Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Cell ; 143(7): 1072-83, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21183071

RESUMEN

The heart has the ability to grow in size in response to exercise, but little is known about the transcriptional mechanisms underlying physiological hypertrophy. Adult cardiomyocytes have also recently been proven to hold the potential for proliferation, a process that could be of great importance for regenerative medicine. Using a unique RT-PCR-based screen against all transcriptional components, we showed that C/EBPß was downregulated with exercise, whereas the expression of CITED4 was increased. Reduction of C/EBPß in vitro and in vivo resulted in a phenocopy of endurance exercise with cardiomyocyte hypertrophy and proliferation. This proliferation was mediated, at least in part, by the increased CITED4. Importantly, mice with reduced cardiac C/EBPß levels displayed substantial resistance to cardiac failure upon pressure overload. These data indicate that C/EBPß represses cardiomyocyte growth and proliferation in the adult mammalian heart and that reduction in C/EBPß is a central signal in physiologic hypertrophy and proliferation.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Corazón/fisiología , Condicionamiento Físico Animal , Animales , Proliferación Celular , Células Cultivadas , Embrión no Mamífero/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Miocardio/citología , Miocitos Cardíacos/metabolismo , Ratas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología
3.
Circ Res ; 130(12): 1994-2014, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35679366

RESUMEN

Acute and chronic animal models of exercise are commonly used in research. Acute exercise testing is used, often in combination with genetic, pharmacological, or other manipulations, to study the impact of these manipulations on the cardiovascular response to exercise and to detect impairments or improvements in cardiovascular function that may not be evident at rest. Chronic exercise conditioning models are used to study the cardiac phenotypic response to regular exercise training and as a platform for discovery of novel pathways mediating cardiovascular benefits conferred by exercise conditioning that could be exploited therapeutically. The cardiovascular benefits of exercise are well established, and, frequently, molecular manipulations that mimic the pathway changes induced by exercise recapitulate at least some of its benefits. This review discusses approaches for assessing cardiovascular function during an acute exercise challenge in rodents, as well as practical and conceptual considerations in the use of common rodent exercise conditioning models. The case for studying feeding in the Burmese python as a model for exercise-like physiological adaptation is also explored.


Asunto(s)
Boidae , Condicionamiento Físico Animal , Animales , Boidae/genética , Fenómenos Fisiológicos Cardiovasculares , Modelos Animales , Condicionamiento Físico Animal/fisiología , Roedores
4.
Arterioscler Thromb Vasc Biol ; 43(2): 330-349, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36453275

RESUMEN

BACKGROUND: Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFß/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS: We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS: Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS: Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.


Asunto(s)
Aterosclerosis , Hígado Graso , Humanos , Animales , Ratones , Hígado/metabolismo , Inflamación/genética , Inflamación/prevención & control , Inflamación/metabolismo , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Activinas/genética , Activinas/metabolismo , Hígado Graso/genética , Hígado Graso/prevención & control , Colesterol/metabolismo , Redes y Vías Metabólicas , Receptores de LDL/genética , Receptores de LDL/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL
5.
Circulation ; 146(5): 412-426, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35862076

RESUMEN

BACKGROUND: The human heart has limited capacity to generate new cardiomyocytes and this capacity declines with age. Because loss of cardiomyocytes may contribute to heart failure, it is crucial to explore stimuli of endogenous cardiac regeneration to favorably shift the balance between loss of cardiomyocytes and the birth of new cardiomyocytes in the aged heart. We have previously shown that cardiomyogenesis can be activated by exercise in the young adult mouse heart. Whether exercise also induces cardiomyogenesis in aged hearts, however, is still unknown. Here, we aim to investigate the effect of exercise on the generation of new cardiomyocytes in the aged heart. METHODS: Aged (20-month-old) mice were subjected to an 8-week voluntary running protocol, and age-matched sedentary animals served as controls. Cardiomyogenesis in aged hearts was assessed on the basis of 15N-thymidine incorporation and multi-isotope imaging mass spectrometry. We analyzed 1793 cardiomyocytes from 5 aged sedentary mice and compared these with 2002 cardiomyocytes from 5 aged exercised mice, followed by advanced histology and imaging to account for ploidy and nucleation status of the cell. RNA sequencing and subsequent bioinformatic analyses were performed to investigate transcriptional changes induced by exercise specifically in aged hearts in comparison with young hearts. RESULTS: Cardiomyogenesis was observed at a significantly higher frequency in exercised compared with sedentary aged hearts on the basis of the detection of mononucleated/diploid 15N-thymidine-labeled cardiomyocytes. No mononucleated/diploid 15N-thymidine-labeled cardiomyocyte was detected in sedentary aged mice. The annual rate of mononucleated/diploid 15N-thymidine-labeled cardiomyocytes in aged exercised mice was 2.3% per year. This compares with our previously reported annual rate of 7.5% in young exercised mice and 1.63% in young sedentary mice. Transcriptional profiling of young and aged exercised murine hearts and their sedentary controls revealed that exercise induces pathways related to circadian rhythm, irrespective of age. One known oscillating transcript, however, that was exclusively upregulated in aged exercised hearts, was isoform 1.4 of regulator of calcineurin, whose regulation and functional role were explored further. CONCLUSIONS: Our data demonstrate that voluntary running in part restores cardiomyogenesis in aged mice and suggest that pathways associated with circadian rhythm may play a role in physiologically stimulated cardiomyogenesis.


Asunto(s)
Miocitos Cardíacos , Condicionamiento Físico Animal , Animales , Calcineurina/metabolismo , Humanos , Lactante , Ratones , Miocitos Cardíacos/citología , Timidina/metabolismo
6.
Circulation ; 145(16): 1218-1233, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35114812

RESUMEN

BACKGROUND: The heart grows in response to pathological and physiological stimuli. The former often precedes cardiomyocyte loss and heart failure; the latter paradoxically protects the heart and enhances cardiomyogenesis. The mechanisms underlying these differences remain incompletely understood. Although long noncoding RNAs (lncRNAs) are important in cardiac development and disease, less is known about their roles in physiological hypertrophy or cardiomyogenesis. METHODS: RNA sequencing was applied to hearts from mice after 8 weeks of voluntary exercise-induced physiological hypertrophy and cardiomyogenesis or transverse aortic constriction for 2 or 8 weeks to induce pathological hypertrophy or heart failure. The top lncRNA candidate was overexpressed in hearts with adeno-associated virus vectors and inhibited with antisense locked nucleic acid-GapmeRs to examine its function. Downstream effectors were identified through promoter analyses and binding assays. The functional roles of a novel downstream effector, dachsous cadherin-related 2 (DCHS2), were examined through transgenic overexpression in zebrafish and cardiac-specific deletion in Cas9-knockin mice. RESULTS: We identified exercise-regulated cardiac lncRNAs, called lncExACTs. lncExACT1 was evolutionarily conserved and decreased in exercised hearts but increased in human and experimental heart failure. Cardiac lncExACT1 overexpression caused pathological hypertrophy and heart failure; lncExACT1 inhibition induced physiological hypertrophy and cardiomyogenesis, protecting against cardiac fibrosis and dysfunction. lncExACT1 functioned by regulating microRNA-222, calcineurin signaling, and Hippo/Yap1 signaling through DCHS2. Cardiomyocyte DCHS2 overexpression in zebrafish induced pathological hypertrophy and impaired cardiac regeneration, promoting scarring after injury. In contrast, murine DCHS2 deletion induced physiological hypertrophy and promoted cardiomyogenesis. CONCLUSIONS: These studies identify lncExACT1-DCHS2 as a novel pathway regulating cardiac hypertrophy and cardiomyogenesis. lncExACT1-DCHS2 acts as a master switch toggling the heart between physiological and pathological growth to determine functional outcomes, providing a potentially tractable therapeutic target for harnessing the beneficial effects of exercise.


Asunto(s)
Proteínas Relacionadas con las Cadherinas/metabolismo , Insuficiencia Cardíaca , MicroARNs , ARN Largo no Codificante , Animales , Cardiomegalia/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Pez Cebra/genética
7.
Circ Res ; 126(4): 533-551, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32078451

RESUMEN

During aging, deterioration in cardiac structure and function leads to increased susceptibility to heart failure. The need for interventions to combat this age-related cardiac decline is becoming increasingly urgent as the elderly population continues to grow. Our understanding of cardiac aging, and aging in general, is limited. However, recent studies of age-related decline and its prevention through interventions like exercise have revealed novel pathological and cardioprotective pathways. In this review, we summarize recent findings concerning the molecular mechanisms of age-related heart failure and highlight exercise as a valuable experimental platform for the discovery of much-needed novel therapeutic targets in this chronic disease.


Asunto(s)
Envejecimiento/fisiología , Ejercicio Físico/fisiología , Insuficiencia Cardíaca/fisiopatología , Corazón/fisiopatología , Miocardio/metabolismo , Transducción de Señal/fisiología , Anciano , Envejecimiento/genética , Envejecimiento/metabolismo , Regulación del Desarrollo de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Humanos , MicroARNs/genética , Transducción de Señal/genética
8.
Circ Res ; 127(5): 631-646, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32418505

RESUMEN

RATIONALE: Cardiac CITED4 (CBP/p300-interacting transactivators with E [glutamic acid]/D [aspartic acid]-rich-carboxylterminal domain4) is induced by exercise and is sufficient to cause physiological hypertrophy and mitigate adverse ventricular remodeling after ischemic injury. However, the role of endogenous CITED4 in response to physiological or pathological stress is unknown. OBJECTIVE: To investigate the role of CITED4 in murine models of exercise and pressure overload. METHODS AND RESULTS: We generated cardiomyocyte-specific CITED4 knockout mice (C4KO) and subjected them to an intensive swim exercise protocol as well as transverse aortic constriction (TAC). Echocardiography, Western blotting, qPCR, immunohistochemistry, immunofluorescence, and transcriptional profiling for mRNA and miRNA (microRNA) expression were performed. Cellular crosstalk was investigated in vitro. CITED4 deletion in cardiomyocytes did not affect baseline cardiac size or function in young adult mice. C4KO mice developed modest cardiac dysfunction and dilation in response to exercise. After TAC, C4KOs developed severe heart failure with left ventricular dilation, impaired cardiomyocyte growth accompanied by reduced mTOR (mammalian target of rapamycin) activity and maladaptive cardiac remodeling with increased apoptosis, autophagy, and impaired mitochondrial signaling. Interstitial fibrosis was markedly increased in C4KO hearts after TAC. RNAseq revealed induction of a profibrotic miRNA network. miR30d was decreased in C4KO hearts after TAC and mediated crosstalk between cardiomyocytes and fibroblasts to modulate fibrosis. miR30d inhibition was sufficient to increase cardiac dysfunction and fibrosis after TAC. CONCLUSIONS: CITED4 protects against pathological cardiac remodeling by regulating mTOR activity and a network of miRNAs mediating cardiomyocyte to fibroblast crosstalk. Our findings highlight the importance of CITED4 in response to both physiological and pathological stimuli.


Asunto(s)
Cardiomegalia Inducida por el Ejercicio , Hipertrofia Ventricular Izquierda/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Comunicación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/patología , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcriptoma
9.
Mol Ther ; 28(7): 1731-1740, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32243833

RESUMEN

VEGF-B gene therapy is a promising proangiogenic treatment for ischemic heart disease, but, unexpectedly, we found that high doses of VEGF-B promote ventricular arrhythmias (VAs). VEGF-B knockout, alpha myosin heavy-chain promoter (αMHC)-VEGF-B transgenic mice, and pigs transduced intramyocardially with adenoviral (Ad)VEGF- B186 were studied. Immunostaining showed a 2-fold increase in the number of nerves per field (76 vs. 39 in controls, p < 0.001) and an abnormal nerve distribution in the hypertrophic hearts of 11- to 20-month-old αMHC-VEGF-B mice. AdVEGF-B186 gene transfer (GT) led to local sprouting of nerve endings in pig myocardium (141 vs. 78 nerves per field in controls, p < 0.05). During dobutamine stress, 60% of the αMHC-VEGF-B hypertrophic mice had arrhythmias as compared to 7% in controls, and 20% of the AdVEGF-B186-transduced pigs and 100% of the combination of AdVEGF-B186- and AdsVEGFR-1-transduced pigs displayed VAs and even ventricular fibrillation. AdVEGF-B186 GT significantly increased the risk of sudden cardiac death in pigs when compared to any other GT with different VEGFs (hazard ratio, 500.5; 95% confidence interval [CI] 46.4-5,396.7; p < 0.0001). In gene expression analysis, VEGF-B induced the upregulation of Nr4a2, ATF6, and MANF in cardiomyocytes, molecules previously linked to nerve growth and differentiation. Thus, high AdVEGF-B186 overexpression induced nerve growth in the adult heart via a VEGFR-1 signaling-independent mechanism, leading to an increased risk of VA and sudden cardiac death.


Asunto(s)
Arritmias Cardíacas/patología , Cadenas Pesadas de Miosina/genética , Sistema Nervioso Simpático/patología , Regulación hacia Arriba , Factor B de Crecimiento Endotelial Vascular/genética , Animales , Animales Modificados Genéticamente , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Dependovirus/genética , Notificación de Enfermedades , Femenino , Técnicas de Inactivación de Genes , Terapia Genética , Vectores Genéticos/administración & dosificación , Masculino , Ratones , Regiones Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Porcinos , Sistema Nervioso Simpático/metabolismo , Transducción Genética , Factor B de Crecimiento Endotelial Vascular/efectos adversos , Factor B de Crecimiento Endotelial Vascular/metabolismo
12.
Circ Res ; 121(12): 1370-1378, 2017 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-28928113

RESUMEN

RATIONALE: Pregnancy profoundly alters maternal physiology. The heart hypertrophies during pregnancy, but its metabolic adaptations, are not well understood. OBJECTIVE: To determine the mechanisms underlying cardiac substrate use during pregnancy. METHODS AND RESULTS: We use here 13C glucose, 13C lactate, and 13C fatty acid tracing analyses to show that hearts in late pregnant mice increase fatty acid uptake and oxidation into the tricarboxylic acid cycle, while reducing glucose and lactate oxidation. Mitochondrial quantity, morphology, and function do not seem altered. Insulin signaling seems intact, and the abundance and localization of the major fatty acid and glucose transporters, CD36 (cluster of differentiation 36) and GLUT4 (glucose transporter type 4), are also unchanged. Rather, we find that the pregnancy hormone progesterone induces PDK4 (pyruvate dehydrogenase kinase 4) in cardiomyocytes and that elevated PDK4 levels in late pregnancy lead to inhibition of PDH (pyruvate dehydrogenase) and pyruvate flux into the tricarboxylic acid cycle. Blocking PDK4 reverses the metabolic changes seen in hearts in late pregnancy. CONCLUSIONS: Taken together, these data indicate that the hormonal environment of late pregnancy promotes metabolic remodeling in the heart at the level of PDH, rather than at the level of insulin signaling.


Asunto(s)
Miocardio/metabolismo , Embarazo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ácido Pirúvico/metabolismo , Animales , Ciclo del Ácido Cítrico , Ácidos Grasos/metabolismo , Femenino , Glucosa/metabolismo , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos C57BL , Progesterona/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
13.
EMBO J ; 33(13): 1438-53, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24825348

RESUMEN

Mice overexpressing the mitotic checkpoint kinase gene BubR1 live longer, whereas mice hypomorphic for BubR1 (BubR1(H/H)) live shorter and show signs of accelerated aging. As wild-type mice age, BubR1 levels decline in many tissues, a process that is proposed to underlie normal aging and age-related diseases. Understanding why BubR1 declines with age and how to slow this process is therefore of considerable interest. The sirtuins (SIRT1-7) are a family of NAD(+)-dependent deacetylases that can delay age-related diseases. Here, we show that the loss of BubR1 levels with age is due to a decline in NAD(+) and the ability of SIRT2 to maintain lysine-668 of BubR1 in a deacetylated state, which is counteracted by the acetyltransferase CBP. Overexpression of SIRT2 or treatment of mice with the NAD(+) precursor nicotinamide mononucleotide (NMN) increases BubR1 abundance in vivo. Overexpression of SIRT2 in BubR1(H/H) animals increases median lifespan, with a greater effect in male mice. Together, these data indicate that further exploration of the potential of SIRT2 and NAD(+) to delay diseases of aging in mammals is warranted.


Asunto(s)
Longevidad/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Sirtuina 2/metabolismo , Animales , Proteínas de Ciclo Celular , Inducción Enzimática/fisiología , Células HeLa , Humanos , Masculino , Ratones , Ratones Noqueados , NAD/genética , NAD/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Sirtuina 2/genética
14.
Circ Res ; 118(2): 279-95, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26838314

RESUMEN

Aging induces structural and functional changes in the heart that are associated with increased risk of cardiovascular disease and impaired functional capacity in the elderly. Exercise is a diagnostic and therapeutic tool, with the potential to provide insights into clinical diagnosis and prognosis, as well as the molecular mechanisms by which aging influences cardiac physiology and function. In this review, we first provide an overview of how aging impacts the cardiac response to exercise, and the implications this has for functional capacity in older adults. We then review the underlying molecular mechanisms by which cardiac aging contributes to exercise intolerance, and conversely how exercise training can potentially modulate aging phenotypes in the heart. Finally, we highlight the potential use of these exercise models to complement models of disease in efforts to uncover new therapeutic targets to prevent or treat heart disease in the aging population.


Asunto(s)
Envejecimiento , Cardiomegalia/fisiopatología , Tolerancia al Ejercicio , Ejercicio Físico , Corazón/fisiopatología , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Señalización del Calcio , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Corazón/inervación , Humanos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Fenotipo , Factores Protectores , Receptores Adrenérgicos beta/metabolismo , Factores de Riesgo
16.
Arterioscler Thromb Vasc Biol ; 36(9): 1854-67, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27386938

RESUMEN

OBJECTIVE: S100A6, a member of the S100 protein family, has been described as relevant for cell cycle entry and progression in endothelial cells. The molecular mechanism conferring S100A6's proliferative actions, however, remained elusive. APPROACH AND RESULTS: Originating from the clinically relevant observation of enhanced S100A6 protein expression in proliferating endothelial cells in remodeling coronary and carotid arteries, our study unveiled S100A6 as a suppressor of antiproliferative signal transducers and activators of transcription 1 signaling. Discovery of the molecular liaison was enabled by combining gene expression time series analysis with bioinformatic pathway modeling in S100A6-silenced human endothelial cells stimulated with vascular endothelial growth factor A. This unbiased approach led to successful identification and experimental validation of interferon-inducible transmembrane protein 1 and protein inhibitors of activated signal transducers and activators of transcription as key components of the link between S100A6 and signal transducers and activators of transcription 1. CONCLUSIONS: Given the important role of coordinated endothelial cell cycle activity for integrity and reconstitution of the inner lining of arterial blood vessels in health and disease, signal transducers and activators of transcription 1 suppression by S100A6 may represent a promising therapeutic target to facilitate reendothelialization in damaged vessels.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proliferación Celular , Células Endoteliales/metabolismo , Proteínas S100/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Biología Computacional , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Silenciador del Gen , Humanos , Masculino , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Interferencia de ARN , Ratas Sprague-Dawley , Repitelización , Proteína A6 de Unión a Calcio de la Familia S100 , Proteínas S100/genética , Factor de Transcripción STAT1/genética , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sus scrofa , Factores de Tiempo , Transcriptoma , Transfección , Factor A de Crecimiento Endotelial Vascular/farmacología , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
17.
Circulation ; 131(25): 2202-2216, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-25995320

RESUMEN

BACKGROUND: Biomarkers that predict response to cardiac resynchronization therapy (CRT) in heart failure patients with dyssynchrony (HFDYS) would be clinically important. Circulating extracellular microRNAs (miRNAs) have emerged as novel biomarkers that may also play important functional roles, but their relevance as markers for CRT response has not been examined. METHODS AND RESULTS: Comprehensive miRNA polymerase chain reaction arrays were used to assess baseline levels of 766 plasma miRNAs in patients undergoing clinically indicated CRT in an initial discovery set (n=12) with and without subsequent echocardiographic improvement at 6 months after CRT. Validation of candidate miRNAs in 61 additional patients confirmed that baseline plasma miR-30d was associated with CRT response (defined as an increase in left ventricular ejection fraction ≥10%). MiR-30d was enriched in coronary sinus blood and increased in late-contracting myocardium in a canine model of HFDYS, indicating cardiac origin with maximal expression in areas of high mechanical stress. We examined the functional effects of miR-30d in cultured cardiomyocytes and determined that miR-30d is expressed in cardiomyocytes and released in vesicles in response to mechanical stress. Overexpression of miR-30d in cultured cardiomyocytes led to cardiomyocyte growth and protected against apoptosis by targeting the mitogen-associated kinase 4, a downstream effector of tumor necrosis factor. In HFDYS patients, miR-30d plasma levels inversely correlated with high-sensitivity troponin T, a marker of myocardial necrosis. CONCLUSIONS: Baseline plasma miR-30d level is associated with response to CRT in HFDYS in this translational pilot study. MiR-30d increase in cardiomyocytes correlates with areas of increased wall stress in HFDYS and is protective against deleterious tumor necrosis factor signaling.


Asunto(s)
Apoptosis/fisiología , Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca/sangre , MicroARNs/sangre , Miocitos Cardíacos/fisiología , Investigación Biomédica Traslacional , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/sangre , Terapia de Resincronización Cardíaca/tendencias , Perros , Femenino , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/terapia , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Ratas , Ratas Sprague-Dawley , Investigación Biomédica Traslacional/tendencias , Resultado del Tratamiento
18.
FASEB J ; 28(10): 4408-19, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25005176

RESUMEN

Mitochondrial dysfunction in adipose tissue occurs in obesity, type 2 diabetes, and some forms of lipodystrophy, but whether this dysfunction contributes to or is the result of these disorders is unknown. To investigate the physiological consequences of severe mitochondrial impairment in adipose tissue, we generated mice deficient in mitochondrial transcription factor A (TFAM) in adipocytes by using mice carrying adiponectin-Cre and TFAM floxed alleles. These adiponectin TFAM-knockout (adipo-TFAM-KO) mice had a 75-81% reduction in TFAM in the subcutaneous and intra-abdominal white adipose tissue (WAT) and interscapular brown adipose tissue (BAT), causing decreased expression and enzymatic activity of proteins in complexes I, III, and IV of the electron transport chain (ETC). This mitochondrial dysfunction led to adipocyte death and inflammation in WAT and a whitening of BAT. As a result, adipo-TFAM-KO mice were resistant to weight gain, but exhibited insulin resistance on both normal chow and high-fat diets. These lipodystrophic mice also developed hypertension, cardiac hypertrophy, and cardiac dysfunction. Thus, isolated mitochondrial dysfunction in adipose tissue can lead a syndrome of lipodystrophy with metabolic syndrome and cardiovascular complications.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Resistencia a la Insulina , Lipodistrofia/metabolismo , Mitocondrias/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/patología , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Proteínas de Unión al ADN/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Proteínas del Grupo de Alta Movilidad/genética , Hipertensión/genética , Hipertensión/metabolismo , Lipodistrofia/genética , Lipodistrofia/fisiopatología , Masculino , Ratones , Aumento de Peso
19.
J Mol Cell Cardiol ; 72: 74-84, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24613264

RESUMEN

Cardiac hypertrophy is controlled by a highly connected signaling network with many effectors of cardiac myocyte size. Quantification of the contribution of individual pathways to specific changes in shape and transcript abundance is needed to better understand hypertrophy signaling and to improve heart failure therapies. We stimulated cardiac myocytes with 15 hypertrophic agonists and quantitatively characterized differential regulation of 5 shape features using high-throughput microscopy and transcript levels of 12 genes using qPCR. Transcripts measured were associated with phenotypes including fibrosis, cell death, contractility, proliferation, angiogenesis, inflammation, and the fetal cardiac gene program. While hypertrophy pathways are highly connected, the agonist screen revealed distinct hypertrophy phenotypic signatures for the 15 receptor agonists. We then used k-means clustering of inputs and outputs to identify a network map linking input modules to output modules. Five modules were identified within inputs and outputs with many maladaptive outputs grouping together in one module: Bax, C/EBPß, Serca2a, TNFα, and CTGF. Subsequently, we identified mechanisms underlying two correlations revealed in the agonist screen: correlation between regulators of fibrosis and cell death signaling (CTGF and Bax mRNA) caused by AngII; and myocyte proliferation (CITED4 mRNA) and elongation caused by Nrg1. Follow-up experiments revealed positive regulation of Bax mRNA level by CTGF and an incoherent feedforward loop linking Nrg1, CITED4 and elongation. With this agonist screen, we identified the most influential inputs in the cardiac hypertrophy signaling network for a variety of features related to pathological and protective hypertrophy signaling and shared regulation among cardiac myocyte phenotypes.


Asunto(s)
Cardiomegalia/genética , Regulación de la Expresión Génica , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , Angiotensina II/genética , Angiotensina II/metabolismo , Animales , Animales Recién Nacidos , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patología , Forma de la Célula/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Perfilación de la Expresión Génica , Miocitos Cardíacos/patología , Neurregulina-1/genética , Neurregulina-1/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
20.
Circulation ; 128(11): 1225-33, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23912910

RESUMEN

BACKGROUND: Cardiomyocyte hypertrophy is a critical precursor to the development of heart failure. Methods to phenotype cellular hypertrophy noninvasively are limited. The goal was to validate a cardiac magnetic resonance-based approach for the combined assessment of extracellular matrix expansion and cardiomyocyte hypertrophy. METHODS AND RESULTS: Two murine models of hypertension (n=18, with n=15 controls) induced by l-N(G)-nitroarginine methyl ester (L-NAME) and pressure overload (n=11) from transaortic constriction (TAC) were imaged by cardiac magnetic resonance at baseline and 7 weeks after L-NAME treatment or up to 7 weeks after TAC. T1 relaxation times were measured before and after gadolinium contrast. The intracellular lifetime of water (τic), a cell size-dependent parameter, and extracellular volume fraction, a marker of interstitial fibrosis, were determined with a model for transcytolemmal water exchange. Cardiomyocyte diameter and length were measured on FITC-wheat germ agglutinin-stained sections. The τic correlated strongly with histological cardiomyocyte volume-to-surface ratio (r=0.78, P<0.001) and cell volume (r=0.75, P<0.001). Histological cardiomyocyte diameters and cell volumes were higher in mice treated with L-NAME compared with controls (P<0.001). In the TAC model, cardiac magnetic resonance and histology showed cell hypertrophy at 2 weeks after TAC without significant fibrosis at this early time point. Mice exposed to TAC demonstrated a significant, longitudinal, and parallel increase in histological cell volume, volume-to-surface ratio, and τic between 2 and 7 weeks after TAC. CONCLUSION: The τic measured by contrast-enhanced cardiac magnetic resonance provides a noninvasive measure of cardiomyocyte hypertrophy. Extracellular volume fraction and τic can track myocardial tissue remodeling from pressure overload.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Miocitos Cardíacos/patología , Remodelación Ventricular/fisiología , Algoritmos , Animales , Estenosis de la Válvula Aórtica/complicaciones , Biomarcadores , Agua Corporal/metabolismo , Cardiomiopatía Hipertrófica/etiología , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , Tamaño de la Célula , Medios de Contraste , Gadolinio DTPA , Hipertensión/inducido químicamente , Hipertensión/etiología , Hipertensión/patología , Hipertrofia , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones , NG-Nitroarginina Metil Éster/toxicidad , Fenotipo , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA