Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-37364662

RESUMEN

Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme involved in reactive aldehyde detoxification. Approximately 560 million people (about 8% of the world's population) carry a point mutation in the aldehyde dehydrogenase 2 gene (ALDH2), identified as ALDH2*2, which leads to decreased ALDH2 catalytic activity. ALDH2*2 variant is associated with an accumulation of toxic reactive aldehydes and consequent disruption of cellular metabolism, which contributes to the establishment and progression of several degenerative diseases. Consequences of aldehyde accumulation include impaired mitochondrial functional, hindered anabolic signaling in the skeletal muscle, impaired cardiovascular and pulmonary function, and reduced osteoblastogenesis. Considering that aldehydes are endogenously produced through redox processes, it is expected that conditions that have a high energy demand, such as exercise, might be affected by impaired aldehyde clearance in ALDH2*2 individuals. Despite the large body of evidence supporting the importance of ALDH2 to ethanol metabolism, redox homeostasis and overall health, specific research investigating the impact of ALDH2*2 on phenotypes relevant to exercise performance are notoriously scarce. In this commentary, we highlight the consolidated knowledge on the impact of ALDH2*2 on physiological processes that are relevant to exercise.


Asunto(s)
Aldehído Deshidrogenasa , Aldehídos , Animales , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Aldehídos/metabolismo , Músculo Esquelético/metabolismo , Oxidación-Reducción
2.
Proc Natl Acad Sci U S A ; 120(2): e2204750120, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36595699

RESUMEN

Exercise is a nonpharmacological intervention that improves health during aging and a valuable tool in the diagnostics of aging-related diseases. In muscle, exercise transiently alters mitochondrial functionality and metabolism. Mitochondrial fission and fusion are critical effectors of mitochondrial plasticity, which allows a fine-tuned regulation of organelle connectiveness, size, and function. Here we have investigated the role of mitochondrial dynamics during exercise in the model organism Caenorhabditis elegans. We show that in body-wall muscle, a single exercise session induces a cycle of mitochondrial fragmentation followed by fusion after a recovery period, and that daily exercise sessions delay the mitochondrial fragmentation and physical fitness decline that occur with aging. Maintenance of proper mitochondrial dynamics is essential for physical fitness, its enhancement by exercise training, and exercise-induced remodeling of the proteome. Surprisingly, among the long-lived genotypes we analyzed (isp-1,nuo-6, daf-2, eat-2, and CA-AAK-2), constitutive activation of AMP-activated protein kinase (AMPK) uniquely preserves physical fitness during aging, a benefit that is abolished by impairment of mitochondrial fission or fusion. AMPK is also required for physical fitness to be enhanced by exercise, with our findings together suggesting that exercise may enhance muscle function through AMPK regulation of mitochondrial dynamics. Our results indicate that mitochondrial connectivity and the mitochondrial dynamics cycle are essential for maintaining physical fitness and exercise responsiveness during aging and suggest that AMPK activation may recapitulate some exercise benefits. Targeting mechanisms to optimize mitochondrial fission and fusion, as well as AMPK activation, may represent promising strategies for promoting muscle function during aging.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Dinámicas Mitocondriales , Animales , Dinámicas Mitocondriales/fisiología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Envejecimiento/fisiología , Caenorhabditis elegans/metabolismo , Ejercicio Físico , Aptitud Física , Músculo Esquelético/metabolismo
3.
Cells ; 11(9)2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35563852

RESUMEN

Mitochondria are major intracellular hubs distributed throughout the cell that play a key role in the spatiotemporal coordination and propagation of signalling events, ensuring that homeostasis is met at baseline or under environmental pressure [...].


Asunto(s)
Mitocondrias , Homeostasis
4.
Antioxid Redox Signal ; 36(13-15): 844-863, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35044229

RESUMEN

Significance: Mitochondria play a critical role in the physiology of the heart by controlling cardiac metabolism, function, and remodeling. Accumulation of fragmented and damaged mitochondria is a hallmark of cardiac diseases. Recent Advances: Disruption of quality control systems that maintain mitochondrial number, size, and shape through fission/fusion balance and mitophagy results in dysfunctional mitochondria, defective mitochondrial segregation, impaired cardiac bioenergetics, and excessive oxidative stress. Critical Issues: Pharmacological tools that improve the cardiac pool of healthy mitochondria through inhibition of excessive mitochondrial fission, boosting mitochondrial fusion, or increasing the clearance of damaged mitochondria have emerged as promising approaches to improve the prognosis of heart diseases. Future Directions: There is a reasonable amount of preclinical evidence supporting the effectiveness of molecules targeting mitochondrial fission and fusion to treat cardiac diseases. The current and future challenges are turning these lead molecules into treatments. Clinical studies focusing on acute (i.e., myocardial infarction) and chronic (i.e., heart failure) cardiac diseases are needed to validate the effectiveness of such strategies in improving mitochondrial morphology, metabolism, and cardiac function. Antioxid. Redox Signal. 36, 844-863.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Insuficiencia Cardíaca/metabolismo , Humanos , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Mitofagia , Infarto del Miocardio/metabolismo
5.
Redox Biol ; 44: 102016, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34038814

RESUMEN

Histidine-containing dipeptides (HCDs) are abundantly expressed in striated muscles. Although important properties have been ascribed to HCDs, including H+ buffering, regulation of Ca2+ transients and protection against oxidative stress, it remains unknown whether they play relevant functions in vivo. To investigate the in vivo roles of HCDs, we developed the first carnosine synthase knockout (CARNS1-/-) rat strain to investigate the impact of an absence of HCDs on skeletal and cardiac muscle function. Male wild-type (WT) and knockout rats (4 months-old) were used. Skeletal muscle function was assessed by an exercise tolerance test, contractile function in situ and muscle buffering capacity in vitro. Cardiac function was assessed in vivo by echocardiography and cardiac electrical activity by electrocardiography. Cardiomyocyte contractile function was assessed in isolated cardiomyocytes by measuring sarcomere contractility, along with the determination of Ca2+ transient. Markers of oxidative stress, mitochondrial function and expression of proteins were also evaluated in cardiac muscle. Animals were supplemented with carnosine (1.8% in drinking water for 12 weeks) in an attempt to rescue tissue HCDs levels and function. CARNS1-/- resulted in the complete absence of carnosine and anserine, but it did not affect exercise capacity, skeletal muscle force production, fatigability or buffering capacity in vitro, indicating that these are not essential for pH regulation and function in skeletal muscle. In cardiac muscle, however, CARNS1-/- resulted in a significant impairment of contractile function, which was confirmed both in vivo and ex vivo in isolated sarcomeres. Impaired systolic and diastolic dysfunction were accompanied by reduced intracellular Ca2+ peaks and slowed Ca2+ removal, but not by increased markers of oxidative stress or impaired mitochondrial respiration. No relevant increases in muscle carnosine content were observed after carnosine supplementation. Results show that a primary function of HCDs in cardiac muscle is the regulation of Ca2+ handling and excitation-contraction coupling.


Asunto(s)
Carnosina , Dipéptidos , Animales , Anserina , Histidina , Masculino , Músculo Esquelético , Miocitos Cardíacos , Ratas
6.
Free Radic Biol Med ; 129: 155-168, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30227272

RESUMEN

Mitochondrial dysfunction characterized by impaired bioenergetics, oxidative stress and aldehydic load is a hallmark of heart failure. Recently, different research groups have provided evidence that selective activation of mitochondrial detoxifying systems that counteract excessive accumulation of ROS, RNS and reactive aldehydes is sufficient to stop cardiac degeneration upon chronic stress, such as heart failure. Therefore, pharmacological and non-pharmacological approaches targeting mitochondria detoxification may play a critical role in the prevention or treatment of heart failure. In this review we discuss the most recent findings on the central role of mitochondrial dysfunction, oxidative stress and aldehydic load in heart failure, highlighting the most recent preclinical and clinical studies using mitochondria-targeted molecules and exercise training as effective tools against heart failure.


Asunto(s)
Antioxidantes/uso terapéutico , Materiales Biomiméticos/uso terapéutico , Cardiotónicos/uso terapéutico , Insuficiencia Cardíaca/terapia , Mitocondrias Cardíacas/efectos de los fármacos , Ubiquinona/análogos & derivados , Aldehídos/antagonistas & inhibidores , Aldehídos/metabolismo , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Ejercicio Físico , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Malondialdehído/antagonistas & inhibidores , Malondialdehído/metabolismo , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Estrés Oxidativo/efectos de los fármacos , Especies de Nitrógeno Reactivo/antagonistas & inhibidores , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/química , Ubiquinona/uso terapéutico
7.
Cardiovasc Res ; 114(7): 1006-1015, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29579152

RESUMEN

Aims: We previously demonstrated that acute ethanol administration protects the heart from ischaemia/reperfusion (I/R) injury thorough activation of aldehyde dehydrogenase 2 (ALDH2). Here, we characterized the role of acetaldehyde, an intermediate product from ethanol metabolism, and its metabolizing enzyme, ALDH2, in an ex vivo model of cardiac I/R injury. Methods and results: We used a combination of homozygous knock-in mice (ALDH2*2), carrying the human inactivating point mutation ALDH2 (E487K), and a direct activator of ALDH2, Alda-1, to investigate the cardiac effect of acetaldehyde. The ALDH2*2 mice have impaired acetaldehyde clearance, recapitulating the human phenotype. Yet, we found a similar infarct size in wild type (WT) and ALDH2*2 mice. Similar to ethanol-induced preconditioning, pre-treatment with 50 µM acetaldehyde increased ALDH2 activity and reduced cardiac injury in hearts of WT mice without affecting cardiac acetaldehyde levels. However, acetaldehyde pre-treatment of hearts of ALDH2*2 mice resulted in a three-fold increase in cardiac acetaldehyde levels and exacerbated I/R injury. Therefore, exogenous acetaldehyde appears to have a bimodal effect in I/R, depending on the ALDH2 genotype. Further supporting an ALDH2 role in cardiac preconditioning, pharmacological ALDH2 inhibition abolished ethanol-induced cardioprotection in hearts of WT mice, whereas a selective activator, Alda-1, protected ALDH2*2 against ethanol-induced cardiotoxicity. Finally, either genetic or pharmacological inhibition of ALDH2 mitigated ischaemic preconditioning. Conclusion: Taken together, our findings suggest that low levels of acetaldehyde are cardioprotective whereas high levels are damaging in an ex vivo model of I/R injury and that ALDH2 is a major, but not the only, regulator of cardiac acetaldehyde levels and protection from I/R.


Asunto(s)
Acetaldehído/farmacología , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Acetaldehído/metabolismo , Acetaldehído/toxicidad , Aldehído Deshidrogenasa Mitocondrial/genética , Animales , Cardiotoxicidad , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Técnicas de Sustitución del Gen , Genotipo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Fenotipo , Mutación Puntual , Ratas , Factores de Tiempo
8.
Rev Assoc Med Bras (1992) ; 63(2): 180-189, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28355380

RESUMEN

Induced pluripotent stem cells (iPSCs) are somatic cells reprogrammed into an embryonic-like pluripotent state by the expression of specific transcription factors. iPSC technology is expected to revolutionize regenerative medicine in the near future. Despite the fact that these cells have the capacity to self-renew, they present low efficiency of reprogramming. Recent studies have demonstrated that the previous somatic epigenetic signature is a limiting factor in iPSC performance. Indeed, the process of effective reprogramming involves a complete remodeling of the existing somatic epigenetic memory, followed by the establishment of a "new epigenetic signature" that complies with the new type of cell to be differentiated. Therefore, further investigations of epigenetic modifications associated with iPSC reprogramming are required in an attempt to improve their self-renew capacity and potency, as well as their application in regenerative medicine, with a new strategy to reduce the damage in degenerative diseases. Our review aimed to summarize the most recent findings on epigenetics and iPSC, focusing on DNA methylation, histone modifications and microRNAs, highlighting their potential in translating cell therapy into clinics.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Medicina Regenerativa , Metilación de ADN , Epigénesis Genética , Histonas , Humanos , MicroARNs
9.
Rev. Assoc. Med. Bras. (1992) ; 63(2): 180-189, Feb. 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-842535

RESUMEN

Summary Induced pluripotent stem cells (iPSCs) are somatic cells reprogrammed into an embryonic-like pluripotent state by the expression of specific transcription factors. iPSC technology is expected to revolutionize regenerative medicine in the near future. Despite the fact that these cells have the capacity to self-renew, they present low efficiency of reprogramming. Recent studies have demonstrated that the previous somatic epigenetic signature is a limiting factor in iPSC performance. Indeed, the process of effective reprogramming involves a complete remodeling of the existing somatic epigenetic memory, followed by the establishment of a "new epigenetic signature" that complies with the new type of cell to be differentiated. Therefore, further investigations of epigenetic modifications associated with iPSC reprogramming are required in an attempt to improve their self-renew capacity and potency, as well as their application in regenerative medicine, with a new strategy to reduce the damage in degenerative diseases. Our review aimed to summarize the most recent findings on epigenetics and iPSC, focusing on DNA methylation, histone modifications and microRNAs, highlighting their potential in translating cell therapy into clinics.


Resumo As células-tronco de pluripotência induzida (CTPI) ou do inglês induced pluripotent stem cells (iPSCs) são células somáticas reprogramadas para o estado embrionário por meio da expressão de fatores ectópicos de transcrição específicos, tornando-as um alvo promissor para a medicina regenerativa. Apesar das CTPI compartilharem características embrionárias, como pluripotência e capacidade de autorrenovação, elas possuem uma baixa eficiência de reprogramação, sendo a memória epigenética uma das principais barreiras nesse processo. A epigenética é caracterizada por alterações reversíveis e herdáveis no genoma funcional que não alteram a sequência de nucleotídeos do DNA. Dentre as diferentes modificações epigenéticas, destacam-se metilação de DNA, alterações em histonas e microRNA. Atualmente, sabe-se que o processo de reprogramação efetivo das CTPI envolve um completo remodelamento da memória epigenética somática existente, seguido pelo estabelecimento de uma "assinatura epigenética" que esteja de acordo com o novo tipo de célula a ser diferenciada. Modificações epigenéticas personalizadas são capazes de melhorar o rendimento e a efetividade das CTPI geradas, abrindo uma nova perspectiva para a terapia celular. Nesta revisão reunimos as principais informações sobre os fatores epigenéticos que afetam a reprogramação das CTPI, bem como seus benefícios na aplicação da terapia celular.


Asunto(s)
Humanos , Medicina Regenerativa , Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Histonas , Metilación de ADN , MicroARNs , Epigénesis Genética
10.
Oxid Med Cell Longev ; 2015: 464195, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25954323

RESUMEN

BACKGROUND: We previously reported that exercise training (ET) facilitates the clearance of damaged proteins in heart failure. Here, we characterized the impact of ET on cardiac protein quality control during compensated ventricular hypertrophy in spontaneously hypertensive rats (SHR). METHODS AND RESULTS: SHR were randomly assigned into sedentary and swimming-trained groups. Sedentary SHR displayed cardiac hypertrophy with preserved ventricular function compared to normotensive rats, characterizing a compensated cardiac hypertrophy. Hypertensive rats presented signs of cardiac oxidative stress, depicted by increased lipid peroxidation. However, these changes were not followed by accumulation of lipid peroxidation-generated reactive aldehydes and damaged proteins. This scenario was explained, at least in part, by the increased catalytic activity of both aldehyde dehydrogenase 2 (ALDH2) and proteasome. Of interest, ET exacerbated cardiac hypertrophy, improved ventricular function, induced resting bradycardia, and decreased blood pressure in SHR. These changes were accompanied by reduced cardiac oxidative stress and a consequent decrease in ALDH2 and proteasome activities, without affecting small chaperones levels and apoptosis in SHR. CONCLUSION: Increased cardiac ALDH2 and proteasomal activities counteract the deleterious effect of excessive oxidative stress in hypertension-induced compensated cardiac hypertrophy in rats. ET has a positive effect in reducing cardiac oxidative stress without affecting protein quality control.


Asunto(s)
Aldehídos/metabolismo , Cardiomegalia/etiología , Hipertensión/complicaciones , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa Mitocondrial , Animales , Presión Sanguínea , Cardiomegalia/metabolismo , Frecuencia Cardíaca , Peroxidación de Lípido , Masculino , Proteínas Mitocondriales/metabolismo , Condicionamiento Físico Animal , Complejo de la Endopetidasa Proteasomal/metabolismo , Carbonilación Proteica , Ratas , Ratas Endogámicas SHR
11.
PLoS One ; 9(3): e90576, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24594607

RESUMEN

Inflammation enhances the peripheral analgesic efficacy of opioid drugs, but the mechanisms involved in this phenomenon have not been fully elucidated. Crotalphine (CRP), a peptide that was first isolated from South American rattlesnake C.d. terrificus venom, induces a potent and long-lasting anti-nociceptive effect that is mediated by the activation of peripheral opioid receptors. Because the high efficacy of CRP is only observed in the presence of inflammation, we aimed to elucidate the mechanisms involved in the CRP anti-nociceptive effect induced by inflammation. Using real-time RT-PCR, western blot analysis and ELISA assays, we demonstrate that the intraplantar injection of prostaglandin E2 (PGE2) increases the mRNA and protein levels of the µ- and κ-opioid receptors in the dorsal root ganglia (DRG) and paw tissue of rats within 3 h of the injection. Using conformation state-sensitive antibodies that recognize activated opioid receptors, we show that PGE2, alone does not increase the activation of these opioid receptors but that in the presence of PGE2, the activation of specific opioid receptors by CRP and selective µ- and κ-opioid receptor agonists (positive controls) increases. Furthermore, PGE2 down-regulated the expression and activation of the δ-opioid receptor. CRP increased the level of activated mitogen-activated protein kinases in cultured DRG neurons, and this increase was dependent on the activation of protein kinase Cζ. This CRP effect was much more prominent when the cells were pretreated with PGE2. These results indicate that the expression and activation of peripheral opioid receptors by opioid-like drugs can be up- or down-regulated in the presence of an acute injury and that acute tissue injury enhances the efficacy of peripheral opioids.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Dinoprostona , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Péptidos/uso terapéutico , Receptores Opioides/genética , Analgésicos Opioides/aislamiento & purificación , Analgésicos Opioides/farmacología , Animales , Crotalus/metabolismo , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/inmunología , Ganglios Espinales/metabolismo , Hiperalgesia/genética , Hiperalgesia/inmunología , Masculino , Péptidos/aislamiento & purificación , Péptidos/farmacología , Ratas , Ratas Wistar , Receptores Opioides/agonistas , Receptores Opioides/inmunología
12.
Physiol Rev ; 94(1): 1-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24382882

RESUMEN

A family of detoxifying enzymes called aldehyde dehydrogenases (ALDHs) has been a subject of recent interest, as its role in detoxifying aldehydes that accumulate through metabolism and to which we are exposed from the environment has been elucidated. Although the human genome has 19 ALDH genes, one ALDH emerges as a particularly important enzyme in a variety of human pathologies. This ALDH, ALDH2, is located in the mitochondrial matrix with much known about its role in ethanol metabolism. Less known is a new body of research to be discussed in this review, suggesting that ALDH2 dysfunction may contribute to a variety of human diseases including cardiovascular diseases, diabetes, neurodegenerative diseases, stroke, and cancer. Recent studies suggest that ALDH2 dysfunction is also associated with Fanconi anemia, pain, osteoporosis, and the process of aging. Furthermore, an ALDH2 inactivating mutation (termed ALDH2*2) is the most common single point mutation in humans, and epidemiological studies suggest a correlation between this inactivating mutation and increased propensity for common human pathologies. These data together with studies in animal models and the use of new pharmacological tools that activate ALDH2 depict a new picture related to ALDH2 as a critical health-promoting enzyme.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Envejecimiento , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa Mitocondrial , Animales , Humanos , Hígado/enzimología , Mutación/genética , Neoplasias/metabolismo
13.
Mol Endocrinol ; 27(12): 2055-65, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24176915

RESUMEN

Although it is well known that the thyroid hormone (T3) is an important positive regulator of cardiac function over a short term and that it also promotes deleterious effects over a long term, the molecular mechanisms for such effects are not yet well understood. Because most alterations in cardiac function are associated with changes in sarcomeric machinery, the present work was undertaken to find novel sarcomeric hot spots driven by T3 in the heart. A microarray analysis indicated that the M-band is a major hot spot, and the structural sarcomeric gene coding for the M-protein is severely down-regulated by T3. Real-time quantitative PCR-based measurements confirmed that T3 (1, 5, 50, and 100 physiological doses for 2 days) sharply decreased the M-protein gene and protein expression in vivo in a dose-dependent manner. Furthermore, the M-protein gene expression was elevated 3.4-fold in hypothyroid rats. Accordingly, T3 was able to rapidly and strongly reduce the M-protein gene expression in neonatal cardiomyocytes. Deletions at the M-protein promoter and bioinformatics approach suggested an area responsive to T3, which was confirmed by chromatin immunoprecipitation assay. Functional assays in cultured neonatal cardiomyocytes revealed that depletion of M-protein (by small interfering RNA) drives a severe decrease in speed of contraction. Interestingly, mRNA and protein levels of other M-band components, myomesin and embryonic-heart myomesin, were not altered by T3. We concluded that the M-protein expression is strongly and rapidly repressed by T3 in cardiomyocytes, which represents an important aspect for the basis of T3-dependent sarcomeric deleterious effects in the heart.


Asunto(s)
Cardiomegalia/genética , Conectina/genética , Regulación hacia Abajo/genética , Hormonas Tiroideas/farmacología , Animales , Animales Recién Nacidos , Secuencia de Bases , Cardiomegalia/etiología , Cardiomegalia/fisiopatología , Línea Celular , Células Cultivadas , Conectina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Hipotiroidismo/genética , Masculino , Ratones , Datos de Secuencia Molecular , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Hormona Tiroidea/metabolismo , Sarcómeros/metabolismo , Tirotoxicosis/complicaciones , Triyodotironina/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Rev. Soc. Cardiol. Estado de Säo Paulo ; 22(3,supl.A): 3-7, jul.-set. 2012. ilus
Artículo en Portugués | LILACS | ID: lil-682783

RESUMEN

A insuficiência cardíaca é uma síndrome de mau prognóstico, caracterizada por disfunçao cardíaca associada à intolerância aos esforços, retenção de fluído e redução de longividade. Recentemente, os conhecimentos sobre a fisiopatoplogia da insuficiência cardíaca estabeleceram que, além dos distúrbios hemodinâmicos e neuro-humorais associados à síndrome, alterações mitocondriais (denominadas mitocondriopatias) podem ser nocivas ao tecido cardíaco. Prejuízos no metabolismo energético mitocondrial, com consequente aumento no estresse oxidativo seguido de morte celular programada, são caracteristicas presentes na insuficiência cardíaca. Além disso, alterações na dinâmica mitocondrial, representada por um desequilíbrio entre os processos de fusão e fissão da organela, contribuem para o agravamento da síndrome, uma vez que esses eventos estão relacionados à manutenção da viabilidade celular. Uma importante estratégia não farmacológica utilizada no tratamento da insuficiência cardíaca é o treinamento físico aeróbico, que, além de contribuir para a melhora da função ventricular, com consequente aumento da tolerância aos esforços e na qualidade de vida desses pacientes, promove importantes adaptações mitocondriais no tecido cardíaco, destacando-se o aumento dos complexos respiratórios mitocondriais e a redução na produção de espécies reativas de oxigênio. Nesse artigo de revisão. será abordada a importância da biologia mitocondrial no contexto cardíaco, retratando aspectos funcionais e morfológicos da organela, bem como sua contribuição para o agravamento da insuficiência cardíaca. Será, também, abordado o papel do treinamento físico aeróbico na reversão da mitocondriopatia observada na síndrome.


Heart failure is a clinical syndrome of poor prognosis characterized by cardiac dysfunction, exercise intolerance, lung edema and reduced longevity. Over the last decades, the knowledge regarding heart failure pathophysiology has established that in addition to hemodynamic and neurohumoral disorders, mitochondrial dysfunctions (called mitochondriopathy) can be deleterious to the heart. Impairment of mitochondrial energy metabolism, with consequent increase in oxidative stress and programmed cell death, is associated to cardiac deterioration in both human failing hearts and heart failure animal models. In addition, changes in mitochondrial dynamic, characterizated by an imbalance between the fusion and fisson processes of the organelle, contribute to the worsening of the pathology, since these events are related to the maintenance of cell viability. Endurance training has been recognized as an important adjuvant in the treatment of heart failure since it improves patient outcomes and quality of life. However, the mechanisms underlying exercise-induced beneficial effect on heart failure are not completely understood. In this review, we describe specific exercise training-mediated changes in mitochondrial function and morphology that contributes to better heart failure prognosis, including improved mitochondrial respiration, reduced oxidative stress and increased mitochondrial fussion.


Asunto(s)
Humanos , Animales , Ratas , Tolerancia al Ejercicio , Insuficiencia Cardíaca/fisiopatología , Mitocondrias Cardíacas/metabolismo , Ciclosporina , Metabolismo Energético
15.
Circ J ; 76(6): 1476-85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22453000

RESUMEN

BACKGROUND: Epsilon-protein kinase C (εPKC) protects the heart from ischemic injury. However, the mechanism(s) of εPKC cardioprotection is still unclear. Identification of the εPKC targets may aid in elucidating the εPKC-mediated cardioprotective mechanisms. Previous studies, using εPKC transgenic mice and difference in gel electrophoresis, identified proteins involved in glucose metabolism, the expression of which was modified by εPKC. Those studies were accompanied by metabolomic analysis, suggesting that increased glucose oxidation may be responsible for the cardioprotective effect of εPKC. Whether these εPKC-mediated alterations were because of differences in protein expression or phosphorylation was not determined. METHODS AND RESULTS: In the present study, we used an εPKC -specific activator peptide, ψεRACK, combined with phosphoproteomics, to find εPKC targets, and identified that the proteins whose phosphorylation was altered by selective activation of εPKC were mostly mitochondrial proteins. Analysis of the mitochondrial phosphoproteome led to the identification of 55 spots, corresponding to 37 individual proteins, exclusively phosphorylated, in the presence of ψεRACK. The majority of the proteins identified were involved in glucose and lipid metabolism, components of the respiratory chain as well as mitochondrial heat shock proteins. CONCLUSIONS: The protective effect of εPKC during ischemia involves phosphorylation of several mitochondrial proteins involved in glucose and lipid metabolism and oxidative phosphorylation. Regulation of these metabolic pathways by εPKC phosphorylation may lead to εPKC-mediated cardioprotection induced by ψεRACK.


Asunto(s)
Metabolismo Energético , Mitocondrias Cardíacas/enzimología , Isquemia Miocárdica/enzimología , Miocardio/enzimología , Proteína Quinasa C-epsilon/metabolismo , Animales , Citoprotección , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Activación Enzimática , Activadores de Enzimas/farmacología , Glucosa/metabolismo , Técnicas In Vitro , Metabolismo de los Lípidos , Isquemia Miocárdica/prevención & control , Oligopéptidos/farmacología , Fosforilación Oxidativa , Perfusión , Fosforilación , Proteómica/métodos , Ratas , Ratas Wistar , Transducción de Señal
16.
Front Biosci (Schol Ed) ; 4(2): 532-46, 2012 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-22202075

RESUMEN

Cardiac excitability and electrical activity are determined by the sum of individual ion channels, gap junctions and exchanger activities. Electrophysiological remodeling during heart disease involves changes in membrane properties of cardiomyocytes and is related to higher prevalence of arrhythmia-associated morbidity and mortality. Pharmacological and genetic manipulation of cardiac cells as well as animal models of cardiovascular diseases are used to identity changes in electrophysiological properties and the molecular mechanisms associated with the disease. Protein kinase C (PKC) and several other kinases play a pivotal role in cardiac electrophysiological remodeling. Therefore, identifying specific therapies that regulate these kinases is the main focus of current research. PKC, a family of serine/threonine kinases, has been implicated as potential signaling nodes associated with biochemical and biophysical stress in cardiovascular diseases. In this review, we describe the role of PKC isozymes that are involved in cardiac excitability and discuss both genetic and pharmacological tools that were used, their attributes and limitations. Selective and effective pharmacological interventions to normalize cardiac electrical activities and correct cardiac arrhythmias will be of great clinical benefit.


Asunto(s)
Miocardio/enzimología , Proteína Quinasa C/metabolismo , Animales , Enfermedades Cardiovasculares/enzimología , Uniones Comunicantes/enzimología , Humanos , Canales Iónicos/metabolismo , Isoenzimas , Miocitos Cardíacos/enzimología
17.
Sci Transl Med ; 3(107): 107ra111, 2011 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22049071

RESUMEN

Nitroglycerin, which treats impaired cardiac function through vasodilation as it is converted to nitric oxide, is used worldwide for patients with various ischemic and congestive cardiac diseases, including angina pectoris. Nevertheless, after continuous treatment, the benefits of nitroglycerin are limited by the development of tolerance to the drug. Nitroglycerin tolerance is a result of inactivation of aldehyde dehydrogenase 2 (ALDH2), an enzyme essential for cardioprotection in animals subjected to myocardial infarction. Here, we tested the hypothesis that the tolerance that develops as a result of sustained nitroglycerin treatment increases cardiac injury by subsequent myocardial infarction. In a rat model of myocardial infarction, 16 hours of prior, sustained nitroglycerin treatment resulted in infarcts that were twice as large as those in untreated control animals and in diminished cardiac function at 3 days and 2 weeks after the myocardial infarction. We also sought to identify a potential treatment to protect against this increased cardiac damage. Nitroglycerin inhibited ALDH2 activity in vitro, an effect that was blocked by Alda-1, an activator of ALDH2. Co-administration of Alda-1 with the nitroglycerin prevented the nitroglycerin-induced increase in cardiac dysfunction after myocardial infarction in rats, at least in part by enhancing metabolism of reactive aldehyde adducts that impair normal protein functions. If our animal studies showing that nitroglycerin tolerance increases cardiac injury upon ischemic insult are corroborated in humans, activators of ALDH2 such as Alda-1 may help to protect patients with myocardial infarction from this nitroglycerin-induced increase in cardiac injury while maintaining the cardiac benefits of the increased nitric oxide concentrations produced by nitroglycerin.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Nitroglicerina/efectos adversos , Nitroglicerina/uso terapéutico , Vasodilatadores/efectos adversos , Aldehído Deshidrogenasa Mitocondrial , Animales , Presión Sanguínea/fisiología , Muerte Celular/fisiología , Frecuencia Cardíaca/fisiología , Masculino , Carbonilación Proteica , Ratas , Ratas Wistar , Vasodilatadores/uso terapéutico
18.
J Cell Mol Med ; 15(8): 1769-77, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20874717

RESUMEN

Protein kinase C ßII (PKCßII) levels increase in the myocardium of patients with end-stage heart failure (HF). Also targeted overexpression of PKCßII in the myocardium of mice leads to dilated cardiomyopathy associated with inflammation, fibrosis and myocardial dysfunction. These reports suggest a deleterious role of PKCßII in HF development. Using a post-myocardial infarction (MI) model of HF in rats, we determined the benefit of chronic inhibition of PKCßII on the progression of HF over a period of 6 weeks after the onset of symptoms and the cellular basis for these effects. Four weeks after MI, rats with HF signs that were treated for 6 weeks with the PKCßII selective inhibitor (ßIIV5-3 conjugated to TAT(47-57) carrier peptide) (3 mg/kg/day) showed improved fractional shortening (from 21% to 35%) compared to control (TAT(47-57) carrier peptide alone). Formalin-fixed mid-ventricle tissue sections stained with picrosirius red, haematoxylin and eosin and toluidine blue dyes exhibited a 150% decrease in collagen deposition, a two-fold decrease in inflammation and a 30% reduction in mast cell degranulation, respectively, in rat hearts treated with the selective PKCßII inhibitor. Further, a 90% decrease in active TGFß1 and a significant reduction in SMAD2/3 phosphorylation indicated that the selective inhibition of PKCßII attenuates cardiac remodelling mediated by the TGF-SMAD signalling pathway. Therefore, sustained selective inhibition of PKCßII in a post-MI HF rat model improves cardiac function and is associated with inhibition of pathological myocardial remodelling.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Infarto del Miocardio/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Secuencia de Aminoácidos , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/etiología , Colágeno/metabolismo , Fibrosis/prevención & control , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Immunoblotting , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Masculino , Infarto del Miocardio/complicaciones , Miocardio/enzimología , Miocardio/metabolismo , Miocardio/patología , Fragmentos de Péptidos/química , Péptidos/química , Péptidos/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/química , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Inhibidores de Proteínas Quinasas/química , Ratas , Ratas Wistar , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
19.
J Mol Cell Cardiol ; 51(4): 479-84, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21035454

RESUMEN

Cardiac hypertrophy is a complex adaptive response to mechanical and neurohumoral stimuli and under continual stressor, it contributes to maladaptive responses, heart failure and death. Protein kinase C (PKC) and several other kinases play a role in the maladaptative cardiac responses, including cardiomyocyte hypertrophy, myocardial fibrosis and inflammation. Identifying specific therapies that regulate these kinases is a major focus of current research. PKC, a family of serine/threonine kinases, has emerged as potential mediators of hypertrophic stimuli associated with neurohumoral hyperactivity in heart failure. In this review, we describe the role of PKC isozymes that is involved in cardiac hypertrophy and heart failure. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure".


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Insuficiencia Cardíaca/tratamiento farmacológico , Miocardio/enzimología , Proteína Quinasa C-epsilon/metabolismo , Proteína Quinasa C/metabolismo , Animales , Cardiomegalia/enzimología , Insuficiencia Cardíaca/enzimología , Humanos , Isoenzimas/metabolismo , Terapia Molecular Dirigida , Proteína Quinasa C beta , Inhibidores de Proteínas Quinasas/uso terapéutico
20.
Hypertension ; 56(4): 629-35, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20644006

RESUMEN

Previous studies show that exercise training and caloric restriction improve cardiac function in obesity. However, the molecular mechanisms underlying this effect on cardiac function remain unknown. Thus, we studied the effect of exercise training and/or caloric restriction on cardiac function and Ca(2+) handling protein expression in obese rats. To accomplish this goal, male rats fed with a high-fat and sucrose diet for 25 weeks were randomly assigned into 4 groups: high-fat and sucrose diet, high-fat and sucrose diet and exercise training, caloric restriction, and exercise training and caloric restriction. An additional lean group was studied. The study was conducted for 10 weeks. Cardiac function was evaluated by echocardiography and Ca(2+) handling protein expression by Western blotting. Our results showed that visceral fat mass, circulating leptin, epinephrine, and norepinephrine levels were higher in rats on the high-fat and sucrose diet compared with the lean rats. Cardiac nitrate levels, reduced/oxidized glutathione, left ventricular fractional shortening, and protein expression of phosphorylated Ser(2808)-ryanodine receptor and Thr(17)-phospholamban were lower in rats on the high-fat and sucrose diet compared with lean rats. Exercise training and/or caloric restriction prevented increases in visceral fat mass, circulating leptin, epinephrine, and norepinephrine levels and prevented reduction in cardiac nitrate levels and reduced:oxidized glutathione ratio. Exercise training and/or caloric restriction prevented reduction in left ventricular fractional shortening and in phosphorylation of the Ser(2808)-ryanodine receptor and Thr(17)-phospholamban. These findings show that exercise training and/or caloric restriction prevent cardiac dysfunction in high-fat and sucrose diet rats, which seems to be attributed to decreased circulating neurohormone levels. In addition, this nonpharmacological paradigm prevents a reduction in the Ser(2808)-ryanodine receptor and Thr(17)-phospholamban phosphorylation and redox status.


Asunto(s)
Calcio/metabolismo , Restricción Calórica , Obesidad/prevención & control , Condicionamiento Físico Animal/fisiología , Animales , Presión Sanguínea/fisiología , Western Blotting , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Carbohidratos de la Dieta/administración & dosificación , Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Miocardio/metabolismo , Obesidad/etiología , Fosforilación , Distribución Aleatoria , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Sacarosa/administración & dosificación , Sacarosa/efectos adversos , Función Ventricular Izquierda/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...