Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Am Heart Assoc ; 5(10)2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27729334

RESUMEN

BACKGROUND: Lifestyle modifications are first-line measures for cardiovascular disease prevention. Whether lifestyle intervention also preserves cardiovascular health is less clear. Our study examined the role of a Health Partner-administered lifestyle intervention on metrics of ideal cardiovascular health. METHODS AND RESULTS: A total of 711 university employees (48±11 years; 66% women, 72% Caucasian/22.5% African Americans) enrolled in a program that promoted healthier lifestyles at Emory University (Atlanta, GA). Anthropometric, laboratory, and physical activity measurements were performed at baseline and at 6 months, 1 year, and 2 years of follow-up. Results were utilized by the Health Partner to generate a personalized plan aimed at meeting ideal health metrics. Compared to baseline, at each of the 6-month, 1-year, and 2-year follow-up visits, systolic blood pressure was lower by 3.6, 4.6, and 3.3 mm Hg (P<0.001), total cholesterol decreased by 5.3, 6.5, and 6.4 mg/dL (P<0.001), body mass index declined by 0.33, 0.45, and 0.38 kg/m2 (P<0.001), and the percentage of smokers decreased by 1.3%, 3.5%, and 3.5% (P<0.01), respectively. Changes were greater in those with greater abnormalities at baseline. Finally, the American Heart Association "Life's Simple 7" ideal cardiovascular health score increased by 0.28, 0.40, and 0.33 at 6 month, 1 year, and 2 years, respectively, compared to baseline visit. CONCLUSIONS: A personalized, goal-directed Health Partner intervention significantly improved the cardiometabolic risk profile and metrics of cardiovascular health. These effects were evident at 6 months following enrollment and were sustained for 2 years. Whether the Health Partner intervention improves long-term morbidity and mortality and is cost-effective needs further investigation.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Dieta , Ejercicio Físico , Conductas Relacionadas con la Salud , Educadores en Salud , Salud Laboral , Conducta de Reducción del Riesgo , Cese del Hábito de Fumar , Adulto , Negro o Afroamericano , American Heart Association , Presión Sanguínea , Índice de Masa Corporal , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/epidemiología , Colesterol/sangre , Femenino , Humanos , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Estados Unidos , Universidades , Población Blanca
2.
Cell Rep ; 12(9): 1391-9, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26299964

RESUMEN

Cellular senescence and organismal aging predispose age-related chronic diseases, such as neurodegenerative, metabolic, and cardiovascular disorders. These diseases emerge coincidently from elevated oxidative/electrophilic stress, inflammation, mitochondrial dysfunction, DNA damage, and telomere dysfunction and shortening. Mechanistic linkages are incompletely understood. Here, we show that ablation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) accelerates vascular aging and atherosclerosis, coinciding with telomere dysfunction and shortening and DNA damage. PGC-1α deletion reduces expression and activity of telomerase reverse transcriptase (TERT) and increases p53 levels. Ectopic expression of PGC-1α coactivates TERT transcription and reverses telomere malfunction and DNA damage. Furthermore, alpha lipoic acid (ALA), a non-dispensable mitochondrial cofactor, upregulates PGC-1α-dependent TERT and the cytoprotective Nrf-2-mediated antioxidant/electrophile-responsive element (ARE/ERE) signaling cascades, and counteracts high-fat-diet-induced, age-dependent arteriopathy. These results illustrate the pivotal importance of PGC-1α in ameliorating senescence, aging, and associated chronic diseases, and may inform novel therapeutic approaches involving electrophilic specificity.


Asunto(s)
Aterosclerosis/genética , Vasos Sanguíneos/crecimiento & desarrollo , Daño del ADN , Acortamiento del Telómero , Factores de Transcripción/genética , Animales , Elementos de Respuesta Antioxidante , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Ácido Tióctico/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
J Am Heart Assoc ; 3(6): e001202, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25527624

RESUMEN

BACKGROUND: Clinical studies show that metformin attenuates all­cause mortality and myocardial infarction compared with other medications for type 2 diabetes, even at similar glycemic levels. However, there is paucity of data in the euglycemic state on the vasculoprotective effects of metformin. The objectives of this study are to evaluate the effects of metformin on ameliorating atherosclerosis. METHODS AND RESULTS: Using ApoE−/− C57BL/6J mice, we found that metformin attenuates atherosclerosis and vascular senescence in mice fed a high­fat diet and prevents the upregulation of angiotensin II type 1 receptor by a high­fat diet in the aortas of mice. Thus, considering the known deleterious effects of angiotensin II mediated by angiotensin II type 1 receptor, the vascular benefits of metformin may be mediated, at least in part, by angiotensin II type 1 receptor downregulation. Moreover, we found that metformin can cause weight loss without hypoglycemia. We also found that metformin increases the antioxidant superoxide dismutase­1. CONCLUSION: Pleiotropic effects of metformin ameliorate atherosclerosis and vascular senescence.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Fármacos Cardiovasculares/farmacología , Hipoglucemiantes/farmacología , Metformina/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Senescencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Pérdida de Peso/efectos de los fármacos
5.
Arterioscler Thromb Vasc Biol ; 33(5): 988-98, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23430617

RESUMEN

OBJECTIVE: Cellular senescence influences organismal aging and increases predisposition to age-related diseases, in particular cardiovascular disease, a leading cause of death and disability worldwide. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis and function, oxidative stress, and insulin resistance. Senescence is associated with telomere and mitochondrial dysfunction and oxidative stress, implying a potential causal role of PGC-1α in senescence pathogenesis. APPROACH AND RESULTS: We generated a PGC-1α(+/-)/apolipoprotein E(-/-) mouse model and showed that PGC-1α deficiency promotes a vascular senescence phenotype that is associated with increased oxidative stress, mitochondrial abnormalities, and reduced telomerase activity. PGC-1α disruption results in reduced expression of the longevity-related deacetylase sirtuin 1 (SIRT1) and the antioxidant catalase, and increased expression of the senescence marker p53 in aortas. Further, angiotensin II, a major hormonal inducer of vascular senescence, induces prolonged lysine acetylation of PGC-1α and releases the PGC-1α-FoxO1 complex from the SIRT1 promoter, thus reducing SIRT1 expression. The phosphorylation-defective mutant PGC-1α S570A is not acetylated, is constitutively active for forkhead box O1-dependent SIRT1 transcription, and prevents angiotensin II-induced senescence. Acetylation of PGC-1α by angiotensin II interrupts the PGC-1α-forkhead box O1-SIRT1 feed-forward signaling circuit leading to SIRT1 and catalase downregulation and vascular senescence. CONCLUSIONS: PGC-1α is a primary negative regulator of vascular senescence. Moreover, the central role of posttranslational modification of PGC-1α in regulating angiotensin II-induced vascular senescence may inform development of novel therapeutic strategies for mitigating age-associated diseases, such as atherosclerosis.


Asunto(s)
Senescencia Celular , Músculo Liso Vascular/citología , Transactivadores/fisiología , Acetilación , Angiotensina II/farmacología , Animales , Catalasa/análisis , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosforilación , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 1/análisis , Sirtuina 1/genética , Telomerasa/metabolismo , Factores de Transcripción
6.
J Biomol Screen ; 17(9): 1163-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22885731

RESUMEN

Vascular injury mobilizes bone marrow-derived proangiogenic cells into the circulation, where these cells can facilitate vascular repair and new vessel formation. We sought to determine the relationship between a new biomarker of circulating bone marrow-derived proangiogenic cell activity, the presence of atherosclerotic cardiovascular disease (CVD) and its risk factors, and clinical outcomes. Circulating proangiogenic cell activity was estimated using a reproducible angiogenic colony-forming unit (CFU-A) assay in 532 clinically stable subjects aged 20 to 90 years and ranging in the CVD risk spectrum from those who are healthy without risk factors to those with active CVD. CFU-A counts increased with the burden of CVD risk factors (p < 0.001). CFU-A counts were higher in subjects with symptomatic CVD than in those without (p < 0.001). During follow-up of 232 subjects with CVD, CFU-A counts were higher in those with death, myocardial infarction, or stroke than in those without (110 [70-173] vs 84 [51-136], p = 0.01). Therefore, we conclude that circulating proangiogenic cell activity, as estimated by CFU-A counts, increases with CVD risk factor burden and in the presence of established CVD. Furthermore, higher circulating proangiogenic cell activity is associated with worse clinical outcome in those with CVD.


Asunto(s)
Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/epidemiología , Ensayo de Unidades Formadoras de Colonias , Células Madre Hematopoyéticas/patología , Adulto , Anciano , Anciano de 80 o más Años , Aterosclerosis/sangre , Aterosclerosis/epidemiología , Aterosclerosis/patología , Biomarcadores/sangre , Enfermedades Cardiovasculares/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/sangre , Infarto del Miocardio/epidemiología , Infarto del Miocardio/patología , Revascularización Miocárdica , Factores de Riesgo , Índice de Severidad de la Enfermedad , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/patología
7.
J Biomol Screen ; 17(9): 1128-35, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22904201

RESUMEN

Although multiple culture assays have been designed to identify endothelial progenitor cells (EPCs), the phenotype of cells grown in culture often remains undefined. We sought to define and characterize the proangiogenic cell population within human peripheral blood mononuclear cells. Mononuclear cells were isolated from peripheral blood and grown under angiogenic conditions for 7 days. Formed colonies (CFU-As) were identified and analyzed for proliferation, mRNA and surface antigen expression, tube-forming ability, and chromosomal content. Colonies were composed of a heterogeneous group of cells expressing the leukocyte antigens CD45, CD14, and CD3, as well as the endothelial proteins vascular endothelial (VE) cadherin, von Willebrand's factor (vWF), CD31, and endothelial nitric oxide synthase (eNOS). Colony cells expressed increased levels of proangiogenic growth factors, and they formed tubes in Matrigel. In comparison with colonies from the CFU-Hill assay, our assay resulted in a greater number of colonies (19 ± 9 vs. 13 ± 7; p < 0.0001) with a substantial number of cells expressing an endothelial phenotype (20.2% ± 7.4% vs. 2.2% ± 1.2% expressing eNOS, p = 0.0006). Chromosomal analysis indicated the colony cells were bone marrow derived. We, therefore, describe a colony-forming unit assay that measures bone marrow-derived circulating mononuclear cells with the capacity to proliferate and mature into proangiogenic leukocytic and endothelial-like cells. This assay, therefore, reflects circulating, bone marrow-derived proangiogenic activity.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias/métodos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Neovascularización Fisiológica , Células Madre/citología , Células Madre/metabolismo , Antígenos CD/análisis , Antígenos CD/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Trasplante de Médula Ósea , Complejo CD3/metabolismo , Cadherinas/análisis , Cadherinas/metabolismo , Diferenciación Celular/fisiología , Colágeno/metabolismo , Combinación de Medicamentos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Comités de Ética en Investigación , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Laminina/metabolismo , Antígenos Comunes de Leucocito/análisis , Antígenos Comunes de Leucocito/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo III/análisis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Cultivo Primario de Células , Proteoglicanos/metabolismo , Factor de von Willebrand/análisis , Factor de von Willebrand/metabolismo
8.
Am J Hypertens ; 25(7): 797-803, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22513829

RESUMEN

BACKGROUND: Genome-wide association studies (GWAS) have identified novel variants associated with myocardial infarction (MI) in Caucasians. We hypothesized that those variants whose mechanism of risk is currently unknown, confer risk via pathways mediating arterial wave reflections which is an increasingly recognized risk factor for cardiovascular disease. METHODS: Single-nucleotide polymorphisms (SNPs) at eight MI risk loci were genotyped and correlated with noninvasively determined pulse wave analysis (PWA)-derived central hemodynamic indexes (augmentation index (AIx); augmented pressure (AP); time to reflected wave (TrW) and central systolic blood pressure (SBP) and diastolic BP (DBP)) in two independent Caucasian populations including (i) those free of measured cardiovascular risk factors (n = 133) and (ii) a community-based population (n = 270). RESULTS: Of the eight SNPs examined in the healthy group, the variants at loci 6p24 (AIx and AP both P < 0.001, TrW P = 0.02) and 21q22 (AIx P = 0.002, TrW P = 0.037) were significantly associated with PWA indexes. In the replication group, only the 6p24 variant correlated with these phenotypes (AIx P = 0.005, AP P = 0.049, TrW P = 0.013). In the pooled population (n = 403), no new associations were identified but the association with 6p24 and AIx remained significant even after Bonferroni correction and adjustment for covariates including age, mean arterial pressure, height, gender, glucose, cholesterol, body mass index (BMI), and smoking (AIx (P = 0.03)). Each copy of the risk allele C increased the AIx by 3.5%. CONCLUSIONS: The GWAS discovered MI risk variant at 6p24 in the protein phosphatase 1 regulator gene (PHACTR1) is associated with adverse arterial wave reflection indexes and may mediate MI risk through this pathway.


Asunto(s)
Cromosomas Humanos Par 6/genética , Proteínas de Microfilamentos/genética , Infarto del Miocardio/genética , Adulto , Anciano , Presión Sanguínea/fisiología , Técnicas de Diagnóstico Cardiovascular , Femenino , Estudio de Asociación del Genoma Completo , Hemodinámica/genética , Hemodinámica/fisiología , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Riesgo , Población Blanca/genética
9.
J Am Coll Cardiol ; 58(2): 186-92, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21718915

RESUMEN

OBJECTIVES: The primary objective of this study was to elucidate mechanisms underlying the link between vitamin D status and cardiovascular disease by exploring the relationship between 25-hydroxyvitamin D (25-OH D), an established marker of vitamin D status, and vascular function in healthy adults. BACKGROUND: Mechanisms underlying vitamin D deficiency-mediated increased risk of cardiovascular disease remain unknown. Vitamin D influences endothelial and smooth muscle cell function, mediates inflammation, and modulates the renin-angiotensin-aldosterone axis. We investigated the relationship between vitamin D status and vascular function in humans, with the hypothesis that vitamin D insufficiency will be associated with increased arterial stiffness and abnormal vascular function. METHODS: We measured serum 25-OH D in 554 subjects. Endothelial function was assessed as brachial artery flow-mediated dilation, and microvascular function was assessed as digital reactive hyperemia index. Carotid-femoral pulse wave velocity and radial tonometry-derived central augmentation index and subendocardial viability ratio were measured to assess arterial stiffness. RESULTS: Mean 25-OH D was 31.8 ± 14 ng/ml. After adjustment for age, sex, race, body mass index, total cholesterol, low-density lipoprotein, triglycerides, C-reactive protein, and medication use, 25-OH D remained independently associated with flow-mediated vasodilation (ß = 0.1, p = 0.03), reactive hyperemia index (ß = 0.23, p < 0.001), pulse wave velocity (ß = -0.09, p = 0.04), augmentation index (ß = -0.11, p = 0.03), and subendocardial viability ratio (ß = 0.18, p = 0.001). In 42 subjects with vitamin D insufficiency, normalization of 25-OH D at 6 months was associated with increases in reactive hyperemia index (0.38 ± 0.14, p = 0.009) and subendocardial viability ratio (7.7 ± 3.1, p = 0.04), and a decrease in mean arterial pressure (4.6 ± 2.3 mm Hg, p = 0.02). CONCLUSIONS: Vitamin D insufficiency is associated with increased arterial stiffness and endothelial dysfunction in the conductance and resistance blood vessels in humans, irrespective of traditional risk burden. Our findings provide impetus for larger trials to assess the effects of vitamin D therapy in cardiovascular disease.


Asunto(s)
Arterias/patología , Resistencia Vascular , Deficiencia de Vitamina D/complicaciones , Vitamina D/metabolismo , Adulto , Arteria Braquial/patología , Enfermedades Cardiovasculares/sangre , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Riesgo , Enfermedades Vasculares/patología , Vitamina D/análogos & derivados , Vitamina D/sangre
10.
Atherosclerosis ; 218(1): 90-5, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21605864

RESUMEN

AIMS: Arterial stiffening may lead to hypertension, greater left ventricular after-load and adverse clinical outcomes. The underlying mechanisms influencing arterial elasticity may involve oxidative injury to the vessel wall. We sought to examine the relationship between novel markers of oxidative stress and arterial elastic properties in healthy humans. METHODS AND RESULTS: We studied 169 subjects (mean age 42.6 ± 14 years, 51.6% male) free of traditional cardiovascular risk factors. Indices of arterial stiffness and wave reflections measured included carotid-femoral Pulse Wave Velocity (PWV), Augmentation Index (Aix) and Pulse Pressure Amplification (PPA). Non-free radical oxidative stress was assessed as plasma oxidized and reduced amino-thiol levels (cysteine/cystine, glutathione/GSSG) and their ratios (redox potentials), and free radical oxidative stress as derivatives of reactive oxygen metabolites (dROMs). Inflammation was assessed as hsCRP and interleukin-6 levels. The non-free radical marker of oxidative stress, cystine was significantly correlated with all arterial indices; PWV (r=0.38, p<0.001), Aix (r=0.35, p<0.001) and PPA (r=-0.30, p<0.001). Its redox potential, was also associated with PWV (r=0.22, p=0.01), while the free radical marker of oxidative stress dROMS was associated with Aix (r=0.25, p<0.01). After multivariate adjustment for age, gender, arterial pressure, height, weight, heart rate and CRP, of these oxidative stress markers, only cystine remained independently associated with PWV (p=0.03), Aix (p=0.01) and PPA (p=0.05). CONCLUSIONS: In healthy subjects without confounding risk factors or significant systemic inflammation, a high cystine level, reflecting extracellular oxidant burden, is associated with increased arterial stiffness and wave reflections. This has implications for understanding the role of oxidant burden in pre-clinical vascular dysfunction.


Asunto(s)
Arterias/patología , Rigidez Vascular , Adulto , Envejecimiento , Proteína C-Reactiva/metabolismo , Enfermedades Cardiovasculares/patología , Cistina/química , Elasticidad , Femenino , Humanos , Inflamación , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oxidación-Reducción , Estrés Oxidativo , Factores de Riesgo
11.
J Biol Chem ; 286(7): 5289-99, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21149440

RESUMEN

Forkhead transcription factor FoxO1 and the NAD(+)-dependent histone deacetylase SIRT1 are evolutionarily conserved regulators of the development of aging, oxidative stress resistance, insulin resistance, and metabolism in species ranging from invertebrates to mammals. SIRT1 deacetylates FoxO1 and enables activation of FoxO1 transcription in multiple systems. The functional consequences of the interactions between FoxO1 and SIRT1 remain incompletely understood. Here, we demonstrate that the 1.5-kb rat sirt1 promoter region contains a cluster of five putative FoxO1 core binding repeat motifs (5×IRS-1) and a forkhead-like consensus binding site (FKHD-L). Luciferase promoter assays demonstrate that FoxO1 directly activates SIRT1 promoter activity and that both the IRS-1 and FKHD-L enable FoxO1-dependent SIRT1 transcription. Electrophoretic mobility shift and chromatin immunoprecipitation assays show that FoxO1 binds to the IRS-1 and FKHD-L sites of the SIRT1 promoter. Consistently, FoxO1 overexpression increases SIRT1 expression, and FoxO1 depletion by siRNA reduces SIRT1 expression at both the messenger RNA and protein levels in vascular smooth muscle cells and HEK293 cells. Thus, endogenous FoxO1 is a positive transcriptional regulator of SIRT1. Conversely, SIRT1 promotes FoxO1-driven SIRT1 autotranscription through interacting with and deacetylating FoxO1. Moreover, resveratrol, a plant polyphenol activator of SIRT1, increases FoxO1-dependent SIRT1 transcription activity and thus induces its expression. These findings suggest that positive feedback mechanisms regulate FoxO1-dependent SIRT1 transcription and indicate a previously unappreciated function for FoxO1. This signaling network may coordinate multiple pathways acting upon immune, inflammatory, regenerative, and metabolic processes.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Elementos de Respuesta/fisiología , Sirtuina 1/biosíntesis , Transcripción Genética/fisiología , Animales , Inhibidores Enzimáticos/farmacología , Factores de Transcripción Forkhead/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Proteínas del Tejido Nervioso/genética , ARN Interferente Pequeño/genética , Ratas , Resveratrol , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirtuina 1/genética , Estilbenos/farmacología , Transcripción Genética/efectos de los fármacos
12.
J Biol Chem ; 286(4): 2886-95, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21097843

RESUMEN

Akt/protein kinase B (PKB) activation/phosphorylation by angiotensin II (Ang II) is a critical signaling event in hypertrophy of vascular smooth muscle cells (VSMCs). Conventional wisdom asserts that Akt activation occurs mainly in plasma membrane domains. Recent evidence that Akt activation may take place within intracellular compartments challenges this dogma. The spatial identity and mechanistic features of these putative signaling domains have not been defined. Using cell fractionation and fluorescence methods, we demonstrate that the early endosomal antigen-1 (EEA1)-positive endosomes are a major site of Ang II-induced Akt activation. Akt moves to and is activated in EEA1 endosomes. The expression of EEA1 is required for phosphorylation of Akt at both Thr-308 and Ser-473 as well as for phosphorylation of its downstream targets mTOR and S6 kinase, but not for Erk1/2 activation. Both Akt and phosphorylated Akt (p-Akt) interact with EEA1. We also found that PKC-α is required for organizing Ang II-induced, EEA1-dependent Akt phosphorylation in VSMC early endosomes. EEA1 expression enables PKC-α phosphorylation, which in turn regulates Akt upstream signaling kinases, PDK1 and p38 MAPK. Our results indicate that PKC-α is a necessary regulator of EEA1-dependent Akt signaling in early endosomes. Finally, EEA1 down-regulation or expression of a dominant negative mutant of PKC-α blunts Ang II-induced leucine incorporation in VSMCs. Thus, EEA1 serves a novel function as an obligate scaffold for Ang II-induced Akt activation in early endosomes.


Asunto(s)
Angiotensina II/farmacología , Endosomas/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Vasoconstrictores/farmacología , Proteínas de Transporte Vesicular/biosíntesis , Animales , Células Cultivadas , Endosomas/genética , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteína Quinasa C-alfa/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
J Biol Chem ; 285(4): 2474-87, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19940161

RESUMEN

Angiotensin II (Ang II) is a pleuripotential hormone that is important in the pathophysiology of multiple conditions including aging, cardiovascular and renal diseases, and insulin resistance. Reactive oxygen species (ROS) are important mediators of Ang II-induced signaling generally and have a well defined role in vascular hypertrophy, which is inhibited by overexpression of catalase, inferring a specific role of H(2)O(2). The molecular mechanisms are understood incompletely. The transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1 alpha) is a key regulator of energy metabolism and ROS-scavenging enzymes including catalase. We show that Ang II stimulates Akt-dependent PGC-1 alpha serine 570 phosphorylation, which is required for the binding of the histone acetyltransferase GCN5 (general control nonderepressible 5) to PGC-1 alpha and for its lysine acetylation. These sequential post-translational modifications suppress PGC-1 alpha activity and prevent its binding to the catalase promoter through the forkhead box O1 transcription factor, thus decreasing catalase expression. We demonstrate that overexpression of the phosphorylation-defective mutant PGC-1 alpha (S570A) prevents Ang II-induced increases in H(2)O(2) levels and hypertrophy ([(3)H]leucine incorporation). Knockdown of PGC-1 alpha by small interfering RNA promotes basal and Ang II-stimulated ROS and hypertrophy, which is reversed by polyethylene glycol-conjugated catalase. Thus, endogenous PGC-1 alpha is a negative regulator of vascular hypertrophy by up-regulating catalase expression and thus reducing ROS levels. We provide novel mechanistic insights by which Ang II may mediate its ROS-dependent pathophysiologic effects on multiple cardiometabolic diseases.


Asunto(s)
Angiotensina II/metabolismo , Enfermedades Cardiovasculares/metabolismo , Catalasa/metabolismo , Músculo Liso Vascular/enzimología , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Angiotensina II/farmacología , Animales , Aorta Torácica/citología , Enfermedades Cardiovasculares/patología , Catalasa/genética , Células Cultivadas , Regulación hacia Abajo/fisiología , Factores de Transcripción Forkhead/metabolismo , Hipertrofia , Luciferasas/genética , Masculino , Músculo Liso Vascular/patología , Proteínas del Tejido Nervioso/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Regiones Promotoras Genéticas/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Factores de Transcripción/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología
15.
Acad Med ; 84(10): 1395-400, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19881429

RESUMEN

To establish guidelines for more effectively incorporating part-time faculty into departments of internal medicine, a task force was convened in early 2007 by the Association of Specialty Professors. The task force used informal surveys, current literature, and consensus building among members of the Alliance for Academic Internal Medicine to produce a consensus statement and a series of recommendations. The task force agreed that part-time faculty could enrich a department of medicine, enhance workforce flexibility, and provide high-quality research, patient care, and education in a cost-effective manner. The task force provided a series of detailed steps for operationalizing part-time practice; to do so, key issues were addressed, such as fixed costs, malpractice insurance, space, cross-coverage, mentoring, career development, productivity targets, and flexible scheduling. Recommendations included (1) increasing respect for work-family balance, (2) allowing flexible time as well as part-time employment, (3) directly addressing negative perceptions about part-time faculty, (4) developing policies to allow flexibility in academic advancement, (5) considering part-time faculty as candidates for leadership positions, (6) encouraging granting agencies, including the National Institutes of Health and Veterans Administration, to consider part-time faculty as eligible for research career development awards, and (7) supporting future research in "best practices" for incorporating part-time faculty into academic departments of medicine.


Asunto(s)
Docentes Médicos/organización & administración , Medicina Interna/organización & administración , Admisión y Programación de Personal/organización & administración , Facultades de Medicina , Eficiencia Organizacional , Humanos , Relaciones Interprofesionales , Estilo de Vida , Admisión y Programación de Personal/clasificación , Admisión y Programación de Personal/economía , Facultades de Medicina/economía , Facultades de Medicina/organización & administración , Estados Unidos , Recursos Humanos , Carga de Trabajo
16.
Am Heart J ; 158(1): 53-60.e1, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19540392

RESUMEN

BACKGROUND: We hypothesized that granulocyte macrophage colony-stimulating factor (GM-CSF) administration will be safe and will improve endothelial dysfunction and exercise capacity by mobilizing progenitor cells in patients with peripheral arterial disease (PAD). METHODS: Forty-five patients with PAD received thrice-weekly injections for 2 weeks of 3, 6, or 10 microg/kg per day of GM-CSF or placebo in successive cohorts of 15 subjects randomized 2:1 to drug or placebo. CD34+ mononuclear cell subsets and colony formation assay, endothelial function, ankle-brachial index, and walking capacity were measured. RESULTS: Granulocyte macrophage colony-stimulating factor administration was safe. After pooling data from GM-CSF cohorts, at 2 weeks, there was a significant increase in total leukocytes (43%, P < .0001), CD34+ cells (46%, P = .035), and colony-forming units (31%, P = .026, week 1). At 12 weeks, endothelial function improved with GM-CSF (flow-mediated vasodilation increased by 59%, P < .01) as did pain-free treadmill walking time (38 seconds, P = .008) and total treadmill walking time (55 seconds, P = .016). Corresponding changes were not observed in the placebo group. CONCLUSIONS: Granulocyte macrophage colony-stimulating factor therapy in patients with PAD was associated with mobilization of progenitor cells, improvement of endothelial dysfunction, and exercise capacity. The efficacy of strategies designed to mobilize bone marrow progenitors warrants further study in patients with PAD.


Asunto(s)
Arteriopatías Oclusivas/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Médula Ósea/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Prueba de Esfuerzo/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Movilización de Célula Madre Hematopoyética/métodos , Anciano , Índice Tobillo Braquial , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Endotelio Vascular/fisiopatología , Femenino , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Humanos , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Vasodilatación
17.
Am J Physiol Regul Integr Comp Physiol ; 297(1): R202-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19458279

RESUMEN

Proton nuclear magnetic resonance ((1)H-NMR) spectroscopy of plasma provides a global metabolic profiling method that shows promise for clinical diagnostics. However, cross-sectional studies are complicated by a lack of understanding of intraindividual variation, and this limits experimental design and interpretation of data. The present study determined the diurnal variation detected by (1)H NMR spectroscopy of human plasma. Data reduction methods revealed three time-of-day metabolic patterns, which were associated with morning, afternoon, and night. Major discriminatory regions for these time-of-day patterns included the various kinds of lipid signals (-CH(2)- and -CH(2)OCOR), and the region between 3 and 4 ppm heavily overlapped with amino acids that had alpha-CH and alpha-CH(2). The phasing and duration of time-of-day patterns were variable among individuals, apparently because of individual difference in food processing/digestion and absorption and clearance of macronutrient energy sources (fat, protein, carbohydrate). The times of day that were most consistent among individuals, and therefore most useful for cross-sectional studies, were fasting morning (0830-0930), postprandial afternoon (1430-1630), and nighttime samples (0430-0530). Importantly, the integrated picture of metabolism provided by (1)H-NMR spectroscopy of plasma suggests that this approach is suitable to study complex regulatory processes, including eating patterns/eating disorders, upper gastrointestinal functions (gastric emptying, pancreatic, biliary functions), and absorption/clearance of macronutrients. Hence, (1)H-NMR spectroscopy of plasma could provide a global metabolic tolerance test to assess complex processes involved in disease, including eating disorders and the range of physiological processes causing dysregulation of energy homeostasis.


Asunto(s)
Ritmo Circadiano , Carbohidratos de la Dieta/sangre , Grasas de la Dieta/sangre , Proteínas en la Dieta/sangre , Ingestión de Alimentos , Metabolismo Energético , Espectroscopía de Resonancia Magnética , Metabolómica/métodos , Adulto , Anciano , Anciano de 80 o más Años , Aminoácidos/sangre , Análisis por Conglomerados , Ayuno/sangre , Femenino , Homeostasis , Humanos , Masculino , Persona de Mediana Edad , Periodo Posprandial , Análisis de Componente Principal , Valores de Referencia , Adulto Joven
18.
Hypertension ; 52(1): 80-5, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18504327

RESUMEN

Endothelial dysfunction is known to precede the development of atherosclerosis and results primarily from increased oxidative degradation of NO. We hypothesized that assessment of oxidative stress in the bloodstream will reliably predict endothelial function in healthy adults. A total of 124 healthy nonsmokers had endothelial function assessed using ultrasound measurement of brachial artery flow-mediated vasodilation. Plasma oxidative stress was estimated by measuring the levels of the reduced and oxidized forms of thiols, including glutathione (reduced glutathione and oxidized glutathione) and cysteine (cysteine and cystine), respectively, and the mixed disulfide. Among the traditional risk factors, there were significant and independent correlations between flow-mediated vasodilation and high-density lipoprotein level, body mass index, gender, and the Framingham risk score. Among the thiol markers, plasma cystine (r=-0.23; P=0.009) and the mixed disulfide (r=-0.23; P=0.01) levels correlated with endothelium-dependent but not endothelium-independent vasodilation, even after adjusting for the Framingham risk score and high-sensitivity C-reactive protein level. A higher level of oxidized metabolites was associated with worse endothelial function. In conclusion, the oxidative stress markers, cystine, and the mixed disulfide are independent predictors of endothelial function. These markers, in combination with the Framingham risk score, may help in the early identification of asymptomatic subjects with endothelial dysfunction who are at potentially increased risk for future atherosclerotic disease progression.


Asunto(s)
Aterosclerosis/diagnóstico , Aterosclerosis/fisiopatología , Endotelio Vascular/fisiopatología , Estrés Oxidativo , Compuestos de Sulfhidrilo/sangre , Adulto , Anciano , Aterosclerosis/sangre , Biomarcadores/sangre , Arteria Braquial/diagnóstico por imagen , Arteria Braquial/efectos de los fármacos , Arteria Braquial/fisiopatología , Cisteína/análogos & derivados , Cisteína/sangre , Endotelio Vascular/efectos de los fármacos , Femenino , Glutatión/análogos & derivados , Glutatión/sangre , Disulfuro de Glutatión/sangre , Humanos , Masculino , Persona de Mediana Edad , Nitroglicerina/farmacología , Factores de Riesgo , Ultrasonografía , Vasodilatación
20.
Eur Heart J ; 27(20): 2400-5, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16782719

RESUMEN

AIMS: The purpose of this analysis was to determine whether the efficacy of adenosine vs. placebo was dependent on the timing of reperfusion therapy in the second Acute Myocardial Infarction Study of Adenosine (AMISTAD-II). METHODS AND RESULTS: Patients presenting with ST-segment elevation anterior AMI were randomized to receive placebo vs. adenosine (50 or 70 microg/kg/min) for 3 h starting within 15 min of reperfusion therapy. In the present post hoc hypothesis generating study, the results were stratified according to the timing of reperfusion, i.e. > or = or < the median 3.17 h, and by reperfusion modality. In patients receiving reperfusion < 3.17 h, adenosine compared with placebo significantly reduced 1-month mortality (5.2 vs. 9.2%, respectively, P = 0.014), 6-month mortality (7.3 vs. 11.2%, P = 0.033), and the occurrence of the primary 6-month composite clinical endpoint of death, in-hospital CHF, or rehospitalization for CHF at 6 months (12.0 vs. 17.2%, P = 0.022). Patients reperfused beyond 3 h did not benefit from adenosine. CONCLUSION: In this post hoc analysis, 3 h adenosine infusion administered as an adjunct to reperfusion therapy within the first 3.17 h onset of evolving anterior ST-segment elevation AMI enhanced early and late survival, and reduced the composite clinical endpoint of death or CHF at 6 months.


Asunto(s)
Adenosina/uso terapéutico , Infarto del Miocardio/terapia , Reperfusión Miocárdica/métodos , Vasodilatadores/uso terapéutico , Angioplastia Coronaria con Balón/mortalidad , Método Doble Ciego , Femenino , Mortalidad Hospitalaria , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/mortalidad , Reperfusión Miocárdica/mortalidad , Recurrencia , Terapia Trombolítica/mortalidad , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA