Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.080
Filtrar
1.
Cardiovasc Diabetol ; 23(1): 138, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664801

RESUMEN

BACKGROUND: Neutral cholesterol ester hydrolase 1 (NCEH1) plays a critical role in the regulation of cholesterol ester metabolism. Deficiency of NCHE1 accelerated atherosclerotic lesion formation in mice. Nonetheless, the role of NCEH1 in endothelial dysfunction associated with diabetes has not been explored. The present study sought to investigate whether NCEH1 improved endothelial function in diabetes, and the underlying mechanisms were explored. METHODS: The expression and activity of NCEH1 were determined in obese mice with high-fat diet (HFD) feeding, high glucose (HG)-induced mouse aortae or primary endothelial cells (ECs). Endothelium-dependent relaxation (EDR) in aortae response to acetylcholine (Ach) was measured. RESULTS: Results showed that the expression and activity of NCEH1 were lower in HFD-induced mouse aortae, HG-exposed mouse aortae ex vivo, and HG-incubated primary ECs. HG exposure reduced EDR in mouse aortae, which was exaggerated by endothelial-specific deficiency of NCEH1, whereas NCEH1 overexpression restored the impaired EDR. Similar results were observed in HFD mice. Mechanically, NCEH1 ameliorated the disrupted EDR by dissociating endothelial nitric oxide synthase (eNOS) from caveolin-1 (Cav-1), leading to eNOS activation and nitric oxide (NO) release. Moreover, interaction of NCEH1 with the E3 ubiquitin-protein ligase ZNRF1 led to the degradation of Cav-1 through the ubiquitination pathway. Silencing Cav-1 and upregulating ZNRF1 were sufficient to improve EDR of diabetic aortas, while overexpression of Cav-1 and downregulation of ZNRF1 abolished the effects of NCEH1 on endothelial function in diabetes. Thus, NCEH1 preserves endothelial function through increasing NO bioavailability secondary to the disruption of the Cav-1/eNOS complex in the endothelium of diabetic mice, depending on ZNRF1-induced ubiquitination of Cav-1. CONCLUSIONS: NCEH1 may be a promising candidate for the prevention and treatment of vascular complications of diabetes.


Asunto(s)
Caveolina 1 , Dieta Alta en Grasa , Células Endoteliales , Endotelio Vascular , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III , Vasodilatación , Animales , Masculino , Ratones , Aorta/enzimología , Aorta/fisiopatología , Aorta/metabolismo , Aorta/efectos de los fármacos , Aorta/patología , Caveolina 1/metabolismo , Caveolina 1/deficiencia , Caveolina 1/genética , Células Cultivadas , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/fisiopatología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/fisiopatología , Endotelio Vascular/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/efectos de los fármacos , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/enzimología , Obesidad/fisiopatología , Obesidad/metabolismo , Transducción de Señal , Esterol Esterasa/metabolismo , Esterol Esterasa/genética , Ubiquitinación , Vasodilatación/efectos de los fármacos
2.
Signal Transduct Target Ther ; 7(1): 6, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34992208

RESUMEN

Revascularization and angiogenesis, as substrates of sustained collateral circulation, play a crucial role in determining the severity and clinical outcome of acute ischemic stroke (AIS) due to large vessel occlusion (LVO). Developing an adjunct biomarker to help identify and monitor collateral status would aid stroke diagnosis and prognosis. To screen the potential biomarkers, proteomic analysis was performed in this study to identify those distinct plasma protein profiles in AIS due to LVO with different collateral status. Interestingly, we found that levels of Plasma Factor VII Activating Protease (FSAP) significantly increased in those AIS patients with poor collaterals, and were correlated with worse neurological outcome. Furtherly, both in vitro and in vivo models of ischemic stroke were used to explore pathological mechanisms of FSAP in endothelial dysfunction. We demonstrated that the FSAP inhibitor, high-molecular-weight hyaluronan (HMW-HA), enhanced the pro-angiogenic vascular factors, improved the integrity of brain blood barrier, and promoted newly formed cerebral microvessels in the ischemic penumbra, consequently improving neurological function. To elucidate the pathways that might contribute to revascularization during LVO, we applied transcriptomic analysis via unbiased RNA sequencing and showed that Wnt signaling was highly involved in FSAP mediated endothelial dysfunction. Notably, inhibition of Wnt5a largely reversed the protective effects from HMW-HA treatment, implying that FSAP might aggravate endothelial dysfunction and neurological deficits by regulating Wnt5a signaling. Therefore, FSAP may represent a potential biomarker for collateral status after LVO and a promising therapeutic target to be explored in the treatment of stroke.


Asunto(s)
Trastornos Cerebrovasculares/enzimología , Endotelio Vascular/enzimología , Accidente Cerebrovascular Isquémico/enzimología , Serina Endopeptidasas/metabolismo , Animales , Humanos , Ratones
3.
Cardiovasc Res ; 118(1): 254-266, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33483748

RESUMEN

AIMS: Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting joints and blood vessels. Despite low levels of low-density lipoprotein cholesterol (LDL-C), RA patients exhibit endothelial dysfunction and are at increased risk of death from cardiovascular complications, but the molecular mechanism of action is unknown. We aimed in the present study to identify the molecular mechanism of endothelial dysfunction in a mouse model of RA and in patients with RA. METHODS AND RESULTS: Endothelium-dependent relaxations to acetylcholine were reduced in aortae of two tumour necrosis factor alpha (TNFα) transgenic mouse lines with either mild (Tg3647) or severe (Tg197) forms of RA in a time- and severity-dependent fashion as assessed by organ chamber myograph. In Tg197, TNFα plasma levels were associated with severe endothelial dysfunction. LOX-1 receptor was markedly up-regulated leading to increased vascular oxLDL uptake and NFκB-mediated enhanced Arg2 expression via direct binding to its promoter resulting in reduced NO bioavailability and vascular cGMP levels as shown by ELISA and chromatin immunoprecipitation. Anti-TNFα treatment with infliximab normalized endothelial function together with LOX-1 and Arg2 serum levels in mice. In RA patients, soluble LOX-1 serum levels were also markedly increased and closely related to serum levels of C-reactive protein. Similarly, ARG2 serum levels were increased. Similarly, anti-TNFα treatment restored LOX-1 and ARG2 serum levels in RA patients. CONCLUSIONS: Increased TNFα levels not only contribute to RA, but also to endothelial dysfunction by increasing vascular oxLDL content and activation of the LOX-1/NFκB/Arg2 pathway leading to reduced NO bioavailability and decreased cGMP levels. Anti-TNFα treatment improved both articular symptoms and endothelial function by reducing LOX-1, vascular oxLDL, and Arg2 levels.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Arginasa/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Receptores Depuradores de Clase E/metabolismo , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo , Vasodilatación/efectos de los fármacos , Adulto , Animales , Animales Modificados Genéticamente , Aorta Torácica/enzimología , Aorta Torácica/inmunología , Aorta Torácica/fisiopatología , Arginasa/genética , Artritis Reumatoide/enzimología , Artritis Reumatoide/inmunología , Artritis Reumatoide/fisiopatología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/inmunología , Endotelio Vascular/enzimología , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Femenino , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Persona de Mediana Edad , FN-kappa B/metabolismo , Receptores Depuradores de Clase E/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética
4.
FASEB J ; 36(1): e22110, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34918393

RESUMEN

Dengue virus (DENV) is a cause of vascular endothelial dysfunction and vascular leakage, which are characterized as hallmarks of dengue hemorrhagic fever or dengue shock syndrome, which become a severe global health emergency with substantial morbidity and mortality. Currently, there are still no promising therapeutics to alleviate the dengue-associated vascular hemorrhage in a clinical setting. In the present study, we first observed that heme oxygenase-1 (HO-1) expression level was highly suppressed in severe DENV-infected patients. In contrast, the overexpression of HO-1 could attenuate DENV-induced pathogenesis, including plasma leakage and thrombocytopenia, in an AG129 mouse model. Our data indicate that overexpression of HO-1 or its metabolite biliverdin can maintain endothelial integrity upon DENV infection in vitro and in vivo. We further characterized the positive regulatory effect of HO-1 on the endothelial adhesion factor vascular endothelial-cadherin to decrease DENV-induced endothelial hyperpermeability. Subsequently, we confirmed that two medicinal plant-derived compounds, andrographolide, and celastrol, widely used as a nutritional or medicinal supplement are useful to attenuate DENV-induced plasma leakage through induction of the HO-1 expression in DENV-infected AG129 mice. In conclusion, our findings reveal that induction of the HO-1 signal pathway is a promising option for the treatment of DENV-induced vascular pathologies.


Asunto(s)
Permeabilidad Capilar , Virus del Dengue/metabolismo , Endotelio Vascular/enzimología , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Dengue Grave/enzimología , Animales , Línea Celular , Virus del Dengue/genética , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Mutantes , Dengue Grave/genética
5.
J Cardiovasc Pharmacol ; 78(Suppl 6): S63-S77, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34840264

RESUMEN

ABSTRACT: SIRT1, a member of the sirtuin family of longevity regulators, possesses potent activities preventing vascular aging. The expression and function of SIRT1 in endothelial cells are downregulated with age, in turn causing early vascular aging and predisposing various vascular abnormalities. Overexpression of SIRT1 in the vascular endothelium prevents aging-associated endothelial dysfunction and senescence, thus the development of hypertension and atherosclerosis. Numerous efforts have been directed to increase SIRT1 signaling as a potential strategy for different aging-associated diseases. However, the complex mechanisms underlying the regulation of SIRT1 have posed a significant challenge toward the design of specific and effective therapeutics. This review aimed to provide a summary on the regulation and function of SIRT1 in the vascular endothelium and to discuss the different approaches targeting this molecule for the prevention and treatment of age-related cardiovascular and cerebrovascular diseases.


Asunto(s)
Senescencia Celular , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Sirtuina 1/metabolismo , Enfermedades Vasculares/enzimología , Animales , Senescencia Celular/efectos de los fármacos , Suplementos Dietéticos , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Regulación Enzimológica de la Expresión Génica , Estilo de Vida Saludable , Humanos , Terapia Molecular Dirigida , Rejuvenecimiento , Conducta de Reducción del Riesgo , Transducción de Señal , Sirtuina 1/genética , Enfermedades Vasculares/patología , Enfermedades Vasculares/fisiopatología , Enfermedades Vasculares/prevención & control
6.
Life Sci ; 285: 119966, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34543641

RESUMEN

AIMS: Androgen deprivation therapy is a common prostate cancer treatment which causes men to have castrate levels of testosterone. Unfortunately, most testosterone deficient patients will suffer severe erectile dysfunction (ED) and have no effective ED treatment options. Testosterone deficiency causes endothelial dysfunction and impairs penile vasodilation necessary to maintain an erection. Recent evidence demonstrates testosterone activates androgen receptors (AR) and generates nitric oxide (NO) through the Akt-endothelial NO synthase (eNOS) pathway; however, it remains unknown how castration impacts this signaling pathway. MATERIALS AND METHODS: In this study, we used a surgically castrated rat model to determine how castration impacts ex vivo internal pudendal artery (IPA) and penile relaxation through the Akt-eNOS pathway. KEY FINDINGS: Unlike systemic vasculature, castration causes significant IPA and penis endothelial dysfunction associated with a 50% AR reduction. Though testosterone and acetylcholine (ACh) both phosphorylate Akt and eNOS, castration did not affect testosterone-mediated IPA and penile Akt or eNOS phosphorylation. Surprisingly, castration increases ACh-mediated Akt and eNOS phosphorylation but reduces the eNOS dimer to monomer ratio. Akt inhibition using 10DEBC preserves IPA eNOS dimers. Functionally, 10DEBC reverses castration induced ex vivo IPA and penile endothelial dysfunction. SIGNIFICANCE: These data demonstrate how castration uncouples eNOS and provide a novel strategy for improving endothelial-dependent relaxation necessary for an erection. Further studies are needed to determine if Akt inhibition may treat or even prevent ED in testosterone deficient prostate cancer survivors.


Asunto(s)
Castración/efectos adversos , Endotelio Vascular/enzimología , Arteria Ilíaca/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Pene/irrigación sanguínea , Proteínas Proto-Oncogénicas c-akt/metabolismo , Testosterona/deficiencia , Vasodilatación/fisiología , Animales , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Arteria Ilíaca/efectos de los fármacos , Arteria Ilíaca/fisiopatología , Masculino , Modelos Animales , Erección Peniana/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
7.
Sci Rep ; 11(1): 17764, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34493753

RESUMEN

Endothelial-mesenchymal transition (EndMT) is a form of endothelial dysfunction wherein endothelial cells acquire a mesenchymal phenotype and lose endothelial functions, which contributes to the pathogenesis of intimal hyperplasia and atherosclerosis. The mitogen activated protein kinase 7 (MAPK7) inhibits EndMT and decreases the expression of the histone methyltransferase Enhancer-of-Zeste homologue 2 (EZH2), thereby maintaining endothelial quiescence. EZH2 is the catalytic subunit of the Polycomb Repressive Complex 2 that methylates lysine 27 on histone 3 (H3K27me3). It is elusive how the crosstalk between MAPK7 and EZH2 is regulated in the endothelium and if the balance between MAPK7 and EZH2 is disturbed in vascular disease. In human coronary artery disease, we assessed the expression levels of MAPK7 and EZH2 and found that with increasing intima/media thickness ratio, MAPK7 expression decreased, whereas EZH2 expression increased. In vitro, MAPK7 activation decreased EZH2 expression, whereas endothelial cells deficient of EZH2 had increased MAPK7 activity. MAPK7 activation results in increased expression of microRNA (miR)-101, a repressor of EZH2. This loss of EZH2 in turn results in the increased expression of the miR-200 family, culminating in decreased expression of the dual-specificity phosphatases 1 and 6 who may repress MAPK7 activity. Transfection of endothelial cells with miR-200 family members decreased the endothelial sensitivity to TGFß1-induced EndMT. In endothelial cells there is reciprocity between MAPK7 signaling and EZH2 expression and disturbances in this reciprocal signaling associate with the induction of EndMT and severity of human coronary artery disease.


Asunto(s)
Transdiferenciación Celular/fisiología , Enfermedad de la Arteria Coronaria/patología , Endotelio Vascular/patología , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Mesodermo/patología , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Transducción de Señal/fisiología , Túnica Íntima/patología , Regiones no Traducidas 3'/genética , Enfermedad de la Arteria Coronaria/enzimología , Estenosis Coronaria/enzimología , Estenosis Coronaria/patología , Fosfatasa 1 de Especificidad Dual/biosíntesis , Fosfatasa 1 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/biosíntesis , Fosfatasa 6 de Especificidad Dual/genética , Endotelio Vascular/enzimología , Activación Enzimática , Regulación de la Expresión Génica , Genes Reporteros , Código de Histonas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hiperplasia , Mesodermo/enzimología , MicroARNs/biosíntesis , MicroARNs/genética , Túnica Media/patología
8.
J Pharmacol Exp Ther ; 379(2): 117-124, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34389653

RESUMEN

Streptolysin O (SLO) is produced by common hemolytic streptococci that cause a wide range of diseases from pharyngitis to life-threatening necrotizing fasciitis and toxic shock syndrome. Although the importance of SLO in invasive hemolytic streptococcus infection has been well demonstrated, the role of circulating SLO in noninvasive infection remains unclear. The aim of this study was to characterize the pharmacological effect of SLO on vascular functions, focusing on cellular signaling pathways. In control Wistar rats, SLO treatment (1-1000 ng/ml) impaired acetylcholine-induced endothelial-dependent relaxation in the aorta and second-order mesenteric artery in a dose-dependent manner without any effects on sodium nitroprusside-induced endothelium-independent relaxation or agonist-induced contractions. SLO also increased phosphorylation of the endothelial NO synthase (eNOS) inhibitory site at Thr495 in the aorta. Pharmacological analysis indicated that either endothelial dysfunction or eNOS phosphorylation was mediated by protein kinase Cß (PKCß), but not by the p38 mitogen-activated protein kinase pathway. Consistent with this, SLO increased phosphorylation levels of protein kinase C substrates in the aorta. In vivo study of control Wistar rats indicated that intravenous administration of SLO did not change basal blood pressure but significantly counteracted the acetylcholine-induced decrease in blood pressure. Interestingly, plasma anti-SLO IgG levels were significantly higher in 10- to 15-week-old spontaneously hypertensive rats compared with age-matched control rats (P < 0.05). These findings demonstrated that SLO causes vascular endothelial dysfunction, which is mediated by PKCß-induced phosphorylation of the eNOS inhibitory site. SIGNIFICANCE STATEMENT: This study showed for the first time that in vitro exposure of vascular tissues to SLO impairs endothelial function, an effect that is mediated by protein kinase C ß-induced phosphorylation of the endothelial NO synthase inhibitory site. Intravenous administration of SLO in control and hypertensive rats blunted the acetylcholine-induced decrease in blood pressure, providing evidence for a possible role of SLO in dysregulation of blood pressure.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Proteína Quinasa C beta/metabolismo , Estreptolisinas/toxicidad , Vasoconstricción/efectos de los fármacos , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , Proteínas Bacterianas/toxicidad , Relación Dosis-Respuesta a Droga , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/enzimología , Técnicas de Cultivo de Órganos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Wistar , Vasoconstricción/fisiología
9.
Biomolecules ; 11(6)2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204548

RESUMEN

Endothelial lipase (EL) is an enzyme capable of HDL phospholipids hydrolysis. Its action leads to a reduction in the serum high-density lipoprotein concentration, and thus, it exerts a pro-atherogenic effect. This study examines the impact of a single bout exercise on the gene and protein expression of the EL in skeletal muscles composed of different fiber types (the soleus-mainly type I, the red gastrocnemius-mostly IIA, and the white gastrocnemius-predominantly IIX fibers), as well as the diaphragm, and the heart. Wistar rats were subjected to a treadmill run: (1) t = 30 [min], V = 18 [m/min]; (2) t = 30 [min], V = 28 [m/min]; (3) t = 120 [min], V = 18 [m/min] (designated: M30, F30, and M120, respectively). We established EL expression in the total muscle homogenates in sedentary animals. Resting values could be ordered with the decreasing EL protein expression as follows: endothelium of left ventricle > diaphragm > red gastrocnemius > right ventricle > soleus > white gastrocnemius. Furthermore, we observed that even a single bout of exercise was capable of inducing changes in the mRNA and protein level of EL, with a clearer pattern observed for the former. After 30 min of running at either exercise intensity, the expression of EL transcript in all the cardiovascular components of muscles tested, except the soleus, was reduced in comparison to the respective sedentary control. The protein content of EL varied with the intensity and/or duration of the run in the studied whole tissue homogenates. The observed differences between EL expression in vascular beds of muscles may indicate the muscle-specific role of the lipase.


Asunto(s)
Endotelio Vascular/enzimología , Regulación de la Expresión Génica , Lipasa/biosíntesis , Músculo Estriado/enzimología , Condicionamiento Físico Animal , Carrera , Animales , Masculino , Ratas , Ratas Wistar
10.
Exp Biol Med (Maywood) ; 246(21): 2338-2345, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34292081

RESUMEN

In sepsis-induced inflammation, polymorphonuclear neutrophils (PMNs) contribute to vascular dysfunction. The serine proteases proteinase 3 (PR3) and human leukocyte elastase (HLE) are abundant in PMNs and are released upon degranulation. While HLE's role in inflammation-induced endothelial dysfunction is well studied, PR3's role is largely uninvestigated. We hypothesized that PR3, similarly to HLE, contributes to vascular barrier dysfunction in sepsis. Plasma PR3 and HLE concentrations and their leukocyte mRNA levels were measured by ELISA and qPCR, respectively, in sepsis patients and controls. Exogenous PR3 or HLE was applied to human umbilical vein endothelial cells (HUVECs) and HUVEC dysfunction was assessed by FITC-dextran permeability and electrical resistance. Both PR3 and HLE protein and mRNA levels were significantly increased in sepsis patients (P < 0.0001 and P < 0.05, respectively). Additionally, each enzyme independently increased HUVEC monolayer FITC-dextran permeability (P < 0.01), and decreased electrical resistance in a time- and dose-dependent manner (P < 0.001), an effect that could be ameliorated by novel treatment with carbon monoxide-releasing molecule 3 (CORM-3). The serine protease PR3, in addition to HLE, lead to vascular dysfunction and increased endothelial permeability, a hallmark pathological consequence of sepsis-induced inflammation. CORMs may offer a new strategy to reduce serine protease-induced vascular dysfunction.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/enzimología , Mieloblastina/metabolismo , Sepsis/enzimología , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Elastasa de Leucocito/sangre , Elastasa de Leucocito/metabolismo , Masculino , Persona de Mediana Edad , Mieloblastina/sangre , Sepsis/etiología
11.
Microvasc Res ; 138: 104227, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34324883

RESUMEN

This study examined the effects of vitamin D deficiency on vascular function and tissue oxidative status in the microcirculation; and whether or not these effects can be ameliorated with calcitriol, the active vitamin D metabolite. Three groups (n = 10 each) of male Sprague Dawley rats were fed for 10 weeks with control diet (CR), vitamin D-deficient diet without (DR), or with oral calcitriol supplementation (0.15 µg/kg) for the last four weeks (DSR). After 10 weeks, rats were sacrificed; mesenteric arterial rings were studied using wire myograph. Oxidative stress biomarkers malondialdehyde (MDA) levels and superoxide dismutase (SOD) activity were measured in the mesenteric arterial tissue. Vascular protein expression of endothelial nitric oxide synthase (eNOS) was determined by Western blotting. Acetylcholine-induced endothelium-dependent relaxation of DR was lower than CR. eNOS expression and SOD activity were lower in mesenteric arterial tissue of DR compared to CR. Calcitriol supplementation to DSR did not ameliorate the above parameters; in fact, augmented endothelium-dependent contraction was observed. Serum calcium was higher in DSR compared to CR and DR. In conclusion, vitamin D deficiency impaired microvascular vasodilation, associated with eNOS downregulation and reduced antioxidant activity. Calcitriol supplementation to vitamin D-deficient rats at the dosage used augmented endothelium-dependent contraction, possibly due to hypercalcaemia.


Asunto(s)
Antioxidantes/metabolismo , Endotelio Vascular/enzimología , Microcirculación , Microvasos/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo , Vasodilatación , Deficiencia de Vitamina D/enzimología , Animales , Calcitriol/farmacología , Calcio/sangre , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Masculino , Malondialdehído/metabolismo , Microcirculación/efectos de los fármacos , Microvasos/efectos de los fármacos , Microvasos/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal , Superóxido Dismutasa/metabolismo , Vasodilatación/efectos de los fármacos , Deficiencia de Vitamina D/tratamiento farmacológico , Deficiencia de Vitamina D/fisiopatología , Vitaminas/farmacología
12.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R228-R237, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34189960

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) signaling complex is emerging as a critical regulator of cardiovascular function with alterations in this pathway implicated in cardiovascular diseases. In this study, we used animal models and human tissues to examine the role of vascular mTORC1 signaling in the endothelial dysfunction associated with obesity. In mice, obesity induced by high-fat/high-sucrose diet feeding for ∼2 mo resulted in aortic endothelial dysfunction without appreciable changes in vascular mTORC1 signaling. On the other hand, chronic high-fat diet feeding (45% or 60% kcal: ∼9 mo) in mice resulted in endothelial dysfunction associated with elevated vascular mTORC1 signaling. Endothelial cells and visceral adipose vessels isolated from obese humans display a trend toward elevated mTORC1 signaling. Surprisingly, genetic disruption of endothelial mTORC1 signaling through constitutive or tamoxifen inducible deletion of endothelial Raptor (critical subunit of mTORC1) did not prevent or rescue the endothelial dysfunction associated with high-fat diet feeding in mice. Endothelial mTORC1 deficiency also failed to reverse the endothelial dysfunction evoked by a high-fat/high-sucrose diet in mice. Taken together, these data show increased vascular mTORC1 signaling in obesity, but this vascular mTORC1 activation appears not to be required for the development of endothelial impairment in obesity.


Asunto(s)
Endotelio Vascular/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina/deficiencia , Obesidad/prevención & control , Grasa Subcutánea/irrigación sanguínea , Vasodilatación , Animales , Aorta Torácica/enzimología , Aorta Torácica/fisiopatología , Estudios de Casos y Controles , Dieta Alta en Grasa , Sacarosa en la Dieta , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/enzimología , Obesidad/genética , Obesidad/fisiopatología , Proteína Reguladora Asociada a mTOR/deficiencia , Proteína Reguladora Asociada a mTOR/genética , Transducción de Señal
13.
Cardiovasc Toxicol ; 21(10): 790-799, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34185244

RESUMEN

Sirtuins are regulators of eNOS and endothelial function; however, no studies have examined the influence of exercise on sirtuin regulation of endothelial function. Effects of the novel sirtuin inhibitor, salermide, on vascular reactivity in rat aortas were investigated following exercise training of different durations. Male Wistar rats (8-9 months old) were divided into four groups (n = 10-12/group): sedentary (SED), 1 day (1D), 2 weeks (2WK), or 6 weeks (6WK) of exercise. Exercise consisted of running on a motor-driven treadmill at 15 m/min, 15% grade, for 40 min (1D) increased up to 1 h at the end of 2 weeks (2WK) and sustained for an additional 4 weeks (6WK). Dose responses to phenylephrine, sodium nitroprusside, and acetylcholine in the presence or absence of salermide (30 µM) were analyzed. SIRT1 and eNOS protein expression as well as nitrotyrosine levels were determined by immunoblotting. Superoxide dismutase activity was determined by colorimetric assay. Sirtuin inhibition significantly impaired acetylcholine-induced vasorelaxtion in aortas in SED, 1D, and 2WK endurance trained rats but not in 6WK. eNOS expression significantly increased ~ 2.0-fold in 1D, 2WK, and 6WK groups. SIRT1 expression and 3-nitrotyrosine levels were significantly increased in 1D and 2WK but were not significantly elevated in 6WK. SOD levels were significantly elevated in 6WK. These data suggest that chronic endurance training diminishes the role of sirtuins in regulating endothelium-dependent relaxation and appears to be related to changes in SIRT1 expression as well as redox status.


Asunto(s)
Aorta/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Naftoles/farmacología , Fenilpropionatos/farmacología , Condicionamiento Físico Animal , Sirtuina 1/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos , Animales , Aorta/enzimología , Endotelio Vascular/enzimología , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Resistencia Física , Ratas Wistar , Carrera , Transducción de Señal , Sirtuina 1/metabolismo , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Tirosina/análogos & derivados , Tirosina/metabolismo
14.
Invest Ophthalmol Vis Sci ; 62(7): 6, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34086044

RESUMEN

Purpose: To investigate the expression of angiotensin-converting enzyme 2 (ACE2), the receptor for SARS-CoV-2 in human retina. Methods: Human post-mortem eyes from 13 non-diabetic control cases and 11 diabetic retinopathy cases were analyzed for the expression of ACE2. To compare the vascular ACE2 expression between different organs that involve in diabetes, the expression of ACE2 was investigated in renal specimens from nondiabetic and diabetic nephropathy patients. Expression of TMPRSS2, a cell-surface protease that facilitates SARS-CoV-2 entry, was also investigated in human nondiabetic retinas. Primary human retinal endothelial cells (HRECs) and primary human retinal pericytes (HRPCs) were further used to confirm the vascular ACE2 expression in human retina. Results: We found that ACE2 was expressed in multiple nonvascular neuroretinal cells, including the retinal ganglion cell layer, inner plexiform layer, inner nuclear layer, and photoreceptor outer segments in both nondiabetic and diabetic retinopathy specimens. Strikingly, we observed significantly more ACE2 positive vessels in the diabetic retinopathy specimens. By contrast, in another end-stage organ affected by diabetes, the kidney, ACE2 in nondiabetic and diabetic nephropathy showed apical expression of ACE2 tubular epithelial cells, but no endothelial expression in glomerular or peritubular capillaries. Western blot analysis of protein lysates from HRECs and HRPCs confirmed expression of ACE2. TMPRSS2 expression was present in multiple retinal neuronal cells, vascular and perivascular cells, and Müller glia. Conclusions: Together, these results indicate that retina expresses ACE2 and TMPRSS2. Moreover, there are increased vascular ACE2 expression in diabetic retinopathy retinas.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Retinopatía Diabética/enzimología , Receptores Virales/metabolismo , Retina/enzimología , SARS-CoV-2/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Sitios de Unión , Western Blotting , Células Cultivadas , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/virología , Retinopatía Diabética/patología , Retinopatía Diabética/virología , Endotelio Vascular/enzimología , Endotelio Vascular/virología , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pericitos/enzimología , Pericitos/virología , Vasos Retinianos/enzimología , Vasos Retinianos/patología , Vasos Retinianos/virología , Serina Endopeptidasas/metabolismo
15.
Elife ; 102021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33908348

RESUMEN

Background: Hypoxia and consequent production of vascular endothelial growth factor A (VEGFA) promote blood vessel leakiness and edema in ocular diseases. Anti-VEGFA therapeutics may aggravate hypoxia; therefore, therapy development is needed. Methods: Oxygen-induced retinopathy was used as a model to test the role of nitric oxide (NO) in pathological neovascularization and vessel permeability. Suppression of NO formation was achieved chemically using L-NMMA, or genetically, in endothelial NO synthase serine to alanine (S1176A) mutant mice. Results: Suppression of NO formation resulted in reduced retinal neoangiogenesis. Remaining vascular tufts exhibited reduced vascular leakage through stabilized endothelial adherens junctions, manifested as reduced phosphorylation of vascular endothelial (VE)-cadherin Y685 in a c-Src-dependent manner. Treatment with a single dose of L-NMMA in established retinopathy restored the vascular barrier and prevented leakage. Conclusions: We conclude that NO destabilizes adheren junctions, resulting in vascular hyperpermeability, by converging with the VEGFA/VEGFR2/c-Src/VE-cadherin pathway. Funding: This study was supported by the Swedish Cancer foundation (19 0119 Pj ), the Swedish Research Council (2020-01349), the Knut and Alice Wallenberg foundation (KAW 2020.0057) and a Fondation Leducq Transatlantic Network of Excellence Grant in Neurovascular Disease (17 CVD 03). KAW also supported LCW with a Wallenberg Scholar grant (2015.0275). WCS was supported by Grants R35 HL139945, P01 HL1070205, AHA MERIT Award. DV was supported by grants from the Deutsche Forschungsgemeinschaft, SFB1450, B03, and CRU342, P2.


Asunto(s)
Antígenos CD/química , Antígenos CD/metabolismo , Proteína Tirosina Quinasa CSK/metabolismo , Cadherinas/química , Cadherinas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Enfermedades de la Retina/enzimología , Tirosina/metabolismo , Uniones Adherentes/genética , Uniones Adherentes/metabolismo , Secuencias de Aminoácidos , Animales , Antígenos CD/genética , Proteína Tirosina Quinasa CSK/genética , Cadherinas/genética , Permeabilidad Capilar , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836561

RESUMEN

Interferonopathies, interferon (IFN)-α/ß therapy, and caveolin-1 (CAV1) loss-of-function have all been associated with pulmonary arterial hypertension (PAH). Here, CAV1-silenced primary human pulmonary artery endothelial cells (PAECs) were proliferative and hypermigratory, with reduced cytoskeletal stress fibers. Signal transducers and activators of transcription (STAT) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) were both constitutively activated in these cells, resulting in a type I IFN-biased inflammatory signature. Cav1-/- mice that spontaneously develop pulmonary hypertension were found to have STAT1 and AKT activation in lung homogenates and increased circulating levels of CXCL10, a hallmark of IFN-mediated inflammation. PAH patients with CAV1 mutations also had elevated serum CXCL10 levels and their fibroblasts mirrored phenotypic and molecular features of CAV1-deficient PAECs. Moreover, immunofluorescence staining revealed endothelial CAV1 loss and STAT1 activation in the pulmonary arterioles of patients with idiopathic PAH, suggesting that this paradigm might not be limited to rare CAV1 frameshift mutations. While blocking JAK/STAT or AKT rescued aspects of CAV1 loss, only AKT inhibitors suppressed activation of both signaling pathways simultaneously. Silencing endothelial nitric oxide synthase (NOS3) prevented STAT1 and AKT activation induced by CAV1 loss, implicating CAV1/NOS3 uncoupling and NOS3 dysregulation in the inflammatory phenotype. Exogenous IFN reduced CAV1 expression, activated STAT1 and AKT, and altered the cytoskeleton of PAECs, implicating these mechanisms in PAH associated with autoimmune and autoinflammatory diseases, as well as IFN therapy. CAV1 insufficiency elicits an IFN inflammatory response that results in a dysfunctional endothelial cell phenotype and targeting this pathway may reduce pathologic vascular remodeling in PAH.


Asunto(s)
Caveolina 1/genética , Endotelio Vascular/metabolismo , Hipertensión Pulmonar/metabolismo , Interferón Tipo I/metabolismo , Animales , Células Cultivadas , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Silenciador del Gen , Humanos , Hipertensión Pulmonar/fisiopatología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
17.
Biochemistry (Mosc) ; 86(2): 146-155, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33832413

RESUMEN

L-arginine is a key metabolite for nitric oxide production by endothelial cells, as well as signaling molecule of the mTOR signaling pathway. mTOR supports endothelial cells homeostasis and regulates activity of L-arginine-metabolizing enzymes, endothelial nitric oxide synthase, and arginase II. Disruption of the L-arginine metabolism in endothelial cells leads to the development of endothelial dysfunction. Conflicting results of the use of L-arginine supplement to improve endothelial function reveals a controversial role of the amino acid in the endothelial cell biology. The review is aimed at analysis of the current data on the role of L-arginine metabolism in the development of endothelial dysfunction.


Asunto(s)
Arginina/metabolismo , Endotelio Vascular/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Arginasa/metabolismo , Endotelio Vascular/enzimología , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo
18.
J Agric Food Chem ; 69(1): 404-413, 2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33395297

RESUMEN

Myeloperoxidase (MPO)-dependent hypochlorous acid (HOCl) generation plays crucial roles in diabetic vascular complications. As a natural polyphenol, quercetin has antioxidant properties in various diabetic models. Herein, we investigated the therapeutic mechanism for quercetin on MPO-mediated HOCl generation and endothelial dysfunction in diabetic vasculature. In vitro, the presence of MPO could amplify high glucose-induced endothelial dysfunction which was significantly inhibited by the NADPH oxidase inhibitor, HOCl or H2O2 scavengers, revealing the contribution of MPO/H2O2/HOCl to vascular endothelial injury. Furthermore, quercetin effectively inhibited MPO/high glucose-mediated HOCl generation and cytotoxicity to vascular endothelial cells. The inhibitive effect on MPO activity was related to the fact that quercetin reduced high glucose-induced H2O2 generation in endothelial cells and directly acted as a competitive substrate for MPO, thus limiting MPO/H2O2-dependent HOCl production. Moreover, quercetin could attenuate HOCl-caused endothelial dysfunction in endothelial cells and isolated aortas. In vivo, dietary quercetin significantly inhibited aortic endothelial dysfunction in diabetic mice, while this compound simultaneously suppressed vascular MPO expression and activity. Therefore, it was demonstrated herein that quercetin inhibited endothelial injury in diabetic vasculature via suppression of MPO/high glucose-dependent HOCl formation.


Asunto(s)
Angiopatías Diabéticas/enzimología , Endotelio Vascular/metabolismo , Ácido Hipocloroso/metabolismo , Peroxidasa/metabolismo , Quercetina/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Aorta/enzimología , Aorta/metabolismo , Angiopatías Diabéticas/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Endotelio Vascular/enzimología , Humanos , Ratones , Peroxidasa/genética
19.
J Ethnopharmacol ; 267: 113462, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33058924

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The Tongmai Yangxin pill (TMYX) is derived from the Zhigancao decoction recorded in Shang han lun by Zhang Zhongjing during the Han dynasty. TMYX is used for the clinical treatment of chest pain, heartache, and qi-yin-deficiency coronary heart disease. Previous studies have confirmed that TMYX can improve vascular endothelial function in patients with coronary heart disease by upregulating nitric oxide activity and then regulating vascular tension. Whether TMYX can further improve myocardial NR by upregulating NO activity and then dilating blood vessels remains unclear. AIM OF THE STUDY: This study aimed to reveal whether TMYX can further improve myocardial NR by upregulating NO activity and then dilating blood vessels. The underlying cAMP/PKA and NO-cGMP signaling pathway-dependent mechanism is also explored. MATERIALS AND METHODS: The left anterior descending coronary arteries of healthy adult male SD rats were ligated to establish the NR model. TMYX (4.0 g/kg) was orally administered throughout the experiment. Cardiac function was measured through echocardiography. Thioflavin S, Evans Blue, and TTC staining were used to evaluate the NR and ischemic areas. Pathological changes in the myocardium were assessed by hematoxylin-eosin staining. An automated biochemical analyzer and kit were used to detect the activities of myocardial enzymes and myocardial oxidants, including CK, CK-MB, LDH, reactive oxygen species, superoxide dismutase, malonaldehyde, and NO. The expression levels of genes and proteins related to the cAMP/PKA and NO/cGMP signaling pathways were detected via real-time fluorescence quantitative PCR and Western blot analysis, respectively. A microvascular tension sensor was used to detect coronary artery diastolic function in vitro. RESULTS: TMYX elevated the EF, FS, LVOT peak, LVPWd and LVPWs values, decreased the LVIDd, LVIDs, LV-mass, IVSd, and LV Vols values, demonstrating cardio-protective effects, and reduced the NR and ischemic areas. Pathological staining showed that TMYX could significantly reduce inflammatory cell number and interstitial edema. The activities of CK, LDH, and MDA were reduced, NO activity was increased, and oxidative stress was suppressed after treatment with TMYX. TMYX not only enhanced the expression of Gs-α, AC, PKA, and eNOS but also increased the expression of sGC and PKG. Furthermore, TMYX treatment significantly decreased ROCK expression. We further showed that TMYX (25-200 mg/mL) relaxed isolated coronary microvessels. CONCLUSIONS: TMYX attenuates myocardial NR after ischemia and reperfusion by activating the cAMP/PKA and NO/cGMP signaling pathways, further upregulating NO activity and relaxing coronary microvessels.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Medicamentos Herbarios Chinos/farmacología , Endotelio Vascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Fenómeno de no Reflujo/tratamiento farmacológico , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/enzimología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Masculino , Microcirculación/efectos de los fármacos , Miocardio/enzimología , Miocardio/patología , Fenómeno de no Reflujo/enzimología , Fenómeno de no Reflujo/patología , Fenómeno de no Reflujo/fisiopatología , Ratas Sprague-Dawley , Transducción de Señal
20.
Cardiovasc Res ; 117(6): 1546-1556, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32653904

RESUMEN

AIMS: Receptor-type vascular endothelial protein tyrosine phosphatase (VE-PTP) dephosphorylates Tie-2 as well as CD31, VE-cadherin, and vascular endothelial growth factor receptor 2 (VEGFR2). The latter form a signal transduction complex that mediates the endothelial cell response to shear stress, including the activation of the endothelial nitric oxide (NO) synthase (eNOS). As VE-PTP expression is increased in diabetes, we investigated the consequences of VE-PTP inhibition (using AKB-9778) on blood pressure in diabetic patients and the role of VE-PTP in the regulation of eNOS activity and vascular reactivity. METHODS AND RESULTS: In diabetic patients AKB-9778 significantly lowered systolic and diastolic blood pressure. This could be linked to elevated NO production, as AKB increased NO generation by cultured endothelial cells and elicited the NOS inhibitor-sensitive relaxation of endothelium-intact rings of mouse aorta. At the molecular level, VE-PTP inhibition increased the phosphorylation of eNOS on Tyr81 and Ser1177 (human sequence). The PIEZO1 activator Yoda1, which was used to mimic the response to shear stress, also increased eNOS Tyr81 phosphorylation, an effect that was enhanced by VE-PTP inhibition. Two kinases, i.e. abelson-tyrosine protein kinase (ABL)1 and Src were identified as eNOS Tyr81 kinases as their inhibition and down-regulation significantly reduced the basal and Yoda1-induced tyrosine phosphorylation and activity of eNOS. VE-PTP, on the other hand, formed a complex with eNOS in endothelial cells and directly dephosphorylated eNOS Tyr81 in vitro. Finally, phosphorylation of eNOS on Tyr80 (murine sequence) was found to be reduced in diabetic mice and diabetes-induced endothelial dysfunction (isolated aortic rings) was blunted by VE-PTP inhibition. CONCLUSIONS: VE-PTP inhibition enhances eNOS activity to improve endothelial function and decrease blood pressure indirectly, through the activation of Tie-2 and the CD31/VE-cadherin/VEGFR2 complex, and directly by dephosphorylating eNOS Tyr81. VE-PTP inhibition, therefore, represents an attractive novel therapeutic option for diabetes-induced endothelial dysfunction and hypertension.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Antihipertensivos/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Hipertensión/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/antagonistas & inhibidores , Ácidos Sulfónicos/uso terapéutico , Animales , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Humanos , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Transducción de Señal , Resultado del Tratamiento , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...