Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genomics ; 13(1): 22, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113495

RESUMEN

BACKGROUND: Elevated resting heart rate (HR) is a risk factor and therapeutic target in patients with heart failure (HF) and reduced ejection fraction (HFrEF). Previous studies indicate a genetic contribution to HR in population samples but there is little data in patients with HFrEF. METHODS: Patients who met Framingham criteria for HF and had an ejection fraction < 50% were prospectively enrolled in a genetic HF registry (2007-2015, n = 1060). All participants donated blood for DNA and underwent genome-wide genotyping with additional variants called via imputation. We performed testing of previously identified variant "hits" (43 loci) as well as a genome-wide association (GWAS) of HR, adjusted for race, using Efficient Mixed-Model Association Expedited (EMMAX). RESULTS: The cohort was 35% female, 51% African American, and averaged 68 years of age. There was a 2 beats per minute (bpm) difference in HR by race, AA being slightly higher. Among 43 candidate variants, 4 single nucleotide polymorphisms (SNPs) in one gene (GJA1) were significantly associated with HR. In genome-wide testing, one statistically significant association peak was identified on chromosome 22q13, with strongest SNP rs535263906 (p = 3.3 × 10-8). The peak is located within the gene Cadherin EGF LAG Seven-Pass G-Type Receptor 1 (CELSR1), encoding a cadherin super-family cell surface protein identified in GWAS of other phenotypes (e.g., stroke). The highest associated SNP was specific to the African American population. CONCLUSIONS: These data confirm GJA1 association with HR in the setting of HFrEF and identify novel candidate genes for HR in HFrEF patients, particularly CELSR1. These associations should be tested in additional cohorts.


Asunto(s)
Cadherinas/genética , Conexina 43/genética , Insuficiencia Cardíaca/genética , Frecuencia Cardíaca/genética , Negro o Afroamericano/genética , Anciano , Cromosomas Humanos Par 22/genética , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Insuficiencia Cardíaca/patología , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Volumen Sistólico/genética
2.
Headache ; 58(5): 715-723, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29878340

RESUMEN

OBJECTIVE: To determine the potential impact of erenumab, a human anti-calcitonin gene-related peptide (CGRP) receptor monoclonal antibody, on total exercise time (TET), time to exercise-induced angina, and ST depression in a double-blind, placebo-controlled study in patients with stable angina due to documented coronary artery disease. BACKGROUND: The relative importance of the CGRP receptor pathway during myocardial ischemia has not been established. METHODS: An exercise treadmill test was conducted following a single IV infusion of erenumab 140 mg or placebo. The primary endpoint was the change from baseline in exercise duration as measured by TET with a noninferiority margin of -90 seconds. Safety follow-up visits occurred through week 12. Eighty-eight participants were included in the analysis. RESULTS: LS mean (SE) change in TET was -2.9 [14.8] seconds in the erenumab group and 8.1 [14.4] seconds in placebo; adjusted mean (90% CI) treatment difference was -11.0 (-44.9, 22.9) seconds. The CI lower bound (-44.9 sec) did not reach pre-defined non-inferiority margin of -90 seconds, demonstrating that TET change from baseline in the erenumab group was non-inferior to placebo. There was no difference in time to exercise-induced angina in erenumab and placebo groups (median [90% CI] time of 500 [420, 540] vs 508 [405, 572] seconds; hazard ratio [90% CI]: 1.11 [0.73, 1.69], P = .69) or time to onset of ≥1 mm ST-segment depression (median [90% CI] time of 407 [380, 443] vs 420 [409,480] seconds; hazard ratio [95% CI]: 1.14 [0.76, 1.69], P = .59). Adverse events were reported by 27% and 32% of patients in erenumab and placebo groups. CONCLUSIONS: Erenumab did not adversely affect exercise time in a high cardiovascular risk population of patients, supporting that inhibition of the canonical CGRP receptor does not worsen myocardial ischemia.


Asunto(s)
Angina Estable/inducido químicamente , Angina Estable/fisiopatología , Anticuerpos Monoclonales Humanizados/efectos adversos , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/efectos adversos , Electrocardiografía/efectos de los fármacos , Prueba de Esfuerzo/efectos de los fármacos , Anciano , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/administración & dosificación , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo
3.
Cardiology ; 136(2): 138-144, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27614723

RESUMEN

OBJECTIVES: In the Systolic Heart Failure Treatment with the If Inhibitor Ivabradine Trial (SHIFT), slowing of the heart rate with ivabradine reduced cardiovascular death or heart failure hospitalizations among patients with systolic chronic heart failure (CHF). Subsequently, in the Study Assessing the Morbidity-Mortality Benefits of the If Inhibitor Ivabradine in Patients with Coronary Artery Disease (SIGNIFY) slowing of the heart rate in patients without CHF provided no benefit for cardiovascular death or nonfatal myocardial infarction (primary composite end point), with secondary analyses suggesting possible harm in the angina subgroup. Therefore, we examined the impact of ivabradine in the patients with CHF plus angina in SHIFT. METHODS: SHIFT enrolled adults with stable, symptomatic CHF, a left ventricular ejection fraction ≤35% and a sinus rhythm with a resting heart rate ≥70 bpm. Outcomes were the SHIFT and SIGNIFY primary composite end points and their components. RESULTS: Of 6,505 patients in SHIFT, 2,220 (34%) reported angina at randomization. Ivabradine numerically, but not significantly, reduced the SIGNIFY primary composite end point by 8, 11 and 11% in the SHIFT angina subgroup, nonangina subgroup and overall population, respectively. Ivabradine also reduced the SHIFT primary composite end point in all 3 subgroups. CONCLUSIONS: In SHIFT, ivabradine showed consistent reduction of cardiovascular outcomes in patients with CHF; similar results were seen in the subgroup of SHIFT patients with angina.


Asunto(s)
Angina de Pecho/tratamiento farmacológico , Benzazepinas/uso terapéutico , Fármacos Cardiovasculares/uso terapéutico , Insuficiencia Cardíaca Sistólica/tratamiento farmacológico , Adulto , Anciano , Angina de Pecho/complicaciones , Femenino , Insuficiencia Cardíaca Sistólica/complicaciones , Insuficiencia Cardíaca Sistólica/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Ivabradina , Masculino , Persona de Mediana Edad , Volumen Sistólico
4.
Circulation ; 124(4): 406-15, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21747053

RESUMEN

BACKGROUND: Cardiac overload, a major cause of heart failure, induces the expression of the heat shock protein H11 kinase/Hsp22 (Hsp22). METHODS AND RESULTS: To determine the specific function of Hsp22 in that context, a knockout mouse model of Hsp22 deletion was generated. Although comparable to wild-type mice in basal conditions, knockout mice exposed to pressure overload developed less hypertrophy and showed ventricular dilation, impaired contractile function, increased myocyte length and accumulation of interstitial collagen, faster transition into heart failure, and increased mortality. Microarrays revealed that hearts from knockout mice failed to transactivate genes regulated by the transcription factor STAT3. Accordingly, nuclear STAT3 tyrosine phosphorylation was decreased in knockout mice. Silencing and overexpression experiments in isolated neonatal rat cardiomyocytes showed that Hsp22 activates STAT3 via production of interleukin-6 by the transcription factor nuclear factor-κB. In addition to its transcriptional function, STAT3 translocates to the mitochondria where it increases oxidative phosphorylation. Both mitochondrial STAT3 translocation and respiration were also significantly decreased in knockout mice. CONCLUSIONS: This study found that Hsp22 represents a previously undescribed activator of both nuclear and mitochondrial functions of STAT3, and its deletion in the context of pressure overload in vivo accelerates the transition into heart failure and increases mortality.


Asunto(s)
Eliminación de Gen , Proteínas del Choque Térmico HSP20/genética , Insuficiencia Cardíaca/genética , Mitocondrias Cardíacas/genética , Proteínas Musculares/genética , Factor de Transcripción STAT3/genética , Animales , Cardiomegalia/enzimología , Cardiomegalia/genética , Núcleo Celular/enzimología , Núcleo Celular/genética , Células Cultivadas , Colágeno/metabolismo , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/mortalidad , Proteínas de Choque Térmico , Interleucina-6/biosíntesis , Masculino , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/enzimología , Chaperonas Moleculares , Miocitos Cardíacos/enzimología , FN-kappa B/metabolismo , Fosforilación Oxidativa , Ratas
5.
Circ Res ; 107(5): 615-9, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20634486

RESUMEN

RATIONALE: Increased aortic stiffness, an important feature of many vascular diseases, eg, aging, hypertension, atherosclerosis, and aortic aneurysms, is assumed because of changes in extracellular matrix (ECM). OBJECTIVE: We tested the hypothesis that the mechanisms also involve intrinsic stiffening of vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: Stiffness was measured in vitro both by atomic force microscopy (AFM) and in a reconstituted tissue model, using VSMCs from aorta of young versus old male monkeys (Macaca fascicularis) (n=7/group), where aortic stiffness increases by 200% in vivo. The apparent elastic modulus was increased (P<0.05) in old (41.7+/-0.5 kPa) versus young (12.8+/-0.3 kPa) VSMCs but not after disassembly of the actin cytoskeleton with cytochalasin D. Stiffness of the VSMCs in the reconstituted tissue model was also higher (P<0.05) in old (23.3+/-3.0 kPa) than in young (13.7+/-2.4 kPa). CONCLUSIONS: These data support the novel concept, not appreciated previously, that increased vascular stiffness with aging is attributable not only to changes in ECM but also to intrinsic changes in VSMCs.


Asunto(s)
Envejecimiento/patología , Enfermedades de la Aorta/patología , Músculo Liso Vascular/patología , Actinas/metabolismo , Factores de Edad , Envejecimiento/metabolismo , Animales , Aorta Torácica/patología , Enfermedades de la Aorta/etiología , Células Cultivadas , Citocalasina D/farmacología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Módulo de Elasticidad , Integrina beta1/metabolismo , Macaca fascicularis , Masculino , Microscopía de Fuerza Atómica , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Tubulina (Proteína)/metabolismo , Vimentina/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 300(4): H1303-10, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21317305

RESUMEN

The second window of ischemic preconditioning (SWOP) provides maximal protection against ischemia through regulation of the inducible nitric oxide synthase (iNOS), yet its application is limited by the inconvenience of the preliminary ischemic stimulus required for prophylaxis. Overexpression of H11 kinase/Hsp22 (Hsp22) in a transgenic mouse model provides cardioprotection against ischemia that is equivalent to that conferred by SWOP. We hypothesized that short-term, prophylactic overexpression of Hsp22 would offer an alternative to SWOP in reducing ischemic damage through a nitric oxide (NO)-dependent mechanism. Adeno-mediated overexpression of Hsp22 was achieved in the area at risk of the left circumflex (Cx) coronary artery in chronically instrumented swine and compared with LacZ controls (n = 5/group). Hsp22-injected myocardium showed an average fourfold increase in Hsp22 protein expression compared with controls and a doubling in iNOS expression (both P < 0.05). Four days after ischemia-reperfusion, regional wall thickening was reduced by 58 ± 2% in the Hsp22 group vs. 82 ± 7% in the LacZ group, and Hsp22 reduced infarct size by 40% (both P < 0.05 vs. LacZ). Treatment with the NOS inhibitor N(G)-nitro-L-arginine (L-NNA) before ischemia suppressed the protection induced by Hsp22. In isolated cardiomyocytes, Hsp22 increased iNOS expression through the transcription factors NF-κB and STAT, the same effectors activated by SWOP, and reduced by 60% H(2)O(2)-mediated apoptosis, which was also abolished by NOS inhibitors. Therefore, short-term, prophylactic conditioning by Hsp22 provides NO-dependent cardioprotection that reproduces the signaling of SWOP, placing Hsp22 as a potential alternative for preemptive treatment of myocardial ischemia.


Asunto(s)
Proteínas de Choque Térmico/biosíntesis , Precondicionamiento Isquémico Miocárdico/métodos , Isquemia Miocárdica/prevención & control , Miocardio/enzimología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Animales Modificados Genéticamente , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiología , Proteínas de Choque Térmico/genética , Ratones , Isquemia Miocárdica/enzimología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/fisiología , FN-kappa B/metabolismo , Nitroarginina/farmacología , Proteínas Serina-Treonina Quinasas/genética , Porcinos
7.
Circ Res ; 104(7): 887-95, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19246680

RESUMEN

H11 kinase/Hsp22 (H11K) is a chaperone promoting cardiac cell growth and survival through the activation of Akt, a downstream effector of phosphatidylinositol 3-kinase (PI3K). In this study, we tested whether H11K-induced activation of the PI3K/Akt pathway is mediated by the bone morphogenetic protein (BMP) signaling, both in a transgenic mouse model with cardiac-specific overexpression of H11K and in isolated cardiac myocytes. Microarrays in hearts from transgenic compared to wild-type mice showed an upregulation of the BMP receptors Alk3 and BMPR-II, and of their ligand BMP4 (P<0.01 versus wild type). Activation of the BMP pathway in transgenic mice was confirmed by increased phosphorylation of the "canonical" BMP effectors Smad 1/5/8 (P<0.01 versus wild type). In isolated myocytes, adenovirus-mediated overexpression of H11K was accompanied by a significant (P<0.01) increase in PI3K activity, phospho-Akt, Smad 1/5/8 phosphorylation and [(3)H]phenylalanine incorporation, and by a 70% reduction in H(2)O(2)-mediated apoptosis. All these effects were abolished by the BMP antagonist noggin. In presence of BMP4, Smad 1/5/8 phosphorylation was enhanced by 5-fold on H11K overexpression but decreased by 3-fold on H11K knockdown (P<0.01 versus control), showing that H11K potentiates the BMP signaling. In pull-down experiments, H11K increased both the association of Alk3 and BMPR-II together, and their interaction with the transforming growth factor-beta-activated kinase (TAK)1, a "noncanonical" mediator of the BMP receptor signaling. TAK1 inhibition prevented H11K-mediated activation of Akt. Therefore, potentiation of the BMP receptor by H11K promotes an activation of the PI3K/Akt pathway mediated by TAK1, which dictates the physiological effects of H11K on cardiac cell growth and survival.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Proteínas de Choque Térmico/metabolismo , Miocitos Cardíacos/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Apoptosis , Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Portadoras/metabolismo , Supervivencia Celular , Células Cultivadas , Proteínas de Choque Térmico/genética , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Ratones Transgénicos , Chaperonas Moleculares , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Proteínas Smad Reguladas por Receptores/metabolismo , Transducción Genética
8.
Am J Physiol Heart Circ Physiol ; 299(3): H752-62, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20581088

RESUMEN

A major difference between experimental ischemic preconditioning (IPC), induced by brief ischemic episodes, and the clinical situation is that patients generally have repetitive episodes of ischemia. We used a swine model to examine differences in genes regulated by classical second-window IPC (SWOP) [two 10-min episodes of coronary artery occlusion (CAO) followed by 24 h reperfusion] compared with repetitive CAO/reperfusion (RCO), i.e., two 10-min CAO 12 h apart, and to repetitive coronary stenosis (RCS), six episodes of 90 min coronary stenosis 12 h apart (n = 5/group). All three models reduced infarct size by 60-85%, which was mediated by nitric oxide in SWOP but not in the other two models. We employed microarray analyses to discover additional molecular pathways intrinsic to models of repetitive ischemia and different from classical SWOP. There was an 85% homology in gene response between the RCO and RCS models, whereas SWOP was qualitatively different. Both RCO and RCS, but not SWOP, showed downregulation of genes encoding proteins involved in oxidative metabolism and upregulation of genes involved in protein synthesis, unfolded protein response, autophagy, heat shock response, protein secretion, and an activation of the NF-kappaB signaling pathway. Therefore, the regulated genes mediating IPC with repetitive ischemia differ radically from SWOP both quantitatively and qualitatively, showing that a repetitive pattern of ischemia, rather than the difference between no-flow vs. low-flow ischemia, dictates the genomic response of the heart. These findings illustrate new cardioprotective mechanisms developed by repetitive IPC, which are potentially more relevant to patients with chronic ischemic heart disease, who are subjected to repetitive episodes of ischemia.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Análisis de Varianza , Animales , Western Blotting , Circulación Coronaria/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos
9.
J Am Heart Assoc ; 9(15): e015916, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32750307

RESUMEN

Background In adults with heart failure, elevated heart rate is associated with lower survival. We determined whether an elevated heart rate was associated with an increased risk of death or heart transplant in children with dilated cardiomyopathy. Methods and Results The study is an analysis of the Pediatric Cardiomyopathy Registry and includes baseline data, annual follow-up, and censoring events (transplant or death) in 557 children (51% male, median age 1.8 years) with dilated cardiomyopathy diagnosed between 1994 and 2011. An elevated heart rate was defined as 2 or more SDs above the mean heart rate of children, adjusted for age. The primary outcomes were heart transplant and death. Heart rate was elevated in 192 children (34%), who were older (median age, 2.3 versus 0.9 years; P<0.001), more likely to have heart failure symptoms (83% versus 67%; P<0.001), had worse ventricular function (median fractional shortening z score, -9.7 versus -9.1; P=0.02), and were more often receiving anticongestive therapies (96% versus 86%; P<0.001) than were children with a normal heart rate. Controlling for age, ventricular function, and cardiac medications, an elevated heart rate was independently associated with death (adjusted hazard ratio [HR] 2.6; P<0.001) and with death or transplant (adjusted HR 1.5; P=0.01). Conclusions In children with dilated cardiomyopathy, elevated heart rate was associated with an increased risk of death and cardiac transplant. Further study is warranted into the association of elevated heart rate and disease severity in children with dilated cardiomyopathy and as a potential target of therapy.


Asunto(s)
Cardiomiopatía Dilatada/mortalidad , Frecuencia Cardíaca , Cardiomiopatía Dilatada/fisiopatología , Niño , Preescolar , Femenino , Trasplante de Corazón/estadística & datos numéricos , Humanos , Lactante , Estimación de Kaplan-Meier , Masculino , Modelos de Riesgos Proporcionales , Sistema de Registros , Factores de Riesgo
10.
Am J Physiol Cell Physiol ; 297(4): C928-34, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19625613

RESUMEN

Cytochrome c oxidase (COX) is composed of 13 subunits, of which COX I, II, and III are encoded by a mitochondrial gene. COX I and II function as the main catalytic components, but the function of COX III is unclear. Because myocardial ischemia affects mitochondrial oxidative metabolism, we hypothesized that COX activity and expression would be affected during postischemic cardiomyopathy. This hypothesis was tested in a monkey model following myocardial infarction (MI) and subsequent pacing-induced heart failure (HF). In this model, COX I protein expression was decreased threefold after MI and fourfold after HF (P < 0.05 vs. sham), whereas COX II expression remained unchanged. COX III protein expression increased 5-fold after MI and further increased 10-fold after HF compared with sham (P < 0.05 vs. sham). The physiological impact of COX III regulation was examined in vitro. Overexpression of COX III in mitochondria of HL-1 cells resulted in an 80% decrease in COX I, 60% decrease in global COX activity, 60% decrease in cell viability, and threefold increase in apoptosis (P < 0.05). Oxidative stress induced by H2O2 significantly (P < 0.05) increased COX III expression. H2O2 decreased cell viability by 47 +/- 3% upon overexpression of COX III, but only by 12 +/- 5% in control conditions (P < 0.05). We conclude that ischemic stress in vivo and oxidative stress in vitro lead to upregulation of COX III, followed by downregulation of COX I expression, impaired COX oxidative activity, and increased apoptosis. Therefore, upregulation of COX III may contribute to the increased susceptibility to apoptosis following MI and subsequent HF.


Asunto(s)
Apoptosis/fisiología , Complejo IV de Transporte de Electrones/fisiología , Insuficiencia Cardíaca/metabolismo , Infarto del Miocardio/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Insuficiencia Cardíaca/patología , Macaca fascicularis , Masculino , Ratones , Mitocondrias/fisiología , Células Musculares/fisiología , Infarto del Miocardio/patología , Estrés Oxidativo/fisiología , Subunidades de Proteína/fisiología , Especies Reactivas de Oxígeno/metabolismo
11.
Circulation ; 118(19): 1961-9, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18936329

RESUMEN

BACKGROUND: The hypothesis of the present study was that molecular mechanisms differ markedly when mediating ischemic preconditioning induced by repetitive episodes of ischemia versus classic first- or second-window preconditioning. METHODS AND RESULTS: To test this, chronically instrumented conscious pigs were subjected to either repetitive coronary stenosis (RCS) or a traditional protocol of second-window ischemic preconditioning (SWIPC). Lethal ischemia, induced by 60 minutes of coronary artery occlusion followed by reperfusion, resulted in an infarct size/area at risk of 6+/-3% after RCS and 16+/-3% after SWIPC (both groups P<0.05, less than shams 42+/-4%). Two molecular signatures of SWIPC, the increased expression of the inducible isoform of nitric oxide synthase and the translocation of protein kinase Cepsilon to the plasma membrane, were observed with SWIPC but not with RCS. Confirming this, pretreatment with a nitric oxide synthase inhibitor prevented the protection conferred by SWIPC but not by RCS. Microarray analysis revealed a qualitatively different genomic profile of cardioprotection between ischemic preconditioning induced by RCS and that induced by SWIPC. The number of genes significantly regulated was greater in RCS (5739) than in SWIPC (2394) animals. Of the 5739 genes regulated in RCS, only 31% were also regulated in SWIPC. Broad categories of genes induced by RCS but not SWIPC included those involved in autophagy, endoplasmic reticulum stress, and mitochondrial oxidative metabolism. The upregulation of these pathways was confirmed by Western blotting. CONCLUSIONS: RCS induces cardioprotection against lethal myocardial ischemia that is at least as powerful as traditional ischemic preconditioning but is mediated through radically different mechanisms.


Asunto(s)
Estenosis Coronaria/metabolismo , Estenosis Coronaria/fisiopatología , Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Animales , Estado de Conciencia , Circulación Coronaria , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Aturdimiento Miocárdico/metabolismo , Aturdimiento Miocárdico/fisiopatología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitroarginina/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Quinasa C-epsilon/metabolismo , Recurrencia , Porcinos
12.
Am J Physiol Heart Circ Physiol ; 297(5): H1776-82, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19734365

RESUMEN

Adenylyl cyclase (AC) types 5 and 6 (AC5 and AC6) are the two major AC isoforms expressed in the mammalian heart that mediate signals from beta-adrenergic receptor stimulation. Because of the unavailability of isoform-specific antibodies, it is difficult to ascertain the expression levels of AC5 protein in the heart. Here we demonstrated the successful generation of an AC5 isoform-specific mouse monoclonal antibody and studied the expression of AC5 protein during cardiac development in different mammalian species. The specificity of the antibody was confirmed using heart and brain tissues from AC5 knockout mice and from transgenic mice overexpressing AC5. In mice, the AC5 protein was highest in the brain but was also detectable in all organs studied, including the heart, brain, lung, liver, stomach, kidney, skeletal muscle, and vascular tissues. Western blot analysis showed that AC5 was most abundant in the neonatal heart and declined to basal levels in the adult heart. AC5 protein increased in the heart with pressure-overload left ventricular hypertrophy. Thus this new AC5 antibody demonstrated that this AC isoform behaves similarly to fetal type genes, such as atrial natriuretic peptide; i.e., it declines with development and increases with pressure-overload hypertrophy.


Asunto(s)
Adenilil Ciclasas/metabolismo , Corazón/crecimiento & desarrollo , Hipertrofia Ventricular Izquierda/enzimología , Isoenzimas/metabolismo , Miocardio/enzimología , Adenilil Ciclasas/deficiencia , Adenilil Ciclasas/genética , Adenilil Ciclasas/inmunología , Factores de Edad , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales , Especificidad de Anticuerpos , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Perros , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiopatología , Hipertrofia Ventricular Izquierda/fisiopatología , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Porcinos , Transfección
13.
Cardiovasc Res ; 77(3): 497-505, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18006445

RESUMEN

AIMS: The regulation of protein degradation by the proteasome during cardiac hypertrophy remains largely unknown. Also, the proteasome translocates to the nuclear periphery in response to cellular stress in yeast, which remains unexplored in mammals. The purpose of this study was to determine the quantitative and qualitative adaptation of the proteasome during stable cardiac hypertrophy. METHODS AND RESULTS: We measured proteasome activity, expression and sub-cellular distribution in a model of chronic cardiac hypertrophy induced by the stress-response chaperone H11 Kinase/Hsp22 (Hsp22). Over-expression of Hsp22 in a transgenic (TG) mouse leads to a 30% increase in myocyte cross-sectional area compared to wild-type (WT) mice (P < 0.01). Characterization of the proteasome in hearts from TG mice vs. WT revealed an increased expression of both 19S and 20S subunits (P < 0.05), a doubling in 20S catalytic activity (P < 0.01), a redistribution of both subunits from the cytosol to the nuclear periphery, and a four-fold increase in nuclear-associated 20S catalytic activity (P < 0.001). The perinuclear proteasome co-localized and interacted with Hsp22. Inhibition of proteasome activity by epoxomicin reduced hypertrophy in TG by 50% (P < 0.05). Adeno-mediated over-expression of Hsp22 in isolated cardiac myocytes increased both cell growth and proteasome activity, and both were prevented upon inhibition of the proteasome. Similarly, stimulation of cardiac cell growth by pro-hypertrophic stimuli increased Hsp22 expression and proteasome activity, and proteasome inhibition in that setting prevented hypertrophy. Proteasome inhibitors also prevented the increase in rate of protein synthesis observed after over-expression of Hsp22 or upon addition of pro-hypertrophic stimuli. CONCLUSIONS: Hsp22-mediated cardiac hypertrophy promotes an increased expression and activity, and a subcellular redistribution of the proteasome. Inhibition of the proteasome reverses cardiac hypertrophy upon Hsp22 over-expression or upon stimulation by pro-hypertrophic hormones, and also blocks the stimulation of protein synthesis in these conditions.


Asunto(s)
Cardiomegalia/etiología , Proteínas del Choque Térmico HSP20/fisiología , Proteínas Musculares/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Animales , Cardiomegalia/enzimología , Cardiomegalia/prevención & control , Proliferación Celular , Activación Enzimática , Proteínas de Choque Térmico , Ratones , Ratones Transgénicos , Chaperonas Moleculares , Inhibidores de Proteasoma
14.
Free Radic Biol Med ; 137: 194-200, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31047988

RESUMEN

H11 kinase/Hsp22 (Hsp22) is a small heat shock protein, which, when overexpressed cardiac specifically in transgenic (TG) mice, induces stable left ventricular (LV) hypertrophy. Hsp22 also increases oxidative phosphorylation and mitochondrial reactive oxygen species (ROS) production, mechanisms mediating LV hypertrophy, senescence and reduced lifespan. Therefore, we investigated whether ROS production mediates LV hypertrophy, senescence and reduced life span in Hsp22 TG mice. Survival curves revealed that TG mice had a 48% reduction in their mean life span compared to wild type (WT) mice. This was associated with a significant increase in senescence markers, such as p16, p19 mRNA levels as well as the percentage of ß-galactosidase positive cells and telomerase activity. Oxidized (GSSG)/reduced (GSH) glutathione ratio, an indicator of oxidative stress, and ROS production from 3 major cellular sources was measured in cardiac tissue. Hearts from TG mice exhibited a decrease in GSH/GSSG ratio together with increased ROS production from all sources. To study the role of ROS, mice were treated with the antioxidant Tempol from weaning to their sacrifice. Chronic Tempol treatment abolished oxidative stress and overproduction of ROS, and reduced myocardial hypertrophy and Akt phosphorylation in TG mice. Tempol also significantly extended life span and prevented aging markers in TG mice. Taken together these results show that overexpression of Hsp22 increases oxidative stress responsible for the induction of hypertrophy and senescence and ultimately reduction in life span.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Mitocondrias/metabolismo , Chaperonas Moleculares/metabolismo , Miocardio/patología , Animales , Antioxidantes/administración & dosificación , Células Cultivadas , Senescencia Celular , Óxidos N-Cíclicos/administración & dosificación , Proteínas de Choque Térmico/genética , Hipertrofia Ventricular Izquierda/genética , Longevidad , Masculino , Ratones , Ratones Transgénicos , Chaperonas Moleculares/genética , Miocardio/metabolismo , Estrés Oxidativo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Marcadores de Spin
15.
Circulation ; 116(6): 669-76, 2007 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-17664374

RESUMEN

BACKGROUND: Our hypothesis was that the changes in vascular properties responsible for aortic stiffness with aging would be greater in old male monkeys than old female monkeys. METHODS AND RESULTS: We analyzed the effects of gender differences in aging on in vivo measurements of aortic pressure and diameter and on extracellular matrix of the thoracic aorta in young adult (age, 6.6+/-0.5 years) versus old adult (age, 21.2+/-0.2 years) monkeys (Macaca fascicularis). Aortic stiffness, as represented by the pressure strain elastic modulus (Ep), increased more in old male monkeys (5.08+/-0.81; P<0.01) than in old females (3.06+/-0.52). In both genders, collagen density was maintained, collagen-bound glycation end products increased, and collagen type 1 decreased. However, elastin density decreased significantly (from 22+/-1.5% to 15+/-1.2%) with aging (P<0.05) only in males. Furthermore, only old males were characterized by a decrease (P<0.05) in collagen type 3 (an isoform that promotes elasticity) and an increase in collagen type 8 (an isoform that promotes the neointimal migration of vascular smooth muscle cells). In contrast to the data in monkeys, collagen types 1 and 3 both increased significantly in aging rats. CONCLUSIONS: There are major species differences in the effects of aging on aortic collagen types 1 and 3. Furthermore, because alterations in collagen density, collagen content, hydroxyproline, and collagen advanced glycation end products were similar in both old male and female monkeys, these factors cannot be responsible for the greater increase in stiffness in old males. However, changes in collagen isoforms and the decrease in elastin observed only in old males likely account for the greater increase in aortic stiffness.


Asunto(s)
Envejecimiento/fisiología , Aorta Torácica/patología , Aorta Torácica/fisiología , Estenosis de la Válvula Aórtica/patología , Caracteres Sexuales , Animales , Estenosis de la Válvula Aórtica/fisiopatología , Presión Sanguínea/fisiología , Colágeno/fisiología , Femenino , Humanos , Macaca fascicularis , Masculino
16.
Circ Res ; 98(2): 280-8, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16373598

RESUMEN

Ischemic preconditioning confers powerful protection against myocardial infarction through pre-emptive activation of survival signaling pathways, but it remains difficult to apply to patients with ischemic heart disease, and its effects are transient. Promoting a sustained activation of preconditioning mechanisms in vivo would represent a novel approach of cardioprotection. We tested the role of the protein H11 kinase (H11K), which accumulates by 4- to 6-fold in myocardium of patients with chronic ischemic heart disease and in experimental models of ischemia. This increased expression was quantitatively reproduced in cardiac myocytes using a transgenic (TG) mouse model. After 45 minutes of coronary artery occlusion and reperfusion, hearts from TG mice showed an 82+/-5% reduction in infarct size compared with wild-type (WT), which was similar to the 84+/-4% reduction of infarct size observed in WT after a protocol of ischemic preconditioning. Hearts from TG mice showed significant activation of survival kinases participating in preconditioning, including Akt and the 5'AMP-activated protein kinase (AMPK). H11K directly binds to both Akt and AMPK and promotes their nuclear translocation and their association in a multiprotein complex, which results in a stimulation of survival mechanisms in cytosol and nucleus, including inhibition of proapoptotic effectors (glycogen synthase kinase-3beta, Bad, and Foxo), activation of antiapoptotic effectors (protein kinase Cepsilon, endothelial and inducible NO synthase isoforms, and heat shock protein 70), increased expression of the hypoxia-inducible factor-1alpha, and genomic switch to glucose utilization. Therefore, activation of survival pathways by H11K preemptively triggers the antiapoptotic and metabolic response to ischemia and is sufficient to confer cardioprotection in vivo equally potent to preconditioning.


Asunto(s)
Proteínas del Choque Térmico HSP20/fisiología , Precondicionamiento Isquémico Miocárdico , Proteínas Musculares/fisiología , Infarto del Miocardio/prevención & control , Proteínas Quinasas Activadas por AMP , Animales , Apoptosis , Supervivencia Celular , Células Cultivadas , Citoprotección , Proteínas de Choque Térmico , Ratones , Ratones Transgénicos , Chaperonas Moleculares , Complejos Multienzimáticos/metabolismo , Miocitos Cardíacos/citología , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Expert Rev Cardiovasc Ther ; 16(7): 515-526, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29902387

RESUMEN

INTRODUCTION: This review summarizes the current management of heart failure (HF) in patients with reduced ejection fraction and the potential role of heart rate lowering agents in select populations, as recommended in the updated guidelines. Areas covered: PubMed was searched for studies that evaluated the role of heart rate lowering or ivabradine in HF management. Expert commentary: Targeting heart rate may offer benefit when added to renin-angiotensin aldosterone antagonists, and beta-blockers. Ivabradine is a heart rate lowering agent that acts on the funny current (If) in the sinoatrial node, thereby reducing heart rate without directly affecting cardiac contraction and relaxation. Clinical data from a phase III trial demonstrated that ivabradine reduced the composite end point of cardiovascular death or hospital admission for worsening systolic HF, while maintaining an acceptable safety profile in patients receiving standard of care therapy. These data, in addition to more recently published guidelines, suggest ivabradine as a promising new treatment option for lowering heart rate after optimizing standard therapy in select patients with chronic HF.


Asunto(s)
Benzazepinas/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Frecuencia Cardíaca/efectos de los fármacos , Antagonistas Adrenérgicos beta/uso terapéutico , Fármacos Cardiovasculares/uso terapéutico , Enfermedad Crónica , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca Sistólica/tratamiento farmacológico , Hospitalización , Humanos , Ivabradina , Resultado del Tratamiento , Disfunción Ventricular Izquierda/tratamiento farmacológico
18.
Physiol Genomics ; 29(2): 169-80, 2007 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-17456900

RESUMEN

Although increased vascular stiffness is more prominent in aging males than females, and males are more prone to vascular disease with aging, no study has investigated the genes potentially responsible for sex differences in vascular aging. We tested the hypothesis that the transcriptional adaptation to aging differs in males and females using a monkey model, which is not only physiologically and phylogenetically closer to humans than the more commonly studied rodent models but also is not afflicted with the most common forms of vascular disease that accompany the aging process in humans, e.g., atherosclerosis, hypertension, and diabetes. The transcriptional profile of the aorta was compared by high-density microarrays between young and old males or females (n = 6/group). About 600 genes were expressed differentially when comparing old versus young animals. Surprisingly, <5% of these genes were shared between males and females. Radical differences between sexes were especially apparent for genes regulating the extracellular matrix, which relates to stiffness. Aging males were also more prone than females to genes switching smooth muscle cells from the "contractile" to "secretory" phenotype. Other sex differences involved genes participating in DNA repair, stress response, and cell signaling. Therefore, major differences of gene regulation exist between males and females in vascular aging, which may underlie the physiological differences characterizing aging arteries in males and females. Furthermore, the analyses in young monkeys demonstrated differences in genes regulating vascular structure, implying that the sex differences in vascular stiffness that develop with aging are programmed at an early age.


Asunto(s)
Envejecimiento/fisiología , Aorta/metabolismo , Regulación de la Expresión Génica/fisiología , Caracteres Sexuales , Envejecimiento/metabolismo , Animales , Biología Computacional , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Macaca fascicularis , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Circulation ; 114(17): 1821-8, 2006 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-17043166

RESUMEN

BACKGROUND: The adaptation of cardiac mass to hemodynamic overload requires an adaptation of protein turnover, ie, the balance between protein synthesis and degradation. We tested 2 hypotheses: (1) chronic left ventricular hypertrophy (LVH) activates the proteasome system of protein degradation, especially in the myocardium submitted to the highest wall stress, ie, the subendocardium, and (2) the proteasome system is required for the development of LVH. METHODS AND RESULTS: Gene and protein expression of proteasome subunits and proteasome activity were measured separately from left ventricular subendocardium and subepicardium, right ventricle, and peripheral tissues in a canine model of severe, chronic (2 years) LVH induced by aortic banding and then were compared with controls. Both gene and protein expressions of proteasome subunits were increased in LVH versus control (P<0.05), which was accompanied by a significant (P<0.05) increase in proteasome activity. Posttranslational modification of the proteasome was also detected by 2-dimensional gel electrophoresis. These changes were found specifically in left ventricular subendocardium but not in left ventricular subepicardium, right ventricle, or noncardiac tissues from the same animals. In a mouse model of chronic pressure overload, a 50% increase in heart mass and a 2-fold increase in proteasome activity (both P<0.05 versus sham) were induced. In that model, the proteasome inhibitor epoxomicin completely prevented LVH while blocking proteasome activation. CONCLUSIONS: The increase in proteasome expression and activity found during chronic pressure overload in myocardium submitted to higher stress is also required for the establishment of LVH.


Asunto(s)
Hipertrofia Ventricular Izquierda/metabolismo , Proteínas Musculares/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Remodelación Ventricular/fisiología , Adaptación Fisiológica , Animales , Aorta Torácica , Modelos Animales de Enfermedad , Perros , Electroforesis en Gel Bidimensional , Femenino , Perfilación de la Expresión Génica , Ligadura , Masculino , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Oligopéptidos/farmacología , Poliubiquitina/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Subunidades de Proteína , Estrés Fisiológico/metabolismo
20.
Circ Res ; 97(3): 219-26, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-16037571

RESUMEN

Endocardial cushions are precursors of mature atrioventricular (AV) valves. Their formation is induced by signaling molecules originating from the AV myocardium, including bone morphogenetic proteins (BMPs). Here, we hypothesized that BMP signaling plays an important role in the AV myocardium during the maturation of AV valves from the cushions. To test our hypothesis, we used a unique Cre/lox system to target the deletion of a floxed Alk3 allele, the type IA receptor for BMPs, to cardiac myocytes of the AV canal (AVC). Lineage analysis indicated that cardiac myocytes of the AVC contributed to the tricuspid mural and posterior leaflets, the mitral septal leaflet, and the atrial border of the annulus fibrosus. When Alk3 was deleted in these cells, defects were seen in the same leaflets, ie, the tricuspid mural leaflet and mitral septal leaflet were longer, the tricuspid posterior leaflet was displaced and adherent to the ventricular wall, and the annulus fibrosus was disrupted resulting in ventricular preexcitation. The defects seen in mice with AVC-targeted deletion of Alk3 provide strong support for a role of Alk3 in human congenital heart diseases, such as Ebstein's anomaly. In conclusion, our mouse model demonstrated critical roles for Alk3 signaling in the AV myocardium during the development of AV valves and the annulus fibrosus.


Asunto(s)
Válvulas Cardíacas/embriología , Corazón/embriología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Proteínas de Unión al ADN/genética , Factor de Transcripción GATA6 , Válvulas Cardíacas/anomalías , Integrasas/genética , Ratones , Ratones Transgénicos , Miocitos Cardíacos/fisiología , Transducción de Señal , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA