Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L748-L759, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28258105

RESUMEN

Right ventricular (RV) dysfunction is associated with numerous smoking-related illnesses, including chronic obstructive pulmonary disease (COPD), in which it is present even in the absence of pulmonary hypertension. It is unknown whether exposure to cigarette smoke (CS) has direct effects on RV function and cardiac fibroblast (CF) proliferation or collagen synthesis. In this study, we evaluated cardiac function and fibrosis in mice exposed to CS and determined mechanisms of smoke-induced changes in CF signaling and fibrosis. AKR mice were exposed to CS for 6 wk followed by echocardiography and evaluation of cardiac hypertrophy, collagen content, and pulmonary muscularization. Proliferation and collagen content were evaluated in primary isolated rat CFs exposed to CS extract (CSE) or nicotine. Markers of cell proliferation, fibrosis, and proliferative signaling were determined by immunoblot or Sircol collagen assay. Mice exposed to CS had significantly decreased RV function, as determined by tricuspid annular plane systolic excursion. There were no changes in left ventricular parameters. RV collagen content was significantly elevated, but there was no change in RV hypertrophy or pulmonary vascular muscularization. CSE directly increased CF proliferation and collagen content in CF. Nicotine alone reproduced these effects. CSE and nicotine-induced fibroblast proliferation and collagen content were mediated through α7 nicotinic acetylcholine receptors and were dependent on PKC-α, PKC-δ, and reduced p38-MAPK phosphorylation. CS and nicotine have direct effects on CFs to induce proliferation and fibrosis, which may negatively affect right heart function.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/patología , Ventrículos Cardíacos/patología , Miocardio/patología , Fumar/efectos adversos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/diagnóstico por imagen , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos AKR , Nicotina/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-delta/metabolismo , Ratas Sprague-Dawley , Remodelación Vascular/efectos de los fármacos , Disfunción Ventricular Derecha/complicaciones , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/patología , Disfunción Ventricular Derecha/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Circ J ; 79(3): 470-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25746522

RESUMEN

Despite the growing number of patients affected, the understanding of diastolic dysfunction and heart failure with preserved ejection fraction (HFpEF) is still poor. Clinical trials, largely based on successful treatments for systolic heart failure, have been disappointing, suggesting that HFpEF has a different pathology to that of systolic dysfunction. In this review, general concepts, epidemiology, diagnosis, and treatment of diastolic dysfunction are summarized, with an emphasis on new experiments suggesting that oxidative stress plays a crucial role in the pathogenesis of at least some forms of the disease. This observation has lead to potential new diagnostics and therapeutics for diastolic dysfunction and heart failure caused by diastolic dysfunction.


Asunto(s)
Diástole , Insuficiencia Cardíaca , Estrés Oxidativo , Volumen Sistólico , Animales , Ensayos Clínicos como Asunto , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos
3.
Circ Res ; 110(6): 841-50, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22343711

RESUMEN

RATIONALE: Previously, we demonstrated that a deoxycorticosterone acetate (DOCA)-salt hypertensive mouse model produces cardiac oxidative stress and diastolic dysfunction with preserved systolic function. Oxidative stress has been shown to increase late inward sodium current (I(Na)), reducing the net cytosolic Ca(2+) efflux. OBJECTIVE: Oxidative stress in the DOCA-salt model may increase late I(Na), resulting in diastolic dysfunction amenable to treatment with ranolazine. METHODS AND RESULTS: Echocardiography detected evidence of diastolic dysfunction in hypertensive mice that improved after treatment with ranolazine (E/E':sham, 31.9 ± 2.8, sham+ranolazine, 30.2 ± 1.9, DOCA-salt, 41.8 ± 2.6, and DOCA-salt+ranolazine, 31.9 ± 2.6; P=0.018). The end-diastolic pressure-volume relationship slope was elevated in DOCA-salt mice, improving to sham levels with treatment (sham, 0.16 ± 0.01 versus sham+ranolazine, 0.18 ± 0.01 versus DOCA-salt, 0.23 ± 0.2 versus DOCA-salt+ranolazine, 0.17 ± 0.0 1 mm Hg/L; P<0.005). DOCA-salt myocytes demonstrated impaired relaxation, τ, improving with ranolazine (DOCA-salt, 0.18 ± 0.02, DOCA-salt+ranolazine, 0.13 ± 0.01, sham, 0.11 ± 0.01, sham+ranolazine, 0.09 ± 0.02 seconds; P=0.0004). Neither late I(Na) nor the Ca(2+) transients were different from sham myocytes. Detergent extracted fiber bundles from DOCA-salt hearts demonstrated increased myofilament response to Ca(2+) with glutathionylation of myosin binding protein C. Treatment with ranolazine ameliorated the Ca(2+) response and cross-bridge kinetics. CONCLUSIONS: Diastolic dysfunction could be reversed by ranolazine, probably resulting from a direct effect on myofilaments, indicating that cardiac oxidative stress may mediate diastolic dysfunction through altering the contractile apparatus.


Asunto(s)
Acetanilidas/farmacología , Calcio/metabolismo , Diástole/efectos de los fármacos , Insuficiencia Cardíaca Diastólica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miofibrillas/efectos de los fármacos , Piperazinas/farmacología , Acetanilidas/sangre , Animales , Desoxicorticosterona/toxicidad , Diástole/fisiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca Diastólica/inducido químicamente , Insuficiencia Cardíaca Diastólica/fisiopatología , Ratones , Mineralocorticoides/toxicidad , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Miofibrillas/metabolismo , Estrés Oxidativo/fisiología , Piperazinas/sangre , Ranolazina , Sodio/metabolismo , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/fisiopatología
4.
J Mol Cell Cardiol ; 54: 25-34, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23123323

RESUMEN

Cardiomyopathy is associated with cardiac Na(+) channel downregulation that may contribute to arrhythmias. Previously, we have shown that elevated intracellular NADH causes a decrease in cardiac Na(+) current (I(Na)) signaled by an increase in mitochondrial reactive oxygen species (ROS). In this study, we tested whether the NADH-mitochondria ROS pathway was involved in the reduction of I(Na) in a nonischemic cardiomyopathic model and correlated the findings with myopathic human hearts. Nonischemic cardiomyopathy was induced in C57BL/6 mice by hypertension after unilateral nephrectomy, deoxycorticosterone acetate (DOCA) pellet implantation, and salt water substitution. Sham operated mice were used as controls. After six weeks, heart tissue and ventricular myocytes isolated from mice were utilized for whole cell patch clamp recording, NADH/NAD(+) level measurements, and mitochondrial ROS monitoring with confocal microscopy. Human explanted hearts were studied using optical mapping. Compared to the sham mice, the arterial blood pressure was higher, the left ventricular volume was significantly enlarged (104.7±3.9 vs. 87.9±6.1 µL, P<0.05), and the ejection fraction was reduced (37.1±1.8% vs. 49.4±3.7%, P<0.05) in DOCA mice. Both the whole cell and cytosolic NADH level were increased (279±70% and 123±2% of sham, respectively, P<0.01), I(Na) was decreased (60±10% of sham, P<0.01), and mitochondrial ROS overproduction was observed (2.9±0.3-fold of sham, P<0.01) in heart tissue and myocytes of myopathic mice vs. sham. Treatment of myocytes with NAD(+) (500 µM), mitoTEMPO (10 µM), chelerythrine (50 µM), or forskolin (5 µM) restored I(Na) back to the level of sham. Injection of NAD(+) (100mg/kg) or mitoTEMPO (0.7 mg/kg) twice (at 24h and 1h before myocyte isolation) to animals also restored I(Na). All treatments simultaneously reduced mitochondrial ROS levels to that of controls. CD38 was found to transduce the extracellular NAD(+) signal. Correlating with the mouse model, failing human hearts showed a reduction in conduction velocity that improved with NAD(+). Nonischemic cardiomyopathy was associated with elevated NADH level, PKC activation, mitochondrial ROS overproduction, and a concomitant decrease in I(Na). Reducing mitochondrial ROS by application of NAD(+), mitoTEMPO, PKC inhibitors, or PKA activators, restored I(Na). NAD(+) improved conduction velocity in human myopathic hearts.


Asunto(s)
Cardiomiopatías/metabolismo , Mitocondrias Cardíacas/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Benzofenantridinas/farmacología , Colforsina/farmacología , Regulación hacia Abajo , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Técnicas In Vitro , Glicoproteínas de Membrana/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , NAD/metabolismo , NAD/farmacología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Compuestos Organofosforados/farmacología , Estrés Oxidativo , Técnicas de Placa-Clamp , Piperidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo
5.
J Mol Cell Cardiol ; 56: 44-54, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23247392

RESUMEN

Despite the increasing prevalence of heart failure with preserved left ventricular function, there are no specific treatments, partially because the mechanism of impaired relaxation is incompletely understood. Evidence indicates that cardiac relaxation may depend on nitric oxide (NO), generated by NO synthase (NOS) requiring the co-factor tetrahydrobiopterin (BH(4)). Recently, we reported that hypertension-induced diastolic dysfunction was accompanied by cardiac BH(4) depletion, NOS uncoupling, a depression in myofilament cross-bridge kinetics, and S-glutathionylation of myosin binding protein C (MyBP-C). We hypothesized that the mechanism by which BH(4) ameliorates diastolic dysfunction is by preventing glutathionylation of MyBP-C and thus reversing changes of myofilament properties that occur during diastolic dysfunction. We used the deoxycorticosterone acetate (DOCA)-salt mouse model, which demonstrates mild hypertension, myocardial oxidative stress, and diastolic dysfunction. Mice were divided into two groups that received control diet and two groups that received BH(4) supplement for 7days after developing diastolic dysfunction at post-operative day 11. Mice were assessed by echocardiography. Left ventricular papillary detergent-extracted fiber bundles were isolated for simultaneous determination of force and ATPase activity. Sarcomeric protein glutathionylation was assessed by immunoblotting. DOCA-salt mice exhibited diastolic dysfunction that was reversed after BH(4) treatment. Diastolic sarcomere length (DOCA-salt 1.70±0.01 vs. DOCA-salt+BH(4) 1.77±0.01µm, P<0.001) and relengthening (relaxation constant, τ, DOCA-salt 0.28±0.02 vs. DOCA-salt+BH(4) 0.08±0.01, P<0.001) were also restored to control by BH(4) treatment. pCa(50) for tension increased in DOCA-salt compared to sham but reverted to sham levels after BH(4) treatment. Maximum ATPase rate and tension cost (ΔATPase/ΔTension) decreased in DOCA-salt compared to sham, but increased after BH(4) treatment. Cardiac MyBP-C glutathionylation increased in DOCA-salt compared to sham, but decreased with BH(4) treatment. MyBP-C glutathionylation correlated with the presence of diastolic dysfunction. Our results suggest that by depressing S-glutathionylation of MyBP-C, BH(4) ameliorates diastolic dysfunction by reversing a decrease in cross-bridge turnover kinetics. These data provide evidence for modulation of cardiac relaxation by post-translational modification of myofilament proteins.


Asunto(s)
Biopterinas/análogos & derivados , Fármacos Cardiovasculares/administración & dosificación , Insuficiencia Cardíaca Diastólica/tratamiento farmacológico , Miofibrillas/fisiología , Adenosina Trifosfatasas/metabolismo , Administración Oral , Animales , Biopterinas/administración & dosificación , Proteínas Portadoras/metabolismo , Células Cultivadas , Desoxicorticosterona/farmacología , Diástole/efectos de los fármacos , Suplementos Dietéticos , Glutatión/metabolismo , Insuficiencia Cardíaca Diastólica/diagnóstico por imagen , Insuficiencia Cardíaca Diastólica/fisiopatología , Ratones , Miofibrillas/efectos de los fármacos , Miofibrillas/enzimología , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Volumen Sistólico/efectos de los fármacos , Ultrasonografía
6.
J Mol Cell Cardiol ; 52(2): 454-63, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21978629

RESUMEN

Cardiac arrhythmias can cause sudden cardiac death (SCD) and add to the current heart failure (HF) health crisis. Nevertheless, the pathological processes underlying arrhythmias are unclear. Arrhythmic conditions are associated with systemic and cardiac oxidative stress caused by reactive oxygen species (ROS). In excitable cardiac cells, ROS regulate both cellular metabolism and ion homeostasis. Increasing evidence suggests that elevated cellular ROS can cause alterations of the cardiac sodium channel (Na(v)1.5), abnormal Ca(2+) handling, changes of mitochondrial function, and gap junction remodeling, leading to arrhythmogenesis. This review summarizes our knowledge of the mechanisms by which ROS may cause arrhythmias and discusses potential therapeutic strategies to prevent arrhythmias by targeting ROS and its consequences. This article is part of a Special Issue entitled "Local Signaling in Myocytes".


Asunto(s)
Arritmias Cardíacas/metabolismo , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Antiarrítmicos/farmacología , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Calcio/metabolismo , Humanos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Oxidación-Reducción/efectos de los fármacos , Canales de Potasio/metabolismo
7.
Circulation ; 124(10): 1124-31, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21859973

RESUMEN

BACKGROUND: Human heart failure is associated with decreased cardiac voltage-gated Na+ channel current (encoded by SCN5A), and the changes have been implicated in the increased risk of sudden death in heart failure. Nevertheless, the mechanism of SCN5A downregulation is unclear. A number of human diseases are associated with alternative mRNA splicing, which has received comparatively little attention in the study of cardiac disease. Splicing factor expression profiles during human heart failure and a specific splicing pathway for SCN5A regulation were explored in this study. METHODS AND RESULTS: Gene array comparisons between normal human and heart failure tissues demonstrated that 17 splicing factors, associated with all major spliceosome components, were upregulated. Two of these splicing factors, RBM25 and LUC7L3, were elevated in human heart failure tissue and mediated truncation of SCN5A mRNA in both Jurkat cells and human embryonic stem cell-derived cardiomyocytes. RBM25/LUC7L3-mediated abnormal SCN5A mRNA splicing reduced Na+ channel current 91.1±9.3% to a range known to cause sudden death. Overexpression of either splicing factor resulted in an increase in truncated mRNA and a concomitant decrease in the full-length SCN5A transcript. CONCLUSIONS: Of the 17 mRNA splicing factors upregulated in heart failure, RBM25 and LUC7L3 were sufficient to explain the increase in truncated forms and the reduction in full-length Na+ channel transcript. Because the reduction in channels was in the range known to be associated with sudden death, interruption of this abnormal mRNA processing may reduce arrhythmic risk in heart failure.


Asunto(s)
Insuficiencia Cardíaca/genética , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Canales de Sodio/genética , Adulto , Anciano , Células Cultivadas , Regulación hacia Abajo , Células Madre Embrionarias/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Células Jurkat , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Proteínas Nucleares , Empalmosomas/metabolismo , Regulación hacia Arriba , Adulto Joven
8.
J Am Heart Assoc ; 11(11): e025295, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35656993

RESUMEN

Background cMyBP-C (Cardiac myosin binding protein-C) regulates cardiac contraction and relaxation. Previously, we demonstrated that elevated myocardial S-glutathionylation of cMyBP-C correlates with diastolic dysfunction (DD) in animal models. In this study, we tested whether circulating S-glutathionylated cMyBP-C would be a biomarker for DD. Methods and Results Humans, African Green monkeys, and mice had DD determined by echocardiography. Blood samples were acquired and analyzed for S-glutathionylated cMyBP-C by immunoprecipitation. Circulating S-glutathionylated cMyBP-C in human participants with DD (n=24) was elevated (1.46±0.13-fold, P=0.014) when compared with the non-DD controls (n=13). Similarly, circulating S-glutathionylated cMyBP-C was upregulated by 2.13±0.47-fold (P=0.047) in DD monkeys (n=6), and by 1.49 (1.22-2.06)-fold (P=0.031) in DD mice (n=5) compared with the respective non-DD controls. Circulating S-glutathionylated cMyBP-C was positively correlated with DD in humans. Conclusions Circulating S-glutathionylated cMyBP-C was elevated in humans, monkeys, and mice with DD. S-glutathionylated cMyBP-C may represent a novel biomarker for the presence of DD.


Asunto(s)
Proteínas Portadoras/análisis , Cardiopatías , Animales , Biomarcadores , Proteínas Portadoras/metabolismo , Chlorocebus aethiops , Diástole/fisiología , Cardiopatías/metabolismo , Humanos , Ratones , Contracción Miocárdica , Miocardio/metabolismo , Fosforilación
9.
Cardiovasc Res ; 118(16): 3211-3224, 2022 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-35018410

RESUMEN

AIMS: Pulmonary arterial hypertension (PAH) is a fatal disease without a cure. Previously, we found that transcription factor RUNX1-dependent haematopoietic transformation of endothelial progenitor cells may contribute to the pathogenesis of PAH. However, the therapeutic potential of RUNX1 inhibition to reverse established PAH remains unknown. In the current study, we aimed to determine whether RUNX1 inhibition was sufficient to reverse Sugen/hypoxia (SuHx)-induced pulmonary hypertension (PH) in rats. We also aimed to demonstrate possible mechanisms involved. METHODS AND RESULTS: We administered a small molecule specific RUNX1 inhibitor Ro5-3335 before, during, and after the development of SuHx-PH in rats to investigate its therapeutic potential. We quantified lung macrophage recruitment and activation in vivo and in vitro in the presence or absence of the RUNX1 inhibitor. We generated conditional VE-cadherin-CreERT2; ZsGreen mice for labelling adult endothelium and lineage tracing in the SuHx-PH model. We also generated conditional Cdh5-CreERT2; Runx1(flox/flox) mice to delete Runx1 gene in adult endothelium and LysM-Cre; Runx1(flox/flox) mice to delete Runx1 gene in cells of myeloid lineage, and then subjected these mice to SuHx-PH induction. RUNX1 inhibition in vivo effectively prevented the development, blocked the progression, and reversed established SuHx-induced PH in rats. RUNX1 inhibition significantly dampened lung macrophage recruitment and activation. Furthermore, lineage tracing with the inducible VE-cadherin-CreERT2; ZsGreen mice demonstrated that a RUNX1-dependent endothelial to haematopoietic transformation occurred during the development of SuHx-PH. Finally, tissue-specific deletion of Runx1 gene either in adult endothelium or in cells of myeloid lineage prevented the mice from developing SuHx-PH, suggesting that RUNX1 is required for the development of PH. CONCLUSION: By blocking RUNX1-dependent endothelial to haematopoietic transformation and pulmonary macrophage recruitment and activation, targeting RUNX1 may be as a novel treatment modality for pulmonary arterial hypertension.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Ratas , Ratones , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/genética , Hipertensión Pulmonar Primaria Familiar , Hipoxia/complicaciones , Arteria Pulmonar , Modelos Animales de Enfermedad
10.
Am J Physiol Heart Circ Physiol ; 301(3): H824-31, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21724869

RESUMEN

Diastolic heart failure is a major cause of mortality in the elderly population. It is often preceded by diastolic dysfunction, which is characterized by impaired active relaxation and increased stiffness. We tested the hypothesis that senescence-prone (SAMP8) mice would develop diastolic dysfunction compared with senescence-resistant controls (SAMR1). Pulsed-wave Doppler imaging of the ratio of blood flow velocity through the mitral valve during early (E) vs. late (A) diastole was reduced from 1.3 ± 0.03 in SAMR1 mice to 1.2 ± 0.03 in SAMP8 mice (P < 0.05). Tissue Doppler imaging of the early (E') and late (A') diastolic mitral annulus velocities found E' reduced from 25.7 ± 0.9 mm/s in SAMR1 to 21.1 ± 0.8 mm/s in SAMP8 mice and E'/A' similarly reduced from 1.1 ± 0.02 to 0.8 ± 0.03 in SAMR1 vs. SAMP8 mice, respectively (P < 0.05). Invasive hemodynamics revealed an increased slope of the end-diastolic pressure-volume relationship (0.5 ± 0.05 vs. 0.8 ± 0.14; P < 0.05), indicating increased left ventricular chamber stiffness. There were no differences in systolic function or mean arterial pressure; however, diastolic dysfunction was accompanied by increased fibrosis in the hearts of SAMP8 mice. In SAMR1 vs. SAMP8 mice, interstitial collagen area increased from 0.3 ± 0.04 to 0.8 ± 0.09% and perivascular collagen area increased from 1.0 ± 0.11 to 1.6 ± 0.14%. Transforming growth factor-ß and connective tissue growth factor gene expression were increased in the hearts of SAMP8 mice (P < 0.05 for all data). In summary, SAMP8 mice show increased fibrosis and diastolic dysfunction similar to those seen in humans with aging and may represent a suitable model for future mechanistic studies.


Asunto(s)
Envejecimiento Prematuro/complicaciones , Envejecimiento , Insuficiencia Cardíaca/etiología , Miocardio/patología , Disfunción Ventricular Izquierda/etiología , Función Ventricular Izquierda , Factores de Edad , Envejecimiento/genética , Envejecimiento Prematuro/genética , Análisis de Varianza , Animales , Cateterismo Cardíaco , Colágeno/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Diástole , Modelos Animales de Enfermedad , Ecocardiografía Doppler de Pulso , Elasticidad , Fibrosis , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Hemodinámica , Ratones , Ratones Transgénicos , Miocardio/metabolismo , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda/genética , Presión Ventricular
11.
Aging (Albany NY) ; 12(24): 25939-25955, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33378745

RESUMEN

Adult hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow (BM) ensuring homeostasis of blood production and immune response throughout life. Sex differences in immunocompetence and mortality are well-documented in humans. However, whether HSPCs behave dimorphically between sexes during aging remains unknown. Here, we show that a significant expansion of BM-derived HSPCs occurs in the middle age of female but in the old age of male mice. We then show that a decline of HSPCs in male mice, as indicated by the expression levels of select hematopoietic genes, occurs much earlier in the aging process than that in female mice. Sex-mismatched heterochronic BM transplantations indicate that the middle-aged female BM microenvironment plays a pivotal role in sustaining hematopoietic gene expression during aging. Furthermore, a higher concentration of the pituitary sex hormone follicle-stimulating hormone (FSH) in the serum and a concomitant higher expression of its receptor on HSPCs in the middle-aged and old female mice than age-matched male mice, suggests that FSH may contribute to the sexual dimorphism in aging hematopoiesis. Our study reveals that HSPCs in the BM niches are possibly regulated in a sex-specific manner and influenced differently by sex hormones during aging hematopoiesis.


Asunto(s)
Envejecimiento/fisiología , Hormona Folículo Estimulante/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Receptores de HFE/metabolismo , Caracteres Sexuales , Animales , Antígenos Ly/metabolismo , Médula Ósea , Trasplante de Médula Ósea , Linaje de la Célula , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo , Nicho de Células Madre
12.
JCI Insight ; 4(1)2019 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-30626750

RESUMEN

In heart failure and type 2 diabetes mellitus (DM), the majority of patients have hypomagnesemia, and magnesium (Mg) supplementation has improved cardiac function and insulin resistance. Recently, we have shown that DM can cause cardiac diastolic dysfunction (DD). Therefore, we hypothesized that Mg supplementation would improve diastolic function in DM. High-fat diet-induced diabetic mouse hearts showed increased cardiac DD and hypertrophy. Mice with DM showed a significantly increased E/e' ratio (the ratio of transmitral Doppler early filling velocity [E] to tissue Doppler early diastolic mitral annular velocity [e']) in the echocardiogram, left ventricular end diastolic volume (LVEDV), incidence of DD, left ventricular posterior wall thickness in diastole (PWTd), and ratio of heart weight to tibia length (HW/TL) when compared with controls. DM mice also had hypomagnesemia. Ventricular cardiomyocytes isolated from DM mice exhibited decreased mitochondrial ATP production, a 1.7- ± 0.2-fold increase of mitochondrial ROS, depolarization of the mitochondrial membrane potential, and mitochondrial Ca2+ overload. Dietary Mg administration (50 mg/ml in the drinking water) for 6 weeks increased plasma Mg concentration and improved cardiac function. At the cellular level, Mg improved mitochondrial function with increased ATP, decreased mitochondrial ROS and Ca2+ overload, and repolarized mitochondrial membrane potential. In conclusion, Mg supplementation improved mitochondrial function, reduced oxidative stress, and prevented DD in DM.

13.
Eur J Pharmacol ; 578(1): 11-8, 2008 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-17961543

RESUMEN

Cadmium (Cd), an ubiquitous heavy metal, is known to be accumulated outside of the blood-brain barrier. In this study, we investigated whether Cd has cytotoxicity in mouse brain microvascular endothelial cells (bEnd.3). Results from the cell viability assay showed that Cd caused a remarkable decrease in cell viability in a dose-dependent manner. The cell death induced by Cd appeared to involve apoptosis, based on our results from annexin V staining, electron microscopy and TUNEL staining. And the cell death induced by Cd was inhibited by caspase inhibitor ZVAD-fmk. To further investigate the mechanism of the Cd-induced cell death, we examined the effects of selective inhibitors for mitogen activated protein kinase (MAPK) pathways on the cell death. The Cd-induced cell death was significantly inhibited by p38 MAPK inhibitor SB202190, but not by either, c-Jun N-terminal kinase (JNK) inhibitor SP600125 or extracellular signal-regulated kinase (ERK) inhibitor U0126. Phosphorylations of p38 MAPK, JNK and ERK were stimulated by treatment with CdCl(2). In summary, our results suggest that Cd can induce apoptotic cell death, at least in part, through the p38 MAPK pathway in brain microvascular endothelial cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Cloruro de Cadmio/toxicidad , Endotelio Vascular/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Animales , Anexina A5/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Cloruro de Cadmio/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Microcirculación/citología , Microcirculación/efectos de los fármacos , Microcirculación/metabolismo , Microscopía Electrónica , Fosforilación/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Heart Rhythm ; 15(7): 1072-1080, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29454929

RESUMEN

BACKGROUND: Downregulated sodium currents in heart failure (HF) have been linked to increased arrhythmic risk. Reduced expression of the messenger RNA (mRNA)-stabilizing protein HuR (also known as ELAVL1) may be responsible for the downregulation of sodium channel gene SCN5A mRNA. OBJECTIVE: The purpose of this article was to investigate whether HuR regulates SCN5A mRNA expression and whether manipulation of HuR benefits arrhythmia control in HF. METHODS: Quantitative real-time reverse-transcriptase polymerase chain reaction was used to investigate the expression of SCN5A. Optical mapping of the intact heart was adopted to study the effects of HuR on the conduction velocity and action potential upstroke in mice with myocardial infarct and HF after injection of AAV9 viral particles carrying HuR. RESULTS: HuR was associated with SCN5A mRNA in cardiomyocytes, and expression of HuR was downregulated in failing hearts. The association of HuR and SCN5A mRNA protected SCN5A mRNA from decay. Injection of AAV9 viral particles carrying HuR increased SCN5A expression in mouse heart tissues after MI. Optical mapping of the intact heart demonstrated that overexpression of HuR improved action potential upstroke and conduction velocity in the infarct border zone, which reduced the risk of reentrant arrhythmia after MI. CONCLUSION: Our data indicate that HuR is an important RNA-binding protein in maintaining SCN5A mRNA abundance in cardiomyocytes. Reduced expression of HuR may be at least partially responsible for the downregulation of SCN5A mRNA expression in ischemic HF. Overexpression of HuR may rescue decreased SCN5A expression and reduce arrhythmic risk in HF. Increasing mRNA stability to increase ion channel currents may correct a fundamental defect in HF and represent a new paradigm in antiarrhythmic therapy.


Asunto(s)
Arritmias Cardíacas/genética , Proteína 1 Similar a ELAV/genética , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Miocardio/patología , Canal de Sodio Activado por Voltaje NAV1.5/genética , ARN Mensajero/genética , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/patología , Células Cultivadas , Proteína 1 Similar a ELAV/biosíntesis , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Am Heart Assoc ; 7(9)2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678826

RESUMEN

BACKGROUND: Although transcription is the initial process of gene expression, posttranscriptional gene expression regulation has also played a critical role for fine-tuning gene expression in a fast, precise, and cost-effective manner. Although the regulation of sodium channel α-subunit (SCN5A) mRNA expression has been studied at both transcriptional and pre-mRNA splicing levels, the molecular mechanisms governing SCN5A mRNA expression are far from clear. METHODS AND RESULTS: Herein, we show that, as evidenced by ribonucleoprotein immunoprecipitation assay, RNA binding protein Hu antigen R/ELAV like RNA binding protein 1 (HuR/ELAVL1) and myocyte enhancer factor-2C (MEF2C) transcription factor mRNA are associated. HuR positively regulated transcription factor MEF2C mRNA expression by protecting its mRNA from degradation. As demonstrated by both chromatin immunoprecipitation-quantitative polymerase chain reaction assay and an electrophoretic mobility shift assay, MEF2C enhanced SCN5A transcription by binding to a putative MEF2C binding site within SCN5A promoter region. Overexpression of HuR increased the expression of SCN5A mRNA, and this effect was attenuated by the presence of MEF2C small interfering RNA in cardiomyocytes. CONCLUSIONS: In conclusion, our results suggested that HuR participates in a combined network at the DNA and RNA levels that regulates SCN5A mRNA expression. HuR upregulates MEF2C mRNA expression by protecting MEF2C mRNA from degradation, and consequently, the elevated MEF2C enhances SCN5A mRNA transcription.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Sitios de Unión , Línea Celular , Proteína 1 Similar a ELAV/genética , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , Activación Transcripcional , Regulación hacia Arriba
16.
J Am Heart Assoc ; 5(5)2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27151515

RESUMEN

BACKGROUND: Diabetes mellitus (DM) is associated with mitochondrial oxidative stress. We have shown that myocardial oxidative stress leads to diastolic dysfunction in a hypertensive mouse model. Therefore, we hypothesized that diabetes mellitus could cause diastolic dysfunction through mitochondrial oxidative stress and that a mitochondria-targeted antioxidant (MitoTEMPO) could prevent diastolic dysfunction in a diabetic mouse model. METHODS AND RESULTS: C57BL/6J mice were fed either 60 kcal % fat diet (high-fat diet [HFD]) or normal chow (control) for 8 weeks with or without concurrent MitoTEMPO administration, followed by in vivo assessment of diastolic function and ex vivo studies. HFD mice developed impaired glucose tolerance compared with the control (serum glucose=495±45 mg/dL versus 236±30 mg/dL at 60 minutes after intraperitoneal glucose injection, P<0.05). Myocardial tagged cardiac magnetic resonance imaging showed significantly reduced diastolic circumferential strain (Ecc) rate in the HFD mice compared with controls (5.0±0.3 1/s versus 7.4±0.5 1/s, P<0.05), indicating diastolic dysfunction in the HFD mice. Systolic function was comparable in both groups (left ventricular ejection fraction=66.4±1.4% versus 66.7±1.2%, P>0.05). MitoTEMPO-treated HFD mice showed significant reduction in mitochondria reactive oxygen species, S-glutathionylation of cardiac myosin binding protein C, and diastolic dysfunction, comparable to the control. The fasting insulin levels of MitoTEMPO-treated HFD mice were also comparable to the controls (P>0.05). CONCLUSIONS: MitoTEMPO treatment prevented insulin resistance and diastolic dysfunction, suggesting that mitochondrial oxidative stress may be involved in the pathophysiology of both conditions.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Insuficiencia Cardíaca Diastólica/metabolismo , Resistencia a la Insulina , Mitocondrias Cardíacas/metabolismo , Estrés Oxidativo , Animales , Fenómenos Biomecánicos , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/metabolismo , Diástole , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Insuficiencia Cardíaca Diastólica/diagnóstico por imagen , Hemodinámica , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Especies Reactivas de Oxígeno/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 22(9): e19-23, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12231575

RESUMEN

OBJECTIVE: We investigated the effects of statin compared with the American Heart Association (AHA) Step I Diet on lipoproteins, vasomotor function, tumor necrosis factor (TNF)-alpha, and serological markers of plaque stability. Furthermore, we investigated the mechanism of regulation suggested by experimental studies. METHODS AND RESULTS: For 14 weeks, we administered AHA diet+placebo and AHA diet+simvastatin (20 mg daily) to 31 and 32 randomly selected patients with coronary artery disease, respectively. Compared with diet alone, simvastatin significantly improved the percent flow-mediated dilator response to hyperemia from 3.37+/-2.28% to 5.89+/-2.35% (P<0.001) and lowered plasma levels of C-reactive protein from 0.48 to 0.10 mg/dL (P<0.001), TNF-alpha from 3.38 to 2.79 pg/mL (P<0.001), total matrix metalloproteinase (MMP)-9 from 36 to 28 ng/mL (P=0.006), and tissue inhibitor of matrix metalloproteinase-1 from 80+/-30 to 74+/-23 ng/mL (P=0.041), and simvastatin lowered to a greater extent MMP-9 activity (from 71 to 52 ng/mL, P=0.006) and MMP-9 activity/tissue inhibitor of matrix metalloproteinase-1 ratios (P=0.018), although this difference did not reach statistical significance. There were significant correlations between the degree of changes in TNF-alpha and the degree of changes in MMP-9 activity (r=0.424, P=0.016). However, no significant correlations between lipoprotein levels or flow-mediated dilation percentages and levels of plaque stability markers were determined (-0.208< or =r< or =0.243). CONCLUSIONS: Simvastatin reduced serological markers of inflammation and plaque stability, independent of lipoprotein changes.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Dieta , Hipercolesterolemia/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Óxido Nítrico/metabolismo , Simvastatina/farmacología , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Enfermedad de la Arteria Coronaria/dietoterapia , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/enzimología , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipercolesterolemia/dietoterapia , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/enzimología , Inflamación/sangre , Inflamación/dietoterapia , Inflamación/tratamiento farmacológico , Lípidos/sangre , Masculino , Metaloproteinasas de la Matriz/fisiología , Persona de Mediana Edad , Óxido Nítrico/fisiología , Estudios Prospectivos , Simvastatina/uso terapéutico , Método Simple Ciego , Factor de Necrosis Tumoral alfa/metabolismo , Sistema Vasomotor/efectos de los fármacos , Sistema Vasomotor/metabolismo , Sistema Vasomotor/fisiología
18.
Coron Artery Dis ; 16(3): 135-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15818081

RESUMEN

BACKGROUND: Coronary artery spasm is not infrequently seen in Korea. Most of the patients with coronary spasm show a focal spasm in coronary angiography. However, the cause of the disease is not well known. There have been a few anecdotal case reports of coronary artery spasm associated with hyperthyroidism, but there has not been a report concerning a large series of such patients. Over a period of 5 years and 8 months, we experienced eight patients having the diffuse or severe type of coronary artery spasm in association with hyperthyroidism. METHODS: We investigated the characteristics of the patients with coronary artery spasm, which was diagnosed by coronary angiography or by provocation with an intracoronary injection of acetylcholine or ergonovine. The demographic data, coronary angiographic findings, thyroid function test results, and the follow-up clinical data of the eight patients having coronary artery spasm associated with hyperthyroidism were analyzed. RESULTS: All eight patients had Graves' disease. In six patients, the coronary arterial vasoconstriction developed during the coronary angiography without an injection of ergonovine. In three patients, the left main stem coronary artery was involved in the spasm. Among these eight patients, five were female, and all of these female patients were < or = 51 years old. All of the patients were treated with anti-thyroid medications, calcium channel blockers, and long-acting nitroglycerines; they all remained free of chest pain during the median follow-up period of 5 years. CONCLUSIONS: A severe form of coronary artery spasm could be associated with hyperthyroidism. A high level of suspicion and the thyroid function study should be mandatory for patients with coronary artery spasm, especially for the young female patients.


Asunto(s)
Vasoespasmo Coronario/complicaciones , Enfermedad de Graves/complicaciones , Angina Inestable/complicaciones , Angina Inestable/tratamiento farmacológico , Antitiroideos/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Vasoespasmo Coronario/tratamiento farmacológico , Vasos Coronarios/fisiopatología , Femenino , Enfermedad de Graves/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad , Nitroglicerina/uso terapéutico , Vasoconstricción/fisiología , Vasodilatadores/uso terapéutico
19.
Biomed Res Int ; 2015: 825027, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26504834

RESUMEN

Animal models have suggested a role of renin-angiotensin system (RAS) activation and subsequent cardiac oxidation in heart failure with preserved ejection fraction (HFpEF). Nevertheless, RAS blockade has failed to show efficacy in treatment of HFpEF. We evaluated the role of RAS activation and subsequent systemic oxidation in HFpEF. Oxidative stress markers were compared in 50 subjects with and without early HFpEF. Derivatives of reactive oxidative metabolites (DROMs), F2-isoprostanes (IsoPs), and ratios of oxidized to reduced glutathione (E h GSH) and cysteine (E h CyS) were measured. Angiotensin converting enzyme (ACE) levels and activity were measured. On univariate analysis, HFpEF was associated with male sex (p = 0.04), higher body mass index (BMI) (p = 0.003), less oxidized E h CyS (p = 0.001), lower DROMs (p = 0.02), and lower IsoP (p = 0.03). Higher BMI (OR: 1.3; 95% CI: 1.1-1.6) and less oxidized E h CyS (OR: 1.2; 95% CI: 1.1-1.4) maintained associations with HFpEF on multivariate analysis. Though ACE levels were higher in early HFpEF (OR: 1.09; 95% CI: 1.01-1.05), ACE activity was similar to that in controls. HFpEF is not associated with significant systemic RAS activation or oxidative stress. This may explain the failure of RAS inhibitors to alter outcomes in HFpEF.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Estrés Oxidativo/fisiología , Sistema Renina-Angiotensina/fisiología , Volumen Sistólico/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios Transversales , Femenino , Insuficiencia Cardíaca/epidemiología , Humanos , Masculino , Persona de Mediana Edad
20.
Atherosclerosis ; 161(2): 255-60, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11888507

RESUMEN

Chemokines such as monocyte chemoattractant protein (MCP) -1 and interleukin (IL)-8 are known to be involved in various processes in atherosclerosis such as plaque formation, plaque rupture, and thrombus formation. We investigated whether a new chemokine, Leukotactin (LKN)-1, is involved in atherosclerosis. We tested the expression of LKN-1 by immunohistochemical methods in carotid atherosclerotic plaque specimen. Induction of pro-inflammatory cytokines, transmigration, and tissue factor (TF) expression were tested in THP-1 cells and human peripheral blood monocytes treated with recombinant human LKN-1. Immunohistochemical analyses revealed that expression of LKN-1 occurs in regions of plaques rich in foam cells. In a Boyden chamber assay, THP-1 cells treated with 0.01--10 nM of LKN-1 transmigrated through gelatin coated filters in a dose dependent manner. LKN-1 also induced the transient expression of TNF-alpha, IL-8, and MCP-1 within 15 min of the treatment of the THP-1 cells. When peripheral blood monocytes were treated with LKN-1, expression levels of TF and TF-mediated procoagulating activity were induced in a time- and dose-dependent manner. These results raise the possibility that LKN-1 is another chemokine that is involved in the atherogenesis. LKN-1 may chemoattract immune cells into the plaque, induce pro-inflammatory cytokines, and produce thrombi by inducing TF expression.


Asunto(s)
Enfermedades de las Arterias Carótidas/patología , Quimiocina CCL2/metabolismo , Quimiocinas CC/farmacología , Tromboplastina/efectos de los fármacos , Tromboplastina/metabolismo , Anciano , Anciano de 80 o más Años , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Interleucina-8/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA