI3Kγ is present in neurons and that the lack of PI3Kγ in mice leads to impaired synaptic plasticity, suggestive of a role in behavioral flexibility. Several neuropsychiatric disorders, such as attention-deficit/hyperactivity disorder (ADHD), involve an impairment of behavioral flexibility. Here, we found a previously unreported expression of PI3Kγ throughout the noradrenergic neurons of the locus coeruleus (LC) in the brainstem, serving as a mechanism that regulates its activity of control on attention, locomotion and sociality. In particular, we show an unprecedented phenotype of PI3Kγ KO mice resembling ADHD symptoms. PI3Kγ KO mice exhibit deficits in the attentive and mnemonic domains, typical hyperactivity, as well as social dysfunctions. Moreover, we demonstrate that the ADHD phenotype depends on a dysregulation of CREB signaling exerted by a kinase-independent PI3Kγ-PDE4D interaction in the noradrenergic neurons of the locus coeruleus, thus uncovering new tools for mechanistic and therapeutic research in ADHD.


Asunto(s)
Neuronas Adrenérgicas/patología , Trastorno por Déficit de Atención con Hiperactividad/patología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Locus Coeruleus/patología , Transducción de Señal , Animales , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Modelos Animales de Enfermedad , Locus Coeruleus/fisiopatología , Ratones , Ratones Noqueados , Unión Proteica
7.
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
8.
Sci Signal ; 7(352): ra110, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25406378

RESUMEN

Obesity is defined as an abnormal increase in white adipose tissue and has become a major medical burden worldwide. Signals from the brain control not only appetite but also energy expenditure, both of which contribute to body weight. We showed that genetic or pharmacological inhibition of two phosphatidylinositol 3-kinases (PI3Kß and PI3Kγ) in mice reduced fat mass by promoting increased energy expenditure. This effect was accompanied by stimulation of lipolysis and the acquisition of the energy-burning characteristics of brown adipocytes by white adipocytes, a process referred to as "browning." The browning of the white adipocytes involved increased norepinephrine release from the sympathetic nervous system. We found that PI3Kß and PI3Kγ together promoted a negative feedback loop downstream of the melanocortin 4 receptor in the central nervous system, which controls appetite and energy expenditure in the periphery. Analysis of mice with drug-induced sympathetic denervation suggested that these kinases controlled the sympathetic drive in the brain. Administration of inhibitors of both PI3Kß and PI3Kγ to mice by intracerebroventricular delivery induced a 10% reduction in fat mass as quickly as 10 days. These results suggest that combined inhibition of PI3Kß and PI3Kγ might represent a promising treatment for obesity.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Obesidad/enzimología , Obesidad/fisiopatología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Sistema Nervioso Simpático/fisiología , alfa-MSH/metabolismo , Células 3T3 , Adipocitos Blancos/metabolismo , Tejido Adiposo/crecimiento & desarrollo , Animales , Western Blotting , AMP Cíclico/metabolismo , Metabolismo Energético/fisiología , Retroalimentación Fisiológica/fisiología , Técnica del Anticuerpo Fluorescente , Técnicas de Sustitución del Gen , Hipotálamo/anatomía & histología , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Lipólisis/efectos de los fármacos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA