Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
Sci Transl Med ; 13(623): eabc7367, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34878822

RESUMEN

Skeletal muscle displays remarkable plasticity upon exercise and is also one of the organs most affected by aging. Despite robust evidence that aging is associated with loss of fast-twitch (type II) muscle fibers, the underlying mechanisms remain to be elucidated. Here, we identified an exercise-induced long noncoding RNA, CYTOR, whose exercise responsiveness was conserved in human and rodents. Cytor overexpression in mouse myogenic progenitor cells enhanced myogenic differentiation by promoting fast-twitch cell fate, whereas Cytor knockdown deteriorated expression of mature type II myotubes. Skeletal muscle Cytor expression was reduced upon mouse aging, and Cytor expression in young mice was required to maintain proper muscle morphology and function. In aged mice, rescuing endogenous Cytor expression using adeno-associated virus serotype 9 delivery of CRISPRa reversed the age-related decrease in type II fibers and improved muscle mass and function. In humans, CYTOR expression correlated with type II isoform expression and was decreased in aged myoblasts. Increased CYTOR expression, mediated by a causal cis­expression quantitative trait locus located within a CYTOR skeletal muscle enhancer element, was associated with improved 6-min walk performance in aged individuals from the Helsinki Birth Cohort Study. Direct CYTOR overexpression using CRISPRa in aged human donor myoblasts enhanced expression of type II myosin isoforms. Mechanistically, Cytor reduced chromatin accessibility and occupancy at binding motifs of the transcription factor Tead1 by binding, and hence sequestering, Tead1. In conclusion, the long noncoding RNA Cytor was found to be a regulator of fast-twitch myogenesis in aging.


Asunto(s)
ARN Largo no Codificante , Envejecimiento/genética , Animales , Diferenciación Celular/genética , Estudios de Cohortes , Humanos , Ratones , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
J Mol Cell Cardiol ; 148: 106-119, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32918915

RESUMEN

AIMS: Endurance training improves aerobic fitness and cardiac function in individuals with heart failure. However, the underlying mechanisms are not well characterized. Exercise training could therefore act as a tool to discover novel targets for heart failure treatment. We aimed to associate changes in Ca2+ handling and electrophysiology with micro-RNA (miRNA) profile in exercise trained heart failure rats to establish which miRNAs induce heart failure-like effects in Ca2+ handling and electrophysiology. METHODS AND RESULTS: Post-myocardial infarction (MI) heart failure was induced in Sprague Dawley rats. Rats with MI were randomized to sedentary control (sed), moderate (mod)- or high-intensity (high) endurance training for 8 weeks. Exercise training improved cardiac function, Ca2+ handling and electrophysiology including reduced susceptibility to arrhythmia in an exercise intensity-dependent manner where high intensity gave a larger effect. Fifty-five miRNAs were significantly regulated (up or down) in MI-sed, of which 18 and 3 were changed towards Sham-sed in MI-high and MI-mod, respectively. Thereafter we experimentally altered expression of these "exercise-miRNAs" individually in human induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CM) in the same direction as they were changed in MI. Of the "exercise-miRNAs", miR-214-3p prolonged AP duration, whereas miR-140 and miR-208a shortened AP duration. miR-497-5p prolonged Ca2+ release whereas miR-214-3p and miR-31a-5p prolonged Ca2+ decay. CONCLUSION: Using exercise training as a tool, we discovered that miR-214-3p, miR-497-5p, miR-31a-5p contribute to heart-failure like behaviour in Ca2+ handling and electrophysiology and could be potential treatment targets.


Asunto(s)
Fenómenos Electrofisiológicos , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Condicionamiento Físico Animal , Aerobiosis , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/fisiopatología , Biomarcadores/metabolismo , Cardiomegalia/complicaciones , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/complicaciones , MicroARNs/metabolismo , Contracción Miocárdica/fisiología , Infarto del Miocardio/complicaciones , Miocitos Cardíacos/metabolismo , Ratas Sprague-Dawley , Fibrilación Ventricular/complicaciones , Fibrilación Ventricular/genética , Fibrilación Ventricular/fisiopatología
4.
Nat Metab ; 2(9): 829-839, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32807982

RESUMEN

The cardiac benefits of exercise have been recognized for centuries. Studies have undisputedly shown that regular exercise is beneficial for the cardiovascular system in young, old, healthy and diseased populations. For these reasons, physical activity has been recommended worldwide for cardiovascular disease prevention and treatment. Although the benefits of exercise are clear, understanding of the molecular triggers that orchestrate these effects remains incomplete and has been a topic of intense research in recent years. Here, we provide a comprehensive review of the cardiac effects of physical activity, beginning with a brief history of exercise in cardiovascular medicine and then discussing seminal work on the physiological effects of exercise in healthy, diseased and aged hearts. Later, we revisit pioneering work on the molecular mechanisms underlying the cardiac benefits of exercise, and we conclude with our view on the translational potential of this knowledge as a powerful platform for cardiovascular disease drug discovery.


Asunto(s)
Ejercicio Físico/fisiología , Estado de Salud , Corazón/fisiología , Miocardio/metabolismo , Animales , Fármacos Cardiovasculares/farmacología , Sistema Cardiovascular , Descubrimiento de Drogas , Humanos
5.
Nat Metab ; 2(8): 653-654, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32694822
6.
Mol Metab ; 39: 101012, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32408015

RESUMEN

OBJECTIVE: We tested the hypothesis that exercise training would attenuate metabolic impairment in a model of severe cancer cachexia. METHODS: We used multiple in vivo and in vitro methods to explore the mechanisms underlying the beneficial effects induced by exercise training in tumor-bearing rats. RESULTS: Exercise training improved running capacity, prolonged lifespan, reduced oxidative stress, and normalized muscle mass and contractile function in tumor-bearing rats. An unbiased proteomic screening revealed COP9 signalosome complex subunit 2 (COPS2) as one of the most downregulated proteins in skeletal muscle at the early stage of cancer cachexia. Exercise training normalized muscle COPS2 protein expression in tumor-bearing rats and mice. Lung cancer patients with low endurance capacity had low muscle COPS2 protein expression as compared to age-matched control subjects. To test whether decrease in COPS2 protein levels could aggravate or be an intrinsic compensatory mechanism to protect myotubes from cancer effects, we performed experiments in vitro using primary myotubes. COPS2 knockdown in human myotubes affected multiple cellular pathways, including regulation of actin cytoskeleton. Incubation of cancer-conditioned media in mouse myotubes decreased F-actin expression, which was partially restored by COPS2 knockdown. Direct repeat 4 (DR4) response elements have been shown to positively regulate gene expression. COPS2 overexpression decreased the DR4 activity in mouse myoblasts, and COPS2 knockdown inhibited the effects of cancer-conditioned media on DR4 activity. CONCLUSIONS: These studies demonstrated that exercise training may be an important adjuvant therapy to counteract cancer cachexia and uncovered novel mechanisms involving COPS2 to regulate myotube homeostasis in cancer cachexia.


Asunto(s)
Complejo del Señalosoma COP9/metabolismo , Músculo Esquelético/metabolismo , Neoplasias/metabolismo , Estrés Oxidativo , Condicionamiento Físico Animal , Proteínas Represoras/metabolismo , Animales , Biomarcadores , Complejo del Señalosoma COP9/genética , Caquexia/etiología , Caquexia/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Mioblastos/metabolismo , Neoplasias/complicaciones , Oxidación-Reducción , Proteómica/métodos , Ratas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Represoras/genética , Transducción de Señal
7.
Prog Cardiovasc Dis ; 62(2): 193-202, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30867130

RESUMEN

The benefits of physical activity in cardiovascular diseases have long been appreciated. However, the molecular mechanisms that trigger and sustain the cardiac benefits of exercise are poorly understood, and it is anticipated that unveiling these mechanisms will identify novel therapeutic targets. In search of these mechanisms we took advantage of unbiased RNA-sequencing (RNA-seq) technology to discover cardiac gene targets whose expression is disrupted in heart failure (HF) and rescued by exercise in a rat model. Upon exhaustive validation in a separate rat cohort (qPCR) and human datasets, we shortlisted 16 targets for a cell-based screening, aiming to evaluate whether targeted disruption of these genes with silencing RNA would affect the abundance of a CVD biomarker (BNP, B-type natriuretic peptide) in human cardiomyocytes. Overall, these experiments showed that Proline Dehydrogenase (PRODH) expression is reduced in human failing hearts, rescued by exercise in a rat model of HF, and its targeted knockdown increases BNP expression in human cardiomyocytes. On the other hand, overexpression of PRODH increases the abundance of metabolism-related gene transcripts, and PRODH appears to be crucial to sustain normal mitochondrial function and maintenance of ATP levels in human cardiomyocytes in a hypoxic environment, as well as for redox homeostasis in both normoxic and hypoxic conditions. Altogether our findings show that PRODH is a novel molecular target of exercise in failing hearts and highlight its role in cardiomyocyte physiology, thereby proposing PRODH as a potential experimental target for gene therapy in HF.


Asunto(s)
Ejercicio Físico/fisiología , Insuficiencia Cardíaca , Prolina Oxidasa/metabolismo , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Humanos , Mitocondrias Cardíacas/metabolismo , Ratas , Transducción de Señal
8.
Sci Rep ; 8(1): 17772, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30538258

RESUMEN

Given the association between high aerobic capacity and the prevention of metabolic diseases, elucidating the mechanisms by which high aerobic capacity regulates whole-body metabolic homeostasis is a major research challenge. Oxidative post-translational modifications (Ox-PTMs) of proteins can regulate cellular homeostasis in skeletal and cardiac muscles, but the relationship between Ox-PTMs and intrinsic components of oxidative energy metabolism is still unclear. Here, we evaluated the Ox-PTM profile in cardiac and skeletal muscles of rats bred for low (LCR) and high (HCR) intrinsic aerobic capacity. Redox proteomics screening revealed different cysteine (Cys) Ox-PTM profile between HCR and LCR rats. HCR showed a higher number of oxidized Cys residues in skeletal muscle compared to LCR, while the opposite was observed in the heart. Most proteins with differentially oxidized Cys residues in the skeletal muscle are important regulators of oxidative metabolism. The most oxidized protein in the skeletal muscle of HCR rats was malate dehydrogenase (MDH1). HCR showed higher MDH1 activity compared to LCR in skeletal, but not cardiac muscle. These novel findings indicate a clear association between Cys Ox-PTMs and aerobic capacity, leading to novel insights into the role of Ox-PTMs as an essential signal to maintain metabolic homeostasis.


Asunto(s)
Cisteína/metabolismo , Metabolismo Energético/fisiología , Estrés Oxidativo/fisiología , Animales , Respiración de la Célula , Tolerancia al Ejercicio/fisiología , Malato Deshidrogenasa/metabolismo , Masculino , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Oxidación-Reducción , Condicionamiento Físico Animal/fisiología , Resistencia Física/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Carrera/fisiología
9.
Sci Rep ; 8(1): 11818, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087400

RESUMEN

Increased proteolytic activity has been widely associated with skeletal muscle atrophy. However, elevated proteolysis is also critical for the maintenance of cellular homeostasis by disposing cytotoxic proteins and non-functioning organelles. We recently demonstrated that exercise activates autophagy and re-establishes proteostasis in cardiac diseases. Here, we characterized the impact of exercise on skeletal muscle autophagy and proteostasis in a model of neurogenic myopathy induced by sciatic nerve constriction in rats. Neurogenic myopathy, characterized by progressive atrophy and impaired contractility, was paralleled by accumulation of autophagy-related markers and loss of acute responsiveness to both colchicine and chloroquine. These changes were correlated with elevated levels of damaged proteins, chaperones and pro-apoptotic markers compared to control animals. Sustained autophagy inhibition using chloroquine in rats (50 mg.kg-1.day-1) or muscle-specific deletion of Atg7 in mice was sufficient to impair muscle contractility in control but not in neurogenic myopathy, suggesting that dysfunctional autophagy is critical in skeletal muscle pathophysiology. Finally, 4 weeks of aerobic exercise training (moderate treadmill running, 5x/week, 1 h/day) prior to neurogenic myopathy improved skeletal muscle autophagic flux and proteostasis. These changes were followed by spared muscle mass and better contractility properties. Taken together, our findings suggest the potential value of exercise in maintaining skeletal muscle proteostasis and slowing down the progression of neurogenic myopathy.


Asunto(s)
Autofagia/fisiología , Enfermedades Neuromusculares/fisiopatología , Condicionamiento Físico Animal/fisiología , Proteostasis/fisiología , Animales , Antirreumáticos/farmacología , Autofagia/genética , Cloroquina/farmacología , Masculino , Ratones Noqueados , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/metabolismo , Proteolisis , Proteostasis/genética , Ratas Sprague-Dawley
11.
PLoS One ; 10(8): e0134844, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26302153

RESUMEN

Metabolic syndrome is a cluster of metabolic risk factors such as obesity, diabetes and cardiovascular diseases. Mitochondria is the main site of ATP production and its dysfunction leads to decreased oxidative phosphorylation, resulting in lipid accumulation and insulin resistance. Our group has demonstrated that kinins can modulate glucose and lipid metabolism as well as skeletal muscle mass. By using B2 receptor knockout mice (B2R-/-) we investigated whether kinin action affects weight gain and physical performance of the animals. Our results show that B2R-/- mice are resistant to high fat diet-induced obesity, have higher glucose tolerance as well as increased mitochondrial mass. These features are accompanied by higher energy expenditure and a lower feed efficiency associated with an increase in the proportion of type I fibers and intermediary fibers characterized by higher mitochondrial content and increased expression of genes related to oxidative metabolism. Additionally, the increased percentage of oxidative skeletal muscle fibers and mitochondrial apparatus in B2R-/- mice is coupled with a higher aerobic exercise performance. Taken together, our data give support to the involvement of kinins in skeletal muscle fiber type distribution and muscle metabolism, which ultimately protects against fat-induced obesity and improves aerobic exercise performance.


Asunto(s)
Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Receptor de Bradiquinina B2/fisiología , Animales , Dieta Alta en Grasa , Expresión Génica/fisiología , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Leptina/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/fisiología , Consumo de Oxígeno/fisiología
12.
J Transl Med ; 13: 76, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25889299

RESUMEN

BACKGROUND: Activation of protein kinase AKT is required for cardioprotection by ischemic preconditioning, and transgenic overexpression of AKT protects the heart against ischemia. However, it is unknown whether acute pharmacological activation of AKT alone, using a therapeutically relevant strategy, induces cardioprotection. In this study we provide the first evidence to clarify this question. METHODS: We used a recently described specific activator of AKT, the small molecule SC79, to treat rat hearts submitted to ischemia and reperfusion. Initially, isolated rat hearts were perfused with increasing doses of SC79 to verify the magnitude of AKT activation. Low and high doses were determined and used to treat hearts submitted to ischemia (35 minutes) and reperfusion (60 minutes), in a randomized and blinded design. AKT activation was verified by western immunobloting. Metabolic profile was determined by cardiac ATP content and mitochondrial enzyme activity, while cytosolic levels of cytochrome C and caspase-3 activity were used as markers of apoptosis. Ischemic injury was assessed by quantification of infarct size and cardiac release of creatine kinase and lactate dehydrogenase. RESULTS: SC79 activated cardiac AKT within 30 minutes in a dose-dependent fashion. ATP content was largely reduced by ischemia, but was not rescued by SC79. Similarly, mitochondrial enzyme activity was not affected by SC79. SC79 administered before ischemia or at reperfusion did not prevent cytosolic accumulation of cytochrome C and overactivation of caspase-3. Finally, SC79 failed to reduce infarct size or release of cardiac injury biomarkers at reperfusion. CONCLUSION: We conclude that selective AKT activation by the synthetic molecule SC79 does not protect the rat heart against ischemic injury, indicating that acute pharmacological activation of AKT is not sufficient for cardioprotection.


Asunto(s)
Activadores de Enzimas/farmacología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Creatina Quinasa/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/enzimología , Miocardio/patología , Ratas Sprague-Dawley
13.
Int J Cardiol ; 177(2): 409-17, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25456576

RESUMEN

BACKGROUND: Understanding the intracellular mechanisms induced by remote ischemic preconditioning (RIPC) in the human left ventricle opens new possibilities for development of pharmacological cardioprotection against ischemia and reperfusion injury. In this study we investigated the effects of RIPC on mitochondrial function, activation of pro-survival protein kinase Akt and microRNA expression in left ventricular biopsies from patients undergoing coronary artery bypass surgery (CABG). METHODS: Sixty patients were randomized to control (n=30) or RIPC (n=30). A blood pressure cuff was applied to the arm of all patients preoperatively. The cuff remained deflated in control group, whereas RIPC was performed by 3 cycles of cuff inflation to 200 mm Hg for 5 min, separated by 5 min deflation intervals. Left ventricular biopsies were obtained before and 15 min after aortic declamping. The primary outcome was mitochondrial respiration measured in situ. Secondary outcomes were activation of protein kinase Akt, assessed by western immunoblotting, and expression of microRNAs assessed by array and real-time polymerase chain reaction. RESULTS: Mitochondrial respiration was preserved during surgery in patients receiving RIPC (+0.2 µmol O2/min/g, p=0.69), and reduced by 15% in controls (-1.5 µmol O2/min/g, p=0.02). Furthermore, RIPC activated protein kinase Akt before aortic clamping (difference from control +43.3%, p=0.04), followed by increased phosphorylation of Akt substrates at reperfusion (+26.8%, p<0.01). No differences were observed in microRNA expression. CONCLUSIONS: RIPC preserves mitochondrial function and activates pro-survival protein kinase Akt in left ventricle of patients undergoing CABG. Modulation of mitochondrial function and Akt activation should be further explored as cardioprotective drug targets. CLINICAL TRIAL REGISTRATION: http://www.clinicaltrials.gov, unique identifier: NCT01308138.


Asunto(s)
Puente de Arteria Coronaria/métodos , Ventrículos Cardíacos/enzimología , Precondicionamiento Isquémico Miocárdico/métodos , Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Robótica/métodos , Anciano , Método Doble Ciego , Activación Enzimática/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monitoreo Intraoperatorio/métodos , Estudios Prospectivos
14.
Int J Cardiol ; 175(3): 499-507, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25023789

RESUMEN

BACKGROUND: Skeletal muscle wasting is associated with poor prognosis and increased mortality in heart failure (HF) patients. Glycolytic muscles are more susceptible to catabolic wasting than oxidative ones. This is particularly important in HF since glycolytic muscle wasting is associated with increased levels of reactive oxygen species (ROS). However, the main ROS sources involved in muscle redox imbalance in HF have not been characterized. Therefore, we hypothesized that NADPH oxidases would be hyperactivated in the plantaris muscle of infarcted rats, contributing to oxidative stress and hyperactivation of the ubiquitin-proteasome system (UPS), ultimately leading to atrophy. METHODS: Rats were submitted to myocardial infarction (MI) or Sham surgery. Four weeks after surgery, MI and Sham groups underwent eight weeks of treatment with apocynin, a NADPH oxidase inhibitor, or placebo. NADPH oxidase activity, oxidative stress markers, NF-κB activity, p38 MAPK phosphorylation, mRNA and sarcolemmal protein levels of NADPH oxidase components, UPS activation and fiber cross-sectional area were assessed in the plantaris muscle. RESULTS: The plantaris of MI rats displayed atrophy associated with increased Nox2 mRNA and sarcolemmal protein levels, NADPH oxidase activity, ROS production, lipid hydroperoxides levels, NF-κB activity, p38 MAPK phosphorylation and UPS activation. NADPH oxidase inhibition by apocynin prevented MI-induced skeletal muscle atrophy by reducing ROS production, NF-κB hyperactivation, p38 MAPK phosphorylation and proteasomal hyperactivity. CONCLUSION: Our data provide evidence for NADPH oxidase hyperactivation as an important source of ROS production leading to plantaris atrophy in heart failure rats, suggesting that this enzyme complex plays key role in skeletal muscle wasting in HF.


Asunto(s)
Insuficiencia Cardíaca/enzimología , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/enzimología , Atrofia Muscular/enzimología , NADPH Oxidasas/metabolismo , Animales , Activación Enzimática/fisiología , Insuficiencia Cardíaca/patología , Masculino , Músculo Esquelético/patología , Atrofia Muscular/patología , NADPH Oxidasa 2 , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
15.
PLoS One ; 9(1): e85820, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24427319

RESUMEN

BACKGROUND: Heart failure (HF)-induced skeletal muscle atrophy is often associated to exercise intolerance and poor prognosis. Better understanding of the molecular mechanisms underlying HF-induced muscle atrophy may contribute to the development of pharmacological strategies to prevent or treat such condition. It has been shown that autophagy-lysosome system is an important mechanism for maintenance of muscle mass. However, its role in HF-induced myopathy has not been addressed yet. Therefore, the aim of the present study was to evaluate autophagy signaling in myocardial infarction (MI)-induced muscle atrophy in rats. METHODS/PRINCIPAL FINDINGS: Wistar rats underwent MI or Sham surgeries, and after 12 weeks were submitted to echocardiography, exercise tolerance and histology evaluations. Cathepsin L activity and expression of autophagy-related genes and proteins were assessed in soleus and plantaris muscles by fluorimetric assay, qRT-PCR and immunoblotting, respectively. MI rats displayed exercise intolerance, left ventricular dysfunction and dilation, thereby suggesting the presence of HF. The key findings of the present study were: a) upregulation of autophagy-related genes (GABARAPL1, ATG7, BNIP3, CTSL1 and LAMP2) was observed only in plantaris while muscle atrophy was observed in both soleus and plantaris muscles, and b) Cathepsin L activity, Bnip3 and Fis1 protein levels, and levels of lipid hydroperoxides were increased specifically in plantaris muscle of MI rats. CONCLUSIONS: Altogether our results provide evidence for autophagy signaling regulation in HF-induced plantaris atrophy but not soleus atrophy. Therefore, autophagy-lysosome system is differentially regulated in atrophic muscles comprising different fiber-types and metabolic characteristics.


Asunto(s)
Autofagia , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Infarto del Miocardio/complicaciones , Transducción de Señal , Animales , Autofagia/genética , Biomarcadores , Catepsina L/metabolismo , Ecocardiografía , Regulación de la Expresión Génica , Masculino , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Atrofia Muscular/patología , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/fisiopatología , Estrés Oxidativo , Condicionamiento Físico Animal , Ratas , Transcriptoma
16.
J Appl Physiol (1985) ; 114(8): 1029-41, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23429866

RESUMEN

Poor skeletal muscle performance was shown to strongly predict mortality and long-term prognosis in a variety of diseases, including heart failure (HF). Despite the known benefits of aerobic exercise training (AET) in improving the skeletal muscle phenotype in HF, the optimal exercise intensity to elicit maximal outcomes is still under debate. Therefore, the aim of the present study was to compare the effects of high-intensity AET with those of a moderate-intensity protocol on skeletal muscle of infarcted rats. Wistar rats underwent myocardial infarction (MI) or sham surgery. MI groups were submitted either to an untrained (MI-UNT); moderate-intensity (MI-CMT, 60% Vo(2)(max)); or matched volume, high-intensity AET (MI-HIT, intervals at 85% Vo(2)(max)) protocol. High-intensity AET (HIT) was superior to moderate-intensity AET (CMT) in improving aerobic capacity, assessed by treadmill running tests. Cardiac contractile function, measured by echocardiography, was equally improved by both AET protocols. CMT and HIT prevented the MI-induced decay of skeletal muscle citrate synthase and hexokinase maximal activities, and increased glycogen content, without significant differences between protocols. Similar improvements in skeletal muscle redox balance and deactivation of the ubiquitin-proteasome system were also observed after CMT and HIT. Such intracellular findings were accompanied by prevented skeletal muscle atrophy in both MI-CMT and MI-HIT groups, whereas no major differences were observed between protocols. Taken together, our data suggest that despite superior effects of HIT in improving functional capacity, skeletal muscle adaptations were remarkably similar among protocols, leading to the conclusion that skeletal myopathy in infarcted rats was equally prevented by either moderate-intensity or high-intensity AET.


Asunto(s)
Terapia por Ejercicio/métodos , Contracción Muscular , Músculo Esquelético/fisiopatología , Atrofia Muscular/prevención & control , Infarto del Miocardio/terapia , Animales , Citrato (si)-Sintasa/metabolismo , Modelos Animales de Enfermedad , Tolerancia al Ejercicio , Glucógeno/metabolismo , Hexoquinasa/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/diagnóstico por imagen , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Contracción Miocárdica , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Oxidación-Reducción , Consumo de Oxígeno , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Ratas Wistar , Ultrasonografía
17.
PLoS One ; 7(8): e41701, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22870245

RESUMEN

BACKGROUND: Heart failure (HF) is known to lead to skeletal muscle atrophy and dysfunction. However, intracellular mechanisms underlying HF-induced myopathy are not fully understood. We hypothesized that HF would increase oxidative stress and ubiquitin-proteasome system (UPS) activation in skeletal muscle of sympathetic hyperactivity mouse model. We also tested the hypothesis that aerobic exercise training (AET) would reestablish UPS activation in mice and human HF. METHODS/PRINCIPAL FINDINGS: Time-course evaluation of plantaris muscle cross-sectional area, lipid hydroperoxidation, protein carbonylation and chymotrypsin-like proteasome activity was performed in a mouse model of sympathetic hyperactivity-induced HF. At the 7(th) month of age, HF mice displayed skeletal muscle atrophy, increased oxidative stress and UPS overactivation. Moderate-intensity AET restored lipid hydroperoxides and carbonylated protein levels paralleled by reduced E3 ligases mRNA levels, and reestablished chymotrypsin-like proteasome activity and plantaris trophicity. In human HF (patients randomized to sedentary or moderate-intensity AET protocol), skeletal muscle chymotrypsin-like proteasome activity was also increased and AET restored it to healthy control subjects' levels. CONCLUSIONS: Collectively, our data provide evidence that AET effectively counteracts redox imbalance and UPS overactivation, preventing skeletal myopathy and exercise intolerance in sympathetic hyperactivity-induced HF in mice. Of particular interest, AET attenuates skeletal muscle proteasome activity paralleled by improved aerobic capacity in HF patients, which is not achieved by drug treatment itself. Altogether these findings strengthen the clinical relevance of AET in the treatment of HF.


Asunto(s)
Terapia por Ejercicio , Proteínas Musculares/metabolismo , Atrofia Muscular , Estrés Oxidativo , Condicionamiento Físico Animal , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Femenino , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos , Peroxidación de Lípido/genética , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Atrofia Muscular/etiología , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Atrofia Muscular/terapia , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética
18.
J Appl Physiol (1985) ; 112(11): 1839-46, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22461440

RESUMEN

Aerobic exercise training (AET) is an important mechanical stimulus that modulates skeletal muscle protein turnover, leading to structural rearrangement. Since the ubiquitin-proteasome system (UPS) and calpain system are major proteolytic pathways involved in protein turnover, we aimed to investigate the effects of intensity-controlled AET on the skeletal muscle UPS and calpain system and their association to training-induced structural adaptations. Long-lasting effects of AET were studied in C57BL/6J mice after 2 or 8 wk of AET. Plantaris cross-sectional area (CSA) and capillarization were assessed by myosin ATPase staining. mRNA and protein expression levels of main components of the UPS and calpain system were evaluated in plantaris by real-time PCR and Western immunoblotting, respectively. No proteolytic system activation was observed after 2 wk of AET. Eight weeks of AET resulted in improved running capacity, plantaris capillarization, and CSA. Muscle RING finger-1 mRNA expression was increased in 8-wk-trained mice. Accordingly, elevated 26S proteasome activity was observed in the 8-wk-trained group, without accumulation of ubiquitinated or carbonylated proteins. In addition, calpain abundance was increased by 8 wk of AET, whereas no difference was observed in its endogenous inhibitor calpastatin. Taken together, our findings indicate that skeletal muscle enhancements, as evidenced by increased running capacity, plantaris capillarization, and CSA, occurred in spite of the upregulated UPS and calpain system, suggesting that overactivation of skeletal muscle proteolytic systems is not restricted to atrophying states. Our data provide evidence for the contribution of the UPS and calpain system to metabolic turnover of myofibrillar proteins and skeletal muscle adaptations to AET.


Asunto(s)
Calpaína/biosíntesis , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Complejo de la Endopetidasa Proteasomal/biosíntesis , Ubiquitina/biosíntesis , Regulación hacia Arriba/fisiología , Animales , Prueba de Esfuerzo/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/enzimología , Condicionamiento Físico Animal/métodos
19.
Life Sci ; 88(13-14): 578-85, 2011 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-21277865

RESUMEN

AIMS: The clinical benefits of angiotensin II type 1 (AT1) receptor blockers (ARB) in heart failure (HF) include cardiac anti-remodeling and improved ventricular function. However, the cellular mechanisms underlying the benefits of ARB on ventricular function need to be better clarified. In the present manuscript, we evaluated the effects of AT1 receptor blockade on the net balance of Ca(2+) handling proteins in hearts of mice lacking α(2A) and α(2C) adrenoceptors (α(2A)/α(2C)ARKO), which develop sympathetic hyperactivity (SH) induced-HF. MAIN METHODS: A cohort of male wild-type (WT) and congenic α(2A)/α(2C)ARKO mice in a C57BL6/J genetic background (5-7mo of age) was randomly assigned to receive either placebo or ARB (Losartan, 10mg/kg for 8wks). Ventricular function (VF) was assessed by echocardiography, and cardiac myocyte width and ventricular fibrosis by a computer-assisted morphometric system. Sarcoplasmic reticulum Ca(2+) ATPase (SERCA2), phospholamban (PLN), phospho-Ser(16)-PLN, phospho-Thr(17)-PLN, phosphatase 1 (PP1), Na(+)-Ca(2+) exchanger (NCX), Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and phospho-Thr(286)-CaMKII were analyzed by Western blot. KEY FINDINGS: α(2A)/α(2C)ARKO mice displayed ventricular dysfunction, cardiomyocyte hypertrophy and cardiac fibrosis paralleled by decreased SERCA2 and increased phospho-Thr(17)-PLN, CaMKII, phospho-Thr(286)-CaMKII and NCX levels. ARB induced anti-cardiac remodeling effect and improved VF in α(2A)/α(2C)ARKO associated with increased SERCA2 and phospho-Ser(16)-PLN levels, and SERCA2:NCX ratio. Additionally, ARB decreased phospho-Thr(17)-PLN levels as well as reestablished NCX, CaMKII and phospho-Thr(286)-CaMKII toward WT levels. SIGNIFICANCE: Altogether, these data provide new insights on intracellular Ca(2+) regulatory mechanisms underlying improved ventricular function by ARB therapy in HF.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Proteínas de Unión al Calcio/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Adrenérgicos alfa 2/fisiología , Sistema Nervioso Simpático/fisiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Western Blotting , Proteínas de Unión al Calcio/genética , Ecocardiografía , Prueba de Esfuerzo , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Receptores Adrenérgicos alfa 2/genética , Sistema Nervioso Simpático/metabolismo , Remodelación Ventricular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...