Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 133(11): 944-958, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37869877

RESUMEN

BACKGROUND: ß1AR (beta-1 adrenergic receptor) and ß2AR (beta-2 adrenergic receptor)-mediated cyclic adenosine monophosphate signaling has distinct effects on cardiac function and heart failure progression. However, the mechanism regulating spatial localization and functional compartmentation of cardiac ß-ARs remains elusive. Emerging evidence suggests that microtubule-dependent trafficking of mRNP (messenger ribonucleoprotein) and localized protein translation modulates protein compartmentation in cardiomyocytes. We hypothesized that ß-AR compartmentation in cardiomyocytes is accomplished by selective trafficking of its mRNAs and localized translation. METHODS: The localization pattern of ß-AR mRNA was investigated using single molecule fluorescence in situ hybridization and subcellular nanobiopsy in rat cardiomyocytes. The role of microtubule on ß-AR mRNA localization was studied using vinblastine, and its effect on receptor localization and function was evaluated with immunofluorescent and high-throughput Förster resonance energy transfer microscopy. An mRNA protein co-detection assay identified plausible ß-AR translation sites in cardiomyocytes. The mechanism by which ß-AR mRNA is redistributed post-heart failure was elucidated by single molecule fluorescence in situ hybridization, nanobiopsy, and high-throughput Förster resonance energy transfer microscopy on 16 weeks post-myocardial infarction and detubulated cardiomyocytes. RESULTS: ß1AR and ß2AR mRNAs show differential localization in cardiomyocytes, with ß1AR found in the perinuclear region and ß2AR showing diffuse distribution throughout the cell. Disruption of microtubules induces a shift of ß2AR transcripts toward the perinuclear region. The close proximity between ß2AR transcripts and translated proteins suggests that the translation process occurs in specialized, precisely defined cellular compartments. Redistribution of ß2AR transcripts is microtubule-dependent, as microtubule depolymerization markedly reduces the number of functional receptors on the membrane. In failing hearts, both ß1AR and ß2AR mRNAs are redistributed toward the cell periphery, similar to what is seen in cardiomyocytes undergoing drug-induced detubulation. This suggests that t-tubule remodeling contributes to ß-AR mRNA redistribution and impaired ß2AR function in failing hearts. CONCLUSIONS: Asymmetrical microtubule-dependent trafficking dictates differential ß1AR and ß2AR localization in healthy cardiomyocyte microtubules, underlying the distinctive compartmentation of the 2 ß-ARs on the plasma membrane. The localization pattern is altered post-myocardial infarction, resulting from transverse tubule remodeling, leading to distorted ß2AR-mediated cyclic adenosine monophosphate signaling.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Ratas , Animales , Hibridación Fluorescente in Situ , Insuficiencia Cardíaca/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , AMP Cíclico/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Microtúbulos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología
2.
Am J Respir Cell Mol Biol ; 68(1): 103-115, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36264759

RESUMEN

Mitochondrial fission and a metabolic switch from oxidative phosphorylation to glycolysis are key features of vascular pathology in pulmonary arterial hypertension (PAH) and are associated with exuberant endothelial proliferation and apoptosis. The underlying mechanisms are poorly understood. We describe the contribution of two intracellular chloride channel proteins, CLIC1 and CLIC4, both highly expressed in PAH and cancer, to mitochondrial dysfunction and energy metabolism in PAH endothelium. Pathological overexpression of CLIC proteins induces mitochondrial fragmentation, inhibits mitochondrial cristae formation, and induces metabolic shift toward glycolysis in human pulmonary artery endothelial cells, consistent with changes observed in patient-derived cells. Interactions of CLIC proteins with structural components of the inner mitochondrial membrane offer mechanistic insights. Endothelial CLIC4 excision and mitofusin 2 supplementation have protective effects in human PAH cells and preclinical PAH. This study is the first to demonstrate the key role of endothelial intracellular chloride channels in the regulation of mitochondrial structure, biogenesis, and metabolic reprogramming in expression of the PAH phenotype.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Humanos , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Células Endoteliales/metabolismo , Hipertensión Pulmonar Primaria Familiar/metabolismo , Arteria Pulmonar/patología , Endotelio/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo
3.
Circ Res ; 124(1): 52-65, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582444

RESUMEN

RATIONALE: Increased expression of CLIC4 (chloride intracellular channel 4) is a feature of endothelial dysfunction in pulmonary arterial hypertension, but its role in disease pathology is not fully understood. OBJECTIVE: To identify CLIC4 effectors and evaluate strategies targeting CLIC4 signaling in pulmonary hypertension. METHODS AND RESULTS: Proteomic analysis of CLIC4-interacting proteins in human pulmonary artery endothelial cells identified regulators of endosomal trafficking, including Arf6 (ADP ribosylation factor 6) GTPase activating proteins and clathrin, while CLIC4 overexpression affected protein regulators of vesicular trafficking, lysosomal function, and inflammation. CLIC4 reduced BMPRII (bone morphogenetic protein receptor II) expression and signaling as a result of Arf6-mediated reduction in gyrating clathrin and increased lysosomal targeting of the receptor. BMPRII expression was restored by Arf6 siRNA, Arf inhibitor Sec7 inhibitor H3 (SecinH3), and inhibitors of clathrin-mediated endocytosis but was unaffected by chloride channel inhibitor, indanyloxyacetic acid 94 or Arf1 siRNA. The effects of CLIC4 on NF-κB (nuclear factor-kappa B), HIF (hypoxia-inducible factor), and angiogenic response were prevented by Arf6 siRNA and SecinH3. Sugen/hypoxia mice and monocrotaline rats showed elevated expression of CLIC4, activation of Arf6 and NF-κB, and reduced expression of BMPRII in the lung. These changes were established early during disease development. Lung endothelium-targeted delivery of CLIC4 siRNA or treatment with SecinH3 attenuated the disease, reduced CLIC4/Arf activation, and restored BMPRII expression in the lung. Endothelial colony-forming cells from idiopathic pulmonary hypertensive patients showed upregulation of CLIC4 expression and Arf6 activity, suggesting potential importance of this pathway in the human condition. CONCLUSIONS: Arf6 is a novel effector of CLIC4 and a new therapeutic target in pulmonary hypertension.


Asunto(s)
Factores de Ribosilacion-ADP/antagonistas & inhibidores , Antihipertensivos/farmacología , Canales de Cloruro/metabolismo , Células Endoteliales/efectos de los fármacos , Hipertensión Pulmonar/prevención & control , Proteínas Mitocondriales/metabolismo , Arteria Pulmonar/efectos de los fármacos , Tratamiento con ARN de Interferencia , Triazoles/farmacología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Cultivadas , Canales de Cloruro/genética , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Terapia Molecular Dirigida , Monocrotalina , Proteómica/métodos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal
4.
Arterioscler Thromb Vasc Biol ; 36(10): 2078-87, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27470511

RESUMEN

OBJECTIVE: Inflammation and dysregulated angiogenesis are features of endothelial dysfunction in pulmonary hypertension. Neutrophil extracellular traps (NETs), produced by dying neutrophils, contribute to pathogenesis of numerous vascular disorders but their role in pulmonary hypertension has not been studied. We sought evidence of (NETs) formation in pulmonary hypertension and investigated the effect of NETs on endothelial function. APPROACH AND RESULTS: Plasma and lung tissues of patients with pulmonary hypertension were analyzed for NET markers. The effects of NETs on endothelial function were studied in vitro and in vivo. Patients with chronic thromboembolic pulmonary hypertension and idiopathic pulmonary hypertension showed elevated plasma levels of DNA, neutrophil elastase, and myeloperoxidase. NET-forming neutrophils and extensive areas of NETosis were found in the occlusive plexiform lesions and vascularized intrapulmonary thrombi. NETs induced nuclear factor κB-dependent endothelial angiogenesis in vitro and increased vascularization of matrigel plugs in vivo. Angiogenic responses were associated with increased release of matrix metalloproteinase-9, heparin-binding epidermal growth factor-like growth factor, latency-associated peptide of the transforming growth factor ß1, and urokinase-type plasminogen activator, accompanied by increased endothelial permeability and cell motility. NETs-induced responses depended on myeloperoxidase/H2O2-dependent activation of Toll-like receptor 4/nuclear factor κB signaling. NETs stimulated the release of endothelin-1 in HPAECs (human pulmonary artery endothelial cells) and stimulated pulmonary smooth muscle cell proliferation in vitro. CONCLUSIONS: We are the first to implicate NETs in angiogenesis and provide a functional link between NETs and inflammatory angiogenesis in vitro and in vivo. We demonstrate the potential pathological relevance of this in 2 diseases of disordered vascular homeostasis, pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension.


Asunto(s)
Células Endoteliales/metabolismo , Trampas Extracelulares/metabolismo , Hipertensión Pulmonar/metabolismo , Neovascularización Patológica , Neutrófilos/metabolismo , Arteria Pulmonar/metabolismo , Animales , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Técnicas de Cocultivo , Células Endoteliales/patología , Humanos , Peróxido de Hidrógeno/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Masculino , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Neutrófilos/patología , Peroxidasa/metabolismo , Arteria Pulmonar/patología , Interferencia de ARN , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Transfección
5.
J Biomed Sci ; 23: 4, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26786759

RESUMEN

BACKGROUND: Increased circulating levels of endoglin(+) endothelial microparticles (EMPs) have been identified in several cardiovascular disorders, related to severity. Endoglin is an auxilary receptor for transforming growth factor ß (TGF-ß) important in the regulation of vascular structure. RESULTS: We quantified the number of microparticles in plasma of six patients with chronic thromboembolic pulmonary hypertension (CTEPH) and age- and sex-matched pulmonary embolic (PE) and healthy controls and investigated the role of microparticle endoglin in the regulation of pulmonary endothelial function in vitro. Results show significantly increased levels of endoglin(+) EMPs in CTEPH plasma, compared to healthy and disease controls. Co-culture of human pulmonary endothelial cells with CTEPH microparticles increased intracellular levels of endoglin and enhanced TGF-ß-induced angiogenesis and Smad1,5,8 phosphorylation in cells, without affecting BMPRII expression. In an in vitro model, we generated endothelium-derived MPs with enforced membrane localization of endoglin. Co-culture of these MPs with endothelial cells increased cellular endoglin content, improved cell survival and stimulated angiogenesis in a manner similar to the effects induced by overexpressed protein. CONCLUSIONS: Increased generation of endoglin(+) EMPs in CTEPH is likely to represent a protective mechanism supporting endothelial cell survival and angiogenesis, set to counteract the effects of vascular occlusion and endothelial damage.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Endotelio Vascular/metabolismo , Hipertensión Pulmonar/metabolismo , Neovascularización Patológica/metabolismo , Embolia Pulmonar/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Micropartículas Derivadas de Células/patología , Endotelio Vascular/patología , Femenino , Humanos , Hipertensión Pulmonar/patología , Masculino , Persona de Mediana Edad , Neovascularización Patológica/patología , Embolia Pulmonar/patología
6.
Circulation ; 129(17): 1770-80, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24503951

RESUMEN

BACKGROUND: Chloride intracellular channel 4 (CLIC4) is highly expressed in the endothelium of remodeled pulmonary vessels and plexiform lesions of patients with pulmonary arterial hypertension. CLIC4 regulates vasculogenesis through endothelial tube formation. Aberrant CLIC4 expression may contribute to the vascular pathology of pulmonary arterial hypertension. METHODS AND RESULTS: CLIC4 protein expression was increased in plasma and blood-derived endothelial cells from patients with idiopathic pulmonary arterial hypertension and in the pulmonary vascular endothelium of 3 rat models of pulmonary hypertension. CLIC4 gene deletion markedly attenuated the development of chronic hypoxia-induced pulmonary hypertension in mice. Adenoviral overexpression of CLIC4 in cultured human pulmonary artery endothelial cells compromised pulmonary endothelial barrier function and enhanced their survival and angiogenic capacity, whereas CLIC4 shRNA had an inhibitory effect. Similarly, inhibition of CLIC4 expression in blood-derived endothelial cells from patients with idiopathic pulmonary arterial hypertension attenuated the abnormal angiogenic behavior that characterizes these cells. The mechanism of CLIC4 effects involves p65-mediated activation of nuclear factor-κB, followed by stabilization of hypoxia-inducible factor-1α and increased downstream production of vascular endothelial growth factor and endothelin-1. CONCLUSION: Increased CLIC4 expression is an early manifestation and mediator of endothelial dysfunction in pulmonary hypertension.


Asunto(s)
Canales de Cloruro/fisiología , Endotelio Vascular/fisiopatología , Hipertensión Pulmonar/fisiopatología , Proteínas Mitocondriales/fisiología , Arteria Pulmonar/fisiopatología , Animales , Células Cultivadas , Canales de Cloruro/genética , Endotelio Vascular/citología , Hipertensión Pulmonar Primaria Familiar , Humanos , Hipertensión Pulmonar/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas Mitocondriales/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiopatología , Arteria Pulmonar/citología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción ReIA/fisiología
7.
Biochem J ; 462(1): 103-12, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24895913

RESUMEN

The NOS (nitric oxide synthase) inhibitor ADMA (asymmetric dimethylarginine) contributes to the pathogenesis of pulmonary hypertension. Reduced levels of the enzymes metabolizing ADMA, dimethylarginine dimethylaminohydrolases (DDAH1 and DDAH2) and increased levels of miR-21 are linked to disease pathology, but the mechanisms are not understood. In the present study we assessed the potential role of miR-21 in the regulation of hypoxia-induced changes in ADMA metabolism in vitro and in vivo. Hypoxia inhibited DDAH1 and DDAH2 expression and increased ADMA levels in cultured human pulmonary endothelial cells. In contrast, in human pulmonary smooth muscle cells, only DDAH2 was reduced whereas ADMA levels remained unchanged. Endothelium-specific down-regulation of DDAH1 by miR-21 in hypoxia induced endothelial dysfunction and was prevented by overexpression of DDAH1 and miR-21 blockade. DDAH1, but not DDAH2, mRNA levels were reduced, whereas miR-21 levels were elevated in lung tissues from patients with pulmonary arterial hypertension and mice with pulmonary hypertension exposed to 2 weeks of hypoxia. Hypoxic mice treated with miR-21 inhibitors and DDAH1 transgenic mice showed elevated lung DDAH1, increased cGMP levels and attenuated pulmonary hypertension. Regulation of DDAH1 by miR-21 plays a role in the development of hypoxia-induced pulmonary hypertension and may be of broader significance in pulmonary hypertension.


Asunto(s)
Amidohidrolasas/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , MicroARNs/fisiología , Animales , Arginina/análogos & derivados , Células Cultivadas , GMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Hipertensión Pulmonar Primaria Familiar , Humanos , Hipertensión Pulmonar/genética , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo
8.
Cell Tissue Res ; 355(3): 675-85, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24599334

RESUMEN

Pulmonary endothelial permeability is an important determinant of vascular adaptation to changes in oxygen tension, blood pressure, levels of growth factors or inflammatory cytokines. The Ras homologous (Rho) family of guanosine triphosphate phosphatases (Rho GTPases), key regulators of the actin cytoskeleton, regulate endothelial barrier function in response to a variety of environmental factors and signalling agents via the reorganization of the actin cytoskeleton, changes in receptor trafficking or the phosphorylation of junctional proteins. This review provides a brief summary of recent knowledge on Rho-GTPase-mediated effects on pulmonary endothelial barrier function and focuses in particular on their role in pulmonary vascular disorders, including pulmonary hypertension, chronic obstructive pulmonary disease, acute lung injury and acute respiratory distress syndrome.


Asunto(s)
Actinas/fisiología , Permeabilidad Capilar/fisiología , Endotelio Vascular/fisiología , Pulmón/fisiología , Proteínas de Unión al GTP rho/fisiología , Actinas/metabolismo , Animales , Endotelio Vascular/metabolismo , Humanos , Pulmón/citología , Pulmón/enzimología , Proteínas de Unión al GTP rho/metabolismo
9.
Circ Res ; 110(11): 1423-34, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22539766

RESUMEN

RATIONALE: RhoA and Rho kinase contribute to pulmonary vasoconstriction and vascular remodeling in pulmonary hypertension. RhoB, a protein homologous to RhoA and activated by hypoxia, regulates neoplastic growth and vasoconstriction but its role in the regulation of pulmonary vascular function is not known. OBJECTIVE: To determine the role of RhoB in pulmonary endothelial and smooth muscle cell responses to hypoxia and in pulmonary vascular remodeling in chronic hypoxia-induced pulmonary hypertension. METHODS AND RESULTS: Hypoxia increased expression and activity of RhoB in human pulmonary artery endothelial and smooth muscle cells, coincidental with activation of RhoA. Hypoxia or adenoviral overexpression of constitutively activated RhoB increased actomyosin contractility, induced endothelial permeability, and promoted cell growth; dominant negative RhoB or manumycin, a farnesyltransferase inhibitor that targets the vascular function of RhoB, inhibited the effects of hypoxia. Coordinated activation of RhoA and RhoB maximized the hypoxia-induced stress fiber formation caused by RhoB/mammalian homolog of Drosophila diaphanous-induced actin polymerization and RhoA/Rho kinase-induced phosphorylation of myosin light chain on Ser19. Notably, RhoB was specifically required for hypoxia-induced factor-1α stabilization and for hypoxia- and platelet-derived growth factor-induced cell proliferation and migration. RhoB deficiency in mice markedly attenuated development of chronic hypoxia-induced pulmonary hypertension, despite compensatory expression of RhoA in the lung. CONCLUSIONS: RhoB mediates adaptational changes to acute hypoxia in the vasculature, but its continual activation by chronic hypoxia can accentuate vascular remodeling to promote development of pulmonary hypertension. RhoB is a potential target for novel approaches (eg, farnesyltransferase inhibitors) aimed at regulating pulmonary vascular tone and structure.


Asunto(s)
Células Endoteliales/enzimología , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Proteína de Unión al GTP rhoB/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Animales , Permeabilidad Capilar , Hipoxia de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Hipertensión Pulmonar Primaria Familiar , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/metabolismo , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/genética , Hipoxia/enzimología , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Polienos/farmacología , Alcamidas Poliinsaturadas/farmacología , Arteria Pulmonar/enzimología , Interferencia de ARN , Serina , Fibras de Estrés/enzimología , Factores de Tiempo , Transfección , Vasoconstricción , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/deficiencia , Proteína de Unión al GTP rhoB/genética
10.
Nat Med ; 13(2): 198-203, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17273169

RESUMEN

Asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA) are endogenously produced amino acids that inhibit all three isoforms of nitric oxide synthase (NOS). ADMA accumulates in various disease states, including renal failure, diabetes and pulmonary hypertension, and its concentration in plasma is strongly predictive of premature cardiovascular disease and death. Both L-NMMA and ADMA are eliminated largely through active metabolism by dimethylarginine dimethylaminohydrolase (DDAH) and thus DDAH dysfunction may be a crucial unifying feature of increased cardiovascular risk. However, despite considerable interest in this pathway and in the role of ADMA as a cardiovascular risk factor, there is little evidence to support a causal role of ADMA in pathophysiology. Here we reveal the structure of human DDAH-1 and probe the function of DDAH-1 both by deleting the DDAH1 gene in mice and by using DDAH-specific inhibitors which, as we demonstrate by crystallography, bind to the active site of human DDAH-1. We show that loss of DDAH-1 activity leads to accumulation of ADMA and reduction in NO signaling. This in turn causes vascular pathophysiology, including endothelial dysfunction, increased systemic vascular resistance and elevated systemic and pulmonary blood pressure. Our results also suggest that DDAH inhibition could be harnessed therapeutically to reduce the vascular collapse associated with sepsis.


Asunto(s)
Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Arginina/análogos & derivados , Fenómenos Fisiológicos Cardiovasculares , Homeostasis/genética , Modelos Moleculares , omega-N-Metilarginina/metabolismo , Acetilcolina/farmacología , Amidohidrolasas/antagonistas & inhibidores , Animales , Arginina/metabolismo , Presión Sanguínea/genética , Vasos Sanguíneos/efectos de los fármacos , Northern Blotting , Western Blotting , Calcimicina/farmacología , Cromatografía Líquida de Alta Presión , Cristalografía , Relación Dosis-Respuesta a Droga , Ecocardiografía , Endotelio/metabolismo , Eliminación de Gen , Humanos , Ratones , Contracción Muscular/efectos de los fármacos , Óxido Nítrico/metabolismo , Nitroprusiato/farmacología , Fenilefrina/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Resistencia Vascular/genética
11.
Am J Respir Crit Care Med ; 187(3): 294-302, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23220912

RESUMEN

RATIONALE: MicroRNAs (miRNAs or miRs) are implicated in the pathogenesis of various cardiovascular diseases, including pulmonary arterial hypertension (PAH). OBJECTIVES: We sought to measure changes in plasma levels of miRNAs in patients with PAH and relate them to the severity of the disease. METHODS: A microarray screen was performed on total plasma RNA from eight patients with PAH and eight healthy control subjects. Quantitative polymerase chain reaction confirmed reduced miR-150 concentrations and was then used to measure miR-150 levels in (1) two separate cohorts of patients with PAH, from London (n = 145) and Sheffield (n = 30), respectively; (2) circulating microvesicles and blood cells; and (3) lungs from a monocrotaline rat model. MEASUREMENTS AND MAIN RESULTS: Fifty-eight miRNAs showed differences in plasma concentration and miR-150 the largest down-regulation in PAH. Receiver-operator-characteristic analysis showed both raw and normalized plasma miR-150 levels correlated with 2-year survival (P < 0.01) in patients with PAH. Cox regression analysis confirmed miR-150 levels as a significant predictor of survival. Age, baseline cardiac index, World Health Organization functional class, 6-minute walk distance, disease duration, and red cell distribution width also predicted survival. Entering these covariates in a multivariable model verified plasma miR-150 levels as an independent predictor of survival in PAH (hazard ratio, 0.533; P = 0.010). miR-150 levels also predicted survival in a second, independent PAH cohort. miR-150 levels were significantly reduced in circulating microvesicles from patients with PAH and the lungs of the monocrotaline rat. CONCLUSIONS: Reduced circulating miR-150 levels are associated with poor survival in PAH.


Asunto(s)
Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/genética , MicroARNs/sangre , Adulto , Distribución por Edad , Animales , Biomarcadores/sangre , Estudios de Cohortes , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hipertensión Pulmonar Primaria Familiar , Femenino , Humanos , Londres , Masculino , Análisis por Micromatrices/métodos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa/métodos , Curva ROC , Ratas , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
12.
Commun Biol ; 5(1): 1192, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36344664

RESUMEN

Pulmonary arterial hypertension (PAH) is an unmet clinical need. The lack of models of human disease is a key obstacle to drug development. We present a biomimetic model of pulmonary arterial endothelial-smooth muscle cell interactions in PAH, combining natural and induced bone morphogenetic protein receptor 2 (BMPR2) dysfunction with hypoxia to induce smooth muscle activation and proliferation, which is responsive to drug treatment. BMPR2- and oxygenation-specific changes in endothelial and smooth muscle gene expression, consistent with observations made in genomic and biochemical studies of PAH, enable insights into underlying disease pathways and mechanisms of drug response. The model captures key changes in the pulmonary endothelial phenotype that are essential for the induction of SMC remodelling, including a BMPR2-SOX17-prostacyclin signalling axis and offers an easily accessible approach for researchers to study pulmonary vascular remodelling and advance drug development in PAH.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Factores de Transcripción SOXF , Humanos , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Epoprostenol/genética , Epoprostenol/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/genética , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo
13.
Int J Biochem Cell Biol ; 134: 105977, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33839307

RESUMEN

Pulmonary arterial hypertension is a rare but deadly disease with a complex pathogenesis. Recent evidence demonstrates that Krüppel-like factors, a diverse family of transcription factors, are involved in several key disease processes such as the phenotypic transition of endothelial cells and smooth muscle cells. Importantly, manipulation of certain Krüppel-like factors enables protection or attenuation against pulmonary arterial hypertension in both animal models and preliminary human studies. In this review, we discuss how Krüppel-like factors, in particular Krüppel-like factors 2, 4 and 5 contribute to the pathological phenomena seen in pulmonary arterial hypertension and how associated signaling and microRNA pathways may be suitable targets for new therapies.


Asunto(s)
Células Endoteliales/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/genética , Miocitos del Músculo Liso/patología , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/patología , Animales , Células Endoteliales/metabolismo , Humanos , Miocitos del Músculo Liso/metabolismo , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar/metabolismo , Transducción de Señal
14.
Mol Ther Nucleic Acids ; 23: 142-153, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33335799

RESUMEN

Circulating levels of endothelial miR-150 are reduced in pulmonary arterial hypertension (PAH) and act as an independent predictor of patient survival, but links between endothelial miR-150 and vascular dysfunction are not well understood. We studied the effects of endothelial miR-150 supplementation and inhibition in PAH mice and cells from patients with idiopathic PAH. The role of selected mediators of miR-150 identified by RNA sequencing was evaluated in vitro and in vivo. Endothelium-targeted miR-150 delivery prevented the disease in Sugen/hypoxia mice, while endothelial knockdown of miR-150 had adverse effects. miR-150 target genes revealed significant associations with PAH pathways, including proliferation, inflammation, and phospholipid signaling, with PTEN-like mitochondrial phosphatase (PTPMT1) most markedly altered. PTPMT1 reduced inflammation and apoptosis and improved mitochondrial function in human pulmonary endothelial cells and blood-derived endothelial colony-forming cells from idiopathic PAH. Beneficial effects of miR-150 in vitro and in vivo were linked with PTPMT1-dependent biosynthesis of mitochondrial phospholipid cardiolipin and reduced expression of pro-apoptotic, pro-inflammatory, and pro-fibrotic genes, including c-MYB, NOTCH3, transforming growth factor ß (TGF-ß), and Col1a1. In conclusion, we are the first to show that miR-150 supplementation attenuates pulmonary endothelial damage induced by vascular stresses and may be considered as a potential therapeutic strategy in PAH.

15.
Hypertension ; 77(2): 605-616, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33356404

RESUMEN

Pulmonary hypertension is a complex disorder characterized by pulmonary vascular remodeling and right ventricular hypertrophy, leading to right heart failure. The mechanisms underlying this process are not well understood. We hypothesize that the structural remodeling occurring in the cardiomyocytes of the right ventricle affects the cytosolic Ca2+ handling leading to arrhythmias. After 12 days of monocrotaline-induced pulmonary hypertension in rats, epicardial mapping showed electrical remodeling in both ventricles. In myocytes isolated from the hypertensive rats, a combination of high-speed camera and confocal line-scan documented a prolongation of Ca2+ transients along with a higher local Ca2+-release activity. These Ca2+ transients were less synchronous than in controls, likely due to disorganized transverse-axial tubular system. In fact, following pulmonary hypertension, hypertrophied right ventricular myocytes showed significantly reduced number of transverse tubules and increased number of axial tubules; however, Stimulation Emission Depletion microscopy demonstrated that the colocalization of L-type Ca2+ channels and RyR2 (ryanodine receptor 2) remained unchanged. Finally, Stimulation Emission Depletion microscopy and super-resolution scanning patch-clamp analysis uncovered a decrease in the density of active L-type Ca2+ channels in right ventricular myocytes with an elevated open probability of the T-tubule anchored channels. This may represent a general mechanism of how nanoscale structural changes at the early stage of pulmonary hypertension impact on the development of the end stage failing phenotype in the right ventricle.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Hipertensión Pulmonar/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Masculino , Monocrotalina , Ratas , Ratas Sprague-Dawley , Remodelación Vascular/fisiología
16.
Arterioscler Thromb Vasc Biol ; 29(12): 2117-24, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19778944

RESUMEN

OBJECTIVE: Asymmetrical dimethylarginine (ADMA) is a nitric oxide synthase (NOS) inhibitor and cardiovascular risk factor associated with angiogenic disorders. Enzymes metabolising ADMA, dimethylarginine dimethylaminohydrolases (DDAH) promote angiogenesis, but the mechanisms are not clear. We hypothesized that ADMA/DDAH modifies endothelial responses to vascular endothelial growth factor (VEGF) by affecting activity of Rho GTPases, regulators of actin polymerization, and focal adhesion dynamics. METHODS AND RESULTS: The effects of ADMA on VEGF-induced endothelial cell motility, focal adhesion turnover, and angiogenesis were studied in human umbilical vein endothelial cells (HUVECs) and DDAH I heterozygous knockout mice. ADMA inhibited VEGF-induced chemotaxis in vitro and angiogenesis in vitro and in vivo in an NO-dependent way. ADMA effects were prevented by overexpression of DDAH but were not associated with decreased proliferation, increased apoptosis, or changes in VEGFR-2 activity or expression. ADMA inhibited endothelial cell polarization, protrusion formation, and decreased focal adhesion dynamics, resulting from Rac1 inhibition after decrease in phosphorylation of vasodilator stimulated phosphoprotein (VASP). Constitutively active Rac1, and to a lesser extent dominant negative RhoA, abrogated ADMA effects in vitro and in vivo. CONCLUSIONS: The ADMA/DDAH pathway regulates VEGF-induced angiogenesis in an NO- and Rac1-dependent manner.


Asunto(s)
Amidohidrolasas/metabolismo , Arginina/análogos & derivados , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Amidohidrolasas/deficiencia , Amidohidrolasas/genética , Animales , Apoptosis , Arginina/metabolismo , Arginina/farmacología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Polaridad Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Adhesiones Focales/efectos de los fármacos , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neuropéptidos/metabolismo , Óxido Nítrico/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo
17.
Vasc Med ; 15(3): 205-13, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20219849

RESUMEN

Dimethylarginine dimethylaminohydrolase (DDAH) degrades asymmetric dimethylarginine (ADMA), an endogenously produced nitric oxide (NO) synthase inhibitor. In mammals, two isoforms of DDAH, DDAH1 and DDAH2, are expressed in the cardiovascular system, suggesting that ADMA concentrations are actively regulated in blood vessels, raising the possibility that cardiovascular metabolism of ADMA constitutes a novel mechanism for the regulation of NO production. The purpose of this study was to determine the role of DDAH-catalyzed asymmetric methylarginine metabolism in the regulation of vascular function. We developed adenoviral vectors for the expression of human DDAH1 and 2. Overexpression of DDAH1 or 2 in human umbilical vein endothelial cells (HUVEC) increases DDAH activity, reduces ADMA concentrations and increases NO production. Similarly, overexpression of DDAH1 or 2 in DDAH1(+/-) mice carotid vessels increases NO production and attenuates the response to phenylephrine (PE), enhances acetylcholine (ACh) relaxation and attenuates the effect of exogenously applied ADMA. Finally, overexpression of either DDAH1 or 2 completely reversed the vascular dysfunction seen in DDAH1(+/-) mice. These data indicate that basal concentrations of ADMA in blood vessels are sufficient to regulate NO production, that increases in the level of either DDAH1 or 2, improves vascular function and that overexpression of either DDAH1 or 2 is sufficient to compensate for life-long exposure to elevated ADMA. Thus, therapeutic manipulation of DDAH expression or activity may represent a novel approach to improve vascular dysfunction in various cardiovascular diseases.


Asunto(s)
Adenoviridae/genética , Amidohidrolasas/genética , Terapia Genética/métodos , Enfermedad Arterial Periférica/terapia , Amidohidrolasas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Arterias Carótidas/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Enfermedad Arterial Periférica/fisiopatología , Venas Umbilicales/citología , Vasoconstricción/fisiología
18.
Cell Death Dis ; 11(9): 729, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908129

RESUMEN

Anti-double stranded DNA antibodies (anti-dsDNA) are a hallmark of SLE but their role in disease pathogenesis is not fully resolved. Anti-dsDNA in serum are highly heterogeneous therefore in this study, we aimed to dissect the functional specificities of anti-dsDNA using a panel of human monoclonal antibodies (humAbs) generated from patients with active lupus nephritis. A total of 46 ANA reactive humAbs were isolated and divided into four broad classes based on their reactivity to histones, DNA and Crithidia. Functional analysis indicated that one subclass of antibodies bound strongly to decondensed DNA areas in neutrophil extracellular traps (NETs) and protected NETs from nuclease digestion, similar to the sera from active SLE patients. In addition, these anti-dsDNA antibodies could stimulate type I interferon responses in mononuclear phagocytic cells, or NF-kB activity in endothelial cells, by uptake of NETs-anti-NETs immune complexes and subsequently trigging inflammatory responses in an Fc-gamma receptor (Fcg-R)-dependant manner. Together our data suggest that only a subset of anti-dsDNA antibodies is capable to amplify inflammatory responses by deposit in the nephritic kidney in vivo, protecting NETs digestion as well as uptake of NETs immune complexes into Fcg-R-expressing cells in vitro.


Asunto(s)
Autoanticuerpos/metabolismo , Inflamación/genética , Lupus Eritematoso Sistémico/complicaciones , Animales , Muerte Celular , Modelos Animales de Enfermedad , Humanos , Lupus Eritematoso Sistémico/patología , Ratones , Transfección
19.
Nat Commun ; 11(1): 1185, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132543

RESUMEN

Pulmonary arterial hypertension (PAH) is a severe disorder of lung vasculature that causes right heart failure. Homoeostatic effects of flow-activated transcription factor Krüppel-like factor 2 (KLF2) are compromised in PAH. Here, we show that KLF2-induced exosomal microRNAs, miR-181a-5p and miR-324-5p act together to attenuate pulmonary vascular remodelling and that their actions are mediated by Notch4 and ETS1 and other key regulators of vascular homoeostasis. Expressions of KLF2, miR-181a-5p and miR-324-5p are reduced, while levels of their target genes are elevated in pre-clinical PAH, idiopathic PAH and heritable PAH with missense p.H288Y KLF2 mutation. Therapeutic supplementation of miR-181a-5p and miR-324-5p reduces proliferative and angiogenic responses in patient-derived cells and attenuates disease progression in PAH mice. This study shows that reduced KLF2 signalling is a common feature of human PAH and highlights the potential therapeutic role of KLF2-regulated exosomal miRNAs in PAH and other diseases associated with vascular remodelling.


Asunto(s)
Terapia Genética/métodos , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/uso terapéutico , Hipertensión Arterial Pulmonar/terapia , Adulto , Anciano , Animales , Proliferación Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales , Exosomas/genética , Exosomas/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Pulmón/irrigación sanguínea , Pulmón/citología , Pulmón/patología , Masculino , Ratones , MicroARNs/metabolismo , Persona de Mediana Edad , Mutación Missense , Cultivo Primario de Células , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/citología , Arteria Pulmonar/patología , Transducción de Señal/genética , Remodelación Vascular/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA