Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.082
Filtrar
1.
Free Radic Biol Med ; 183: 75-88, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35318101

RESUMEN

Myocardial dysfunction is associated with an imbalance in mitochondrial fusion/fission dynamics in patients with diabetes. However, effective strategies to regulate mitochondrial dynamics in the diabetic heart are still lacking. Nicotinamide riboside (NR) supplementation ameliorated mitochondrial dysfunction and oxidative stress in both cardiovascular and aging-related diseases. This study investigated whether NR protects against diabetes-induced cardiac dysfunction by regulating mitochondrial fusion/fission and further explored the underlying mechanisms. Here, we showed an evident decrease in NAD+ (nicotinamide adenine dinucleotide) levels and mitochondrial fragmentation in the hearts of leptin receptor-deficient diabetic (db/db) mouse models. NR supplementation significantly increased NAD+ content in the diabetic hearts and promoted mitochondrial fusion by elevating Mfn2 level. Furthermore, NR-induced mitochondrial fusion suppressed mitochondrial H2O2 and O2•- production and reduced cardiomyocyte apoptosis in both db/db mice hearts and neonatal primary cardiomyocytes. Mechanistically, chromatin immunoprecipitation (ChIP) and luciferase reporter assay analyses revealed that PGC1α and PPARα interdependently regulated Mfn2 transcription by binding to its promoter region. NR treatment elevated NAD+ levels and activated SIRT1, resulting in the deacetylation of PGC1α and promoting the transcription of Mfn2. These findings suggested the promotion of mitochondrial fusion via oral supplementation of NR as a potential strategy for delaying cardiac complications in patients with diabetes.


Asunto(s)
Diabetes Mellitus , GTP Fosfohidrolasas , Mitocondrias Cardíacas , Dinámicas Mitocondriales , Animales , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Mitocondrias Cardíacas/fisiología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacología , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Compuestos de Piridinio , Sirtuina 1/genética , Sirtuina 1/metabolismo
2.
Int J Mol Sci ; 23(4)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35216368

RESUMEN

Analysis of the function, structure, and intracellular organization of mitochondria is important for elucidating energy metabolism and intracellular energy transfer. In addition, basic and clinically oriented studies that investigate organ/tissue/cell dysfunction in various human diseases, including myopathies, cardiac/brain ischemia-reperfusion injuries, neurodegenerative diseases, cancer, and aging, require precise estimation of mitochondrial function. It should be noted that the main metabolic and functional characteristics of mitochondria obtained in situ (in permeabilized cells and tissue samples) and in vitro (in isolated organelles) are quite different, thereby compromising interpretations of experimental and clinical data. These differences are explained by the existence of the mitochondrial network, which possesses multiple interactions between the cytoplasm and other subcellular organelles. Metabolic and functional crosstalk between mitochondria and extra-mitochondrial cellular environments plays a crucial role in the regulation of mitochondrial metabolism and physiology. Therefore, it is important to analyze mitochondria in vivo or in situ without their isolation from the natural cellular environment. This review summarizes previous studies and discusses existing approaches and methods for the analysis of mitochondrial function, structure, and intracellular organization in situ.


Asunto(s)
Mitocondrias Cardíacas/fisiología , Músculo Esquelético/fisiología , Miocitos Cardíacos/fisiología , Animales , Respiración de la Célula/fisiología , Metabolismo Energético/fisiología , Humanos
3.
Sci Rep ; 12(1): 978, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35046471

RESUMEN

Mitochondrial dysfunction is a feature of type I and type II diabetes, but there is a lack of consistency between reports and links to disease development. We aimed to investigate if mitochondrial structure-function remodelling occurs in the early stages of diabetes by employing a mouse model (GENA348) of Maturity Onset Diabetes in the Young, exhibiting hyperglycemia, but not hyperinsulinemia, with mild left ventricular dysfunction. Employing 3-D electron microscopy (SBF-SEM) we determined that compared to wild-type, WT, the GENA348 subsarcolemma mitochondria (SSM) are ~ 2-fold larger, consistent with up-regulation of fusion proteins Mfn1, Mfn2 and Opa1. Further, in comparison, GENA348 mitochondria are more irregular in shape, have more tubular projections with SSM projections being longer and wider. Mitochondrial density is also increased in the GENA348 myocardium consistent with up-regulation of PGC1-α and stalled mitophagy (down-regulation of PINK1, Parkin and Miro1). GENA348 mitochondria have more irregular cristae arrangements but cristae dimensions and density are similar to WT. GENA348 Complex activity (I, II, IV, V) activity is decreased but the OCR is increased, potentially linked to a shift towards fatty acid oxidation due to impaired glycolysis. These novel data reveal that dysregulated mitochondrial morphology, dynamics and function develop in the early stages of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/patología , Mitocondrias Cardíacas/ultraestructura , Dinámicas Mitocondriales , Miocardio/ultraestructura , Animales , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Ratones , Mitocondrias Cardíacas/fisiología
4.
J Pharmacol Sci ; 148(2): 238-247, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35063139

RESUMEN

Chronic magnesium (Mg) deficiency induces and exacerbates various cardiovascular diseases. We previously investigated the mechanisms underlying decline in cardiac function caused by chronic Mg deficiency and the effectiveness of Mg supplementation on this decline using the Langendorff-perfused isolated mouse heart model. Herein, we used the Langendorff-perfused isolated rat heart model to demonstrate the chronic Mg-deficient rats (Mg-deficient group) had lower the heart rate (HR) and left ventricular pressure (LVDP) than rats with normal Mg levels (normal group). Furthermore, decline in cardiac function due to hypoxia/reoxygenation injury was significantly greater in the Mg-deficient group than in the normal group. Experiments on mitochondrial permeability transition pore (mPTP) using isolated mitochondria revealed that mitochondrial membrane was fragile in the Mg-deficient group, implying that cardiac function decline through hypoxia/reoxygenation injury is associated with mitochondrial function. Mg supplementation for chronic Mg-deficient rats not only improved hypomagnesemia but also almost completely restored cardiac and mitochondrial functions. Therefore, proactive Mg supplementation in pathological conditions induced by Mg deficiency or for those at risk of developing hypomagnesemia may suppress the development and exacerbation of certain disease states.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Hipoxia/etiología , Deficiencia de Magnesio/complicaciones , Mitocondrias Cardíacas , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Animales , Presión Sanguínea , Enfermedades Cardiovasculares/prevención & control , Enfermedad Crónica , Suplementos Dietéticos , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Magnesio/administración & dosificación , Deficiencia de Magnesio/patología , Deficiencia de Magnesio/fisiopatología , Deficiencia de Magnesio/terapia , Masculino , Mitocondrias Cardíacas/fisiología , Membranas Mitocondriales/patología , Ratas Sprague-Dawley , Función Ventricular Izquierda
5.
Biomark Med ; 16(1): 11-21, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34856814

RESUMEN

Aim: We aimed to investigate the association of SIRT4 gene polymorphisms with the susceptibility and prognosis of dilated cardiomyopathy (DCM) in a Chinese population. Materials & methods: A total of 369 controls and 373 DCM patients were enrolled. Three tag single-nucleotide polymorphisms (rs2261612, rs2522138 and rs16950058) on SIRT4 were evaluated. Results: G carriers of rs2261612 were associated with the susceptibility of DCM in codominant, dominant and overdominant genetic models (all p < 0.01). Furthermore, the AG/GG and AG genotype of rs2261612 in dominant and overdominant models correlated with poor prognosis of DCM, independent of left ventricular ejection fraction and cardiac resynchronization therapy (p < 0.001). Conclusion:SIRT4 polymorphisms were associated with the susceptibility and prognosis of DCM in a Chinese population.


Asunto(s)
Cardiomiopatía Dilatada/genética , Etnicidad/genética , Proteínas Mitocondriales/genética , Polimorfismo de Nucleótido Simple , Sirtuinas/genética , Adulto , Cardiomiopatía Dilatada/fisiopatología , China , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias Cardíacas/fisiología , Función Ventricular Izquierda
6.
Pharmacol Res ; 175: 105986, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34800627

RESUMEN

During cardiac reperfusion after myocardial infarction, the heart is subjected to cascading cycles of ischaemia reperfusion injury (IRI). Patients presenting with this injury succumb to myocardial dysfunction resulting in myocardial cell death, which contributes to morbidity and mortality. New targeted therapies are required if the myocardium is to be protected from this injury and improve patient outcomes. Extensive research into the role of mitochondria during ischaemia and reperfusion has unveiled one of the most important sites contributing towards this injury; specifically, the opening of the mitochondrial permeability transition pore. The opening of this pore occurs during reperfusion and results in mitochondria swelling and dysfunction, promoting apoptotic cell death. Activation of mitochondrial ATP-sensitive potassium channels (mitoKATP) channels, uncoupling proteins, and inhibition of glycogen synthase kinase-3ß (GSK3ß) phosphorylation have been identified to delay mitochondrial permeability transition pore opening and reduce reactive oxygen species formation, thereby decreasing infarct size. Statins have recently been identified to provide a direct cardioprotective effect on these specific mitochondrial components, all of which reduce the severity of myocardial IRI, promoting the ability of statins to be a considerate preconditioning agent. This review will outline what has currently been shown in regard to statins cardioprotective effects on mitochondria during myocardial IRI.


Asunto(s)
Cardiotónicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Cardiotónicos/farmacología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Mitofagia/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Canales de Potasio/fisiología
7.
Mitochondrion ; 62: 159-175, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34902622

RESUMEN

Sigmar1 is a widely expressed molecular chaperone protein in mammalian cell systems. Accumulating research demonstrated the cardioprotective roles of pharmacologic Sigmar1 activation by ligands in preclinical rodent models of cardiac injury. Extensive biochemical and immuno-electron microscopic research demonstrated Sigmar1's sub-cellular localization largely depends on cell and organ types. Despite comprehensive studies, Sigmar1's direct molecular role in cardiomyocytes remains elusive. In the present study, we determined Sigmar1's subcellular localization, transmembrane topology, and function using complementary microscopy, biochemical, and functional assays in cardiomyocytes. Quantum dots in transmission electron microscopy showed Sigmar1 labeled quantum dots on the mitochondrial membranes, lysosomes, and sarcoplasmic reticulum-mitochondrial interface. Subcellular fractionation of heart cell lysates confirmed Sigmar1's localization in purified mitochondria fraction and lysosome fraction. Immunocytochemistry confirmed Sigmar1 colocalization with mitochondrial proteins in isolated adult mouse cardiomyocytes. Sigmar1's mitochondrial localization was further confirmed by Sigmar1 colocalization with Mito-Tracker in isolated mouse heart mitochondria. A series of biochemical experiments, including alkaline extraction and proteinase K treatment of purified heart mitochondria, demonstrated Sigmar1 as an integral mitochondrial membrane protein. Sigmar1's structural requirement for mitochondrial localization was determined by expressing FLAG-tagged Sigmar1 fragments in cells. Full-length Sigmar1 and Sigmar1's C terminal-deletion fragments were able to localize to the mitochondrial membrane, whereas N-terminal deletion fragment was unable to incorporate into the mitochondria. Finally, functional assays using extracellular flux analyzer and high-resolution respirometry showed Sigmar1 siRNA knockdown significantly altered mitochondrial respiration in cardiomyocytes. Overall, we found that Sigmar1 localizes to mitochondrial membranes and is indispensable for maintaining mitochondrial respiratory homeostasis in cardiomyocytes.


Asunto(s)
Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/metabolismo , Transporte de Proteínas/fisiología , Receptores sigma/metabolismo , Animales , Metabolismo Energético/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Ratones , ARN Interferente Pequeño , Ratas , Receptores sigma/genética , Receptor Sigma-1
8.
Sci Rep ; 11(1): 24129, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34916603

RESUMEN

Abuse of the potent psychostimulant cocaine is widely established to have cardiovascular consequences. The cardiotoxicity of cocaine is mainly associated with oxidative stress and mitochondrial dysfunction. Mitochondrial dynamics and biogenesis, as well as the mitochondrial unfolded protein response (UPRmt), guarantee cardiac mitochondrial homeostasis. Collectively, these mechanisms act to protect against stress, injury, and the detrimental effects of chemicals on mitochondria. In this study, we examined the effects of cocaine on cardiac mitochondrial dynamics, biogenesis, and UPRmt in vivo. Rats administered cocaine via the tail vein at a dose of 20 mg/kg/day for 7 days showed no structural changes in the myocardium, but electron microscopy revealed a significant increase in the number of cardiac mitochondria. Correspondingly, the expressions of the mitochondrial fission gene and mitochondrial biogenesis were increased after cocaine administration. Significant increase in the expression and nuclear translocation of activating transcription factor 5, the major active regulator of UPRmt, were observed after cocaine administration. Accordingly, our findings show that before any structural changes are observable in the myocardium, cocaine alters mitochondrial dynamics, elevates mitochondrial biogenesis, and induces the activation of UPRmt. These alterations might reflect cardiac mitochondrial compensation to protect against the cardiotoxicity of cocaine.


Asunto(s)
Cocaína/efectos adversos , Mitocondrias Cardíacas/efectos de los fármacos , Biogénesis de Organelos , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción Activadores/metabolismo , Animales , Cocaína/toxicidad , Homeostasis/efectos de los fármacos , Masculino , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/fisiología , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/genética , Ratas Sprague-Dawley , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética
9.
Sci Rep ; 11(1): 22106, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764359

RESUMEN

O-GlcNAcylation is a prevalent form of glycosylation that regulates proteins within the cytosol, nucleus, and mitochondria. The O-GlcNAc modification can affect protein cellular localization, function, and signaling interactions. The specific impact of O-GlcNAcylation on mitochondrial morphology and function has been elusive. In this manuscript, the role of O-GlcNAcylation on mitochondrial fission, oxidative phosphorylation (Oxphos), and the activity of electron transport chain (ETC) complexes were evaluated. In a cellular environment with hyper O-GlcNAcylation due to the deletion of O-GlcNAcase (OGA), mitochondria showed a dramatic reduction in size and a corresponding increase in number and total mitochondrial mass. Because of the increased mitochondrial content, OGA knockout cells exhibited comparable coupled mitochondrial Oxphos and ATP levels when compared to WT cells. However, we observed reduced protein levels for complex I and II when comparing normalized mitochondrial content and reduced linked activity for complexes I and III when examining individual ETC complex activities. In assessing mitochondrial fission, we observed increased amounts of O-GlcNAcylated dynamin-related protein 1 (Drp1) in cells genetically null for OGA and in glioblastoma cells. Individual regions of Drp1 were evaluated for O-GlcNAc modifications, and we found that this post-translational modification (PTM) was not limited to the previously characterized residues in the variable domain (VD). Additional modification sites are predicted in the GTPase domain, which may influence enzyme activity. Collectively, these results highlight the impact of O-GlcNAcylation on mitochondrial dynamics and ETC function and mimic the changes that may occur during glucose toxicity from hyperglycemia.


Asunto(s)
Acilación/genética , Acilación/fisiología , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/fisiología , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Dinaminas/genética , Dinaminas/metabolismo , Glucosa/genética , Glucosa/metabolismo , Glicosilación , Células HCT116 , Humanos , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Dinámicas Mitocondriales/genética , Dinámicas Mitocondriales/fisiología , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , N-Acetilglucosaminiltransferasas/genética , Fosforilación Oxidativa , Procesamiento Proteico-Postraduccional/genética , Transducción de Señal/genética
10.
Biol Sex Differ ; 12(1): 58, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34727994

RESUMEN

Women with preeclampsia (PE) have a greater risk of developing hypertension, cardiovascular disease (CVD), and renal disease later in life. Angiotensin II type I receptor agonistic autoantibodies (AT1-AAs) are elevated in women with PE during pregnancy and up to 2-year postpartum (PP), and in the reduced uterine perfusion pressure (RUPP) rat model of PE. Blockade of AT1-AA with a specific 7 amino acid peptide binding sequence ('n7AAc') improves pathophysiology observed in RUPP rats; however, the long-term effects of AT1-AA inhibition in PP is unknown. Pregnant Sprague Dawley rats were divided into three groups: normal pregnant (NP) (n = 16), RUPP (n = 15), and RUPP + 'n7AAc' (n = 16). Gestational day 14, RUPP surgery was performed and 'n7AAc' (144 µg/day) administered via osmotic minipump. At 10-week PP, mean arterial pressure (MAP), renal glomerular filtration rate (GFR) and cardiac functions, and cardiac mitochondria function were assessed. MAP was elevated PP in RUPP vs. NP (126 ± 4 vs. 116 ± 3 mmHg, p < 0.05), but was normalized in in RUPP + 'n7AAc' (109 ± 3 mmHg) vs. RUPP (p < 0.05). PP heart size was reduced by RUPP + 'n7AAc' vs. RUPP rats (p < 0.05). Complex IV protein abundance and enzymatic activity, along with glutamate/malate-driven respiration (complexes I, III, and IV), were reduced in the heart of RUPP vs. NP rats which was prevented with 'n7AAc'. AT1-AA inhibition during pregnancy not only improves blood pressure and pathophysiology of PE in rats during pregnancy, but also long-term changes in blood pressure, cardiac hypertrophy, and cardiac mitochondrial function PP.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Autoanticuerpos/farmacología , Hipertensión , Mitocondrias Cardíacas/fisiología , Preeclampsia , Animales , Femenino , Hipertensión/tratamiento farmacológico , Placenta , Periodo Posparto , Preeclampsia/tratamiento farmacológico , Embarazo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1
11.
J Biol Chem ; 297(6): 101358, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34756888

RESUMEN

Preserving optimal mitochondrial function is critical in the heart, which is the most ATP-avid organ in the body. Recently, we showed that global deficiency of the nuclear receptor RORα in the "staggerer" mouse exacerbates angiotensin II-induced cardiac hypertrophy and compromises cardiomyocyte mitochondrial function. However, the mechanisms underlying these observations have not been defined previously. Here, we used pharmacological and genetic gain- and loss-of-function tools to demonstrate that RORα regulates cardiomyocyte mitophagy to preserve mitochondrial abundance and function. We found that cardiomyocyte mitochondria in staggerer mice with lack of functional RORα were less numerous and exhibited fewer mitophagy events than those in WT controls. The hearts of our novel cardiomyocyte-specific RORα KO mouse line demonstrated impaired contractile function, enhanced oxidative stress, increased apoptosis, and reduced autophagic flux relative to Cre(-) littermates. We found that cardiomyocyte mitochondria in "staggerer" mice with lack of functional RORα were upregulated by hypoxia, a classical inducer of mitophagy. The loss of RORα blunted mitophagy and broadly compromised mitochondrial function in normoxic and hypoxic conditions in vivo and in vitro. We also show that RORα is a direct transcriptional regulator of the mitophagy mediator caveolin-3 in cardiomyocytes and that enhanced expression of RORα increases caveolin-3 abundance and enhances mitophagy. Finally, knockdown of RORα impairs cardiomyocyte mitophagy, compromises mitochondrial function, and induces apoptosis, but these defects could be rescued by caveolin-3 overexpression. Collectively, these findings reveal a novel role for RORα in regulating mitophagy through caveolin-3 and expand our currently limited understanding of the mechanisms underlying RORα-mediated cardioprotection.


Asunto(s)
Caveolina 3/fisiología , Mitocondrias Cardíacas/fisiología , Mitofagia/fisiología , Miocitos Cardíacos/fisiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Animales , Ratones , Mitocondrias Cardíacas/metabolismo
12.
Naunyn Schmiedebergs Arch Pharmacol ; 394(10): 2103-2115, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34338837

RESUMEN

PURPOSE: Cardioprotective effect of carbon monoxide, a gasotransmitter against myocardial ischemia-reperfusion injury (I/R), is well established in preclinical studies with male rats. However, its ischemic tolerance in post-menopausal animals has not been examined due to functional perturbations at the cellular level. METHODS: The protective role of carbon monoxide releasing molecule-2 (CORM-2) on myocardial I/R was studied in female Wistar rats using the Langendorff apparatus. The animals were randomly divided into normal and ovariectomized (Ovx) female rats and were maintained 2 months post-surgery. Each group was further divided into 4 subgroups (n = 6/subgroup): normal, I/R, CORM-2-control (20 µmol/L), and CORM-2-I/R. The cardiac injury was estimated via myocardial infarct size, lactate dehydrogenase, and creatine kinase levels in coronary effluent and cardiac hemodynamic indices. Mitochondrial functional activity was assessed by measuring mitochondrial electron transport chain enzyme activities, swelling behavior, mitochondrial membrane potential, and oxidative stress. RESULTS: Hemodynamic indices were significantly lower in ovariectomized rat hearts than in normal rat hearts. Sixty minutes of reperfusion of ischemic heart exhibited deteriorated cardiac physiological recovery in both ovariectomized and normal groups, where prominent decline was observed in ovariectomized rat. However, preconditioning the isolated heart with CORM-2 improved hemodynamics parameters significantly in both ovariectomized and normal rat hearts challenged with I/R, but with a limited degree of protection in ovariectomized rat hearts. The protective effect of CORM-2 was further confirmed via a reduction in cardiac injury, preservation of mitochondrial enzymes, and reduction in oxidative stress in all groups. CONCLUSION: CORM-2 administration significantly attenuated myocardial I/R injury in ovariectomized rat hearts by attenuating I/R-associated mitochondrial perturbations and reducing oxidative stress.


Asunto(s)
Cardiotónicos/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Compuestos Organometálicos/uso terapéutico , Animales , Monóxido de Carbono/metabolismo , Cardiotónicos/farmacología , Femenino , Hemodinámica , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Compuestos Organometálicos/farmacología , Ovariectomía , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar
13.
Sci Rep ; 11(1): 16542, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34400686

RESUMEN

Left ventricular remodeling due to pressure overload is associated with poor prognosis. Sacubitril/valsartan is the first-in-class Angiotensin Receptor Neprilysin Inhibitor and has been demonstrated to have superior beneficial effects in the settings of heart failure. The aim of this study was to determine whether sacubitril/valsartan has cardioprotective effect in the early intervention of pressure overloaded hearts and whether it is superior to valsartan alone. We induced persistent left ventricular pressure overload in rats by ascending aortic constriction surgery and orally administrated sacubitril/valsartan, valsartan, or vehicle one week post operation for 10 weeks. We also determined the effects of sacubitril/valsartan over valsartan on adult ventricular myocytes and fibroblasts that were isolated from healthy rats and treated in culture. We found that early intervention with sacubitril/valsartan is superior to valsartan in reducing pressure overload-induced ventricular fibrosis and in reducing angiotensin II-induced adult ventricular fibroblast activation. While neither sacubitril/valsartan nor valsartan changes cardiac hypertrophy development, early intervention with sacubitril/valsartan protects ventricular myocytes from mitochondrial dysfunction and is superior to valsartan in reducing mitochondrial oxidative stress in response to persistent left ventricular pressure overload. In conclusion, our findings demonstrate that sacubitril/valsartan has a superior cardioprotective effect over valsartan in the early intervention of pressure overloaded hearts, which is independent of the reduction of left ventricular afterload. Our study provides evidence in support of potential benefits of the use of sacubitril/valsartan in patients with resistant hypertension or in patients with severe aortic stenosis.


Asunto(s)
Aminobutiratos/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Cardiotónicos/uso terapéutico , Intervención Médica Temprana , Valsartán/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Aminobutiratos/farmacología , Antagonistas de Receptores de Angiotensina/farmacología , Animales , Aorta , Compuestos de Bifenilo/farmacología , Cardiotónicos/farmacología , Constricción , Modelos Animales de Enfermedad , Combinación de Medicamentos , Fibroblastos/efectos de los fármacos , Fibrosis , Ventrículos Cardíacos/patología , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Superóxidos/metabolismo , Valsartán/farmacología
14.
Am J Physiol Cell Physiol ; 321(3): C489-C503, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34191626

RESUMEN

Mitochondrial transplantation is emerging as a novel cellular biotherapy to alleviate mitochondrial damage and dysfunction. Mitochondria play a crucial role in establishing cellular homeostasis and providing cell with the energy necessary to accomplish its function. Owing to its endosymbiotic origin, mitochondria share many features with their bacterial ancestors. Unlike the nuclear DNA, which is packaged into nucleosomes and protected from adverse environmental effects, mitochondrial DNA are more prone to harsh environmental effects, in particular that of the reactive oxygen species. Mitochondrial damage and dysfunction are implicated in many diseases ranging from metabolic diseases to cardiovascular and neurodegenerative diseases, among others. While it was once thought that transplantation of mitochondria would not be possible due to their semiautonomous nature and reliance on the nucleus, recent advances have shown that it is possible to transplant viable functional intact mitochondria from autologous, allogenic, and xenogeneic sources into different cell types. Moreover, current research suggests that the transplantation could positively modulate bioenergetics and improve disease outcome. Mitochondrial transplantation techniques and consequences of transplantation in cardiomyocytes are the theme of this review. We outline the different mitochondrial isolation and transfer techniques. Finally, we detail the consequences of mitochondrial transplantation in the cardiovascular system, more specifically in the context of cardiomyopathies and ischemia.


Asunto(s)
Cardiomiopatías/terapia , Diabetes Mellitus Experimental/terapia , Mitocondrias Cardíacas/trasplante , Infarto del Miocardio/terapia , Daño por Reperfusión Miocárdica/terapia , Miocitos Cardíacos/metabolismo , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Fraccionamiento Celular/métodos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Humanos , Inyecciones Intralesiones , Mitocondrias Cardíacas/fisiología , Mitocondrias Cardíacas/ultraestructura , Dinámicas Mitocondriales/fisiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Fosforilación Oxidativa , Conejos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Porcinos
15.
Curr Opin Genet Dev ; 70: 54-60, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34130066

RESUMEN

The adult mammalian heart cannot regenerate after myocardial injury because most cardiomyocytes lack the ability to proliferate. In contrast, cardiomyocytes of vertebrates such as zebrafish and urodele amphibians, but also those of fetal and early neonatal mammals, maintain the ability to proliferate and therefore support regeneration of injured tissue and recovery of cardiac function. Whether evolutionarily conserved regulatory mechanisms of cardiomyocyte proliferation exist and, if so, whether they are modifiable to allow cardiac regeneration in adult mammals are questions of great scientific and medical interest. Environmental hypoxia, hypoxia-induced cellular signaling, and mitochondrial metabolism have recently emerged as key regulators of the cardiomyocyte cell cycle and cardiac regeneration in vertebrates. In this review, we address the cardiac regenerative capacity of several model animals and discuss potential strategies related to hypoxia and mitochondrial metabolism for induction of therapeutic heart regeneration.


Asunto(s)
Hipoxia de la Célula , Metabolismo Energético , Homeostasis , Mitocondrias Cardíacas/fisiología , Dinámicas Mitocondriales , Infarto del Miocardio/metabolismo , Regeneración , Animales , Humanos , Infarto del Miocardio/fisiopatología
16.
Shock ; 56(5): 857-864, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33978607

RESUMEN

ABSTRACT: Mitochondria is often considered as the common nexus of cardiac and cerebral dysfunction after cardiac arrest. Here, our goal was to determine whether the time course of cardiac and cerebral mitochondrial dysfunction is similar after shockable versus non-shockable cardiac arrest in rabbits. Anesthetized rabbits were submitted to 10 min of no-flow by ventricular fibrillation (VF group) or asphyxia (non-shockable group). They were euthanized at the end of the no-flow period or 30 min, 120 min, or 24 h after resuscitation for in vitro evaluation of oxygen consumption and calcium retention capacity. In the brain (cortex and hippocampus), moderate mitochondrial dysfunction was evidenced at the end of the no-flow period after both causes of cardiac arrest versus baseline. It partly recovered at 30 and 120 min after cardiac arrest, with lower calcium retention capacity and higher substrate-dependant oxygen consumption after VF versus non-shockable cardiac arrest. However, after 24 h of follow-up, mitochondrial dysfunction dramatically increased after both VF and non-shockable cardiac arrest, despite greater neurological dysfunction after the latter one. In the heart, mitochondrial dysfunction was also maximal after 24 h following resuscitation, with no significant difference among the causes of the cardiac arrest. During the earlier timing of evaluation, calcium retention capacity and ADP-dependant oxygen consumption were lower and higher, respectively, after non-shockable cardiac arrest versus VF. In conclusion, the kinetics of cardiac and cerebral mitochondrial dysfunction suggests that mitochondrial function does not play a major role in the early phase of the post-resuscitation process but is only involved in the longer pathophysiological events.


Asunto(s)
Encefalopatías/fisiopatología , Encéfalo/ultraestructura , Paro Cardíaco/fisiopatología , Mitocondrias/fisiología , Fibrilación Ventricular/fisiopatología , Animales , Masculino , Mitocondrias Cardíacas/fisiología , Conejos
17.
Circ Res ; 128(10): 1594-1612, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33983828

RESUMEN

This review provides a comprehensive overview of the past 25+ years of research into the development of left ventricular assist device (LVAD) to improve clinical outcomes in patients with severe end-stage heart failure and basic insights gained into the biology of heart failure gleaned from studies of hearts and myocardium of patients undergoing LVAD support. Clinical aspects of contemporary LVAD therapy, including evolving device technology, overall mortality, and complications, are reviewed. We explain the hemodynamic effects of LVAD support and how these lead to ventricular unloading. This includes a detailed review of the structural, cellular, and molecular aspects of LVAD-associated reverse remodeling. Synergisms between LVAD support and medical therapies for heart failure related to reverse remodeling, remission, and recovery are discussed within the context of both clinical outcomes and fundamental effects on myocardial biology. The incidence, clinical implications and factors most likely to be associated with improved ventricular function and remission of the heart failure are reviewed. Finally, we discuss recognized impediments to achieving myocardial recovery in the vast majority of LVAD-supported hearts and their implications for future research aimed at improving the overall rates of recovery.


Asunto(s)
Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Función Ventricular Izquierda/fisiología , Remodelación Ventricular , Animales , Calcio/metabolismo , Muerte Celular/fisiología , Citocinas/metabolismo , Citoesqueleto/fisiología , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Matriz Extracelular/fisiología , Corazón Auxiliar/efectos adversos , Corazón Auxiliar/tendencias , Hemodinámica , Humanos , Macrófagos/citología , Mitocondrias Cardíacas/fisiología , Contracción Miocárdica , Miocitos Cardíacos/fisiología , Transcriptoma
18.
FASEB J ; 35(4): e21278, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33769614

RESUMEN

Mitochondria share attributes of vesicular transport with their bacterial ancestors given their ability to form mitochondrial-derived vesicles (MDVs). MDVs are involved in mitochondrial quality control and their formation is enhanced with stress and may, therefore, play a potential role in mitochondrial-cellular communication. However, MDV proteomic cargo has remained mostly undefined. In this study, we strategically used an in vitro MDV budding/reconstitution assay on cardiac mitochondria, followed by graded oxidative stress, to identify and characterize the MDV proteome. Our results confirmed previously identified cardiac MDV markers, while also revealing a complete map of the MDV proteome, paving the way to a better understanding of the role of MDVs. The oxidative stress vulnerability of proteins directed the cargo loading of MDVs, which was enhanced by antimycin A (Ant-A). Among OXPHOS complexes, complexes III and V were found to be Ant-A-sensitive. Proteins from metabolic pathways such as the TCA cycle and fatty acid metabolism, along with Fe-S cluster, antioxidant response proteins, and autophagy were also found to be Ant-A sensitive. Intriguingly, proteins containing hyper-reactive cysteine residues, metabolic redox switches, including professional redox enzymes and those that mediate iron metabolism, were found to be components of MDV cargo with Ant-A sensitivity. Last, we revealed a possible contribution of MDVs to the formation of extracellular vesicles, which may indicate mitochondrial stress. In conclusion, our study provides an MDV proteomics signature that delineates MDV cargo selectivity and hints at the potential for MDVs and their novel protein cargo to serve as vital biomarkers during mitochondrial stress and related pathologies.


Asunto(s)
Mitocondrias Cardíacas/fisiología , Estrés Oxidativo , Vesículas Transportadoras/fisiología , Animales , Línea Celular , Regulación de la Expresión Génica , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mioblastos , Proteómica , Ratas
19.
Mol Genet Metab ; 132(4): 244-253, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33674196

RESUMEN

Osteogenesis imperfecta (OI) is a heritable connective tissue disorder with patients exhibiting bone fragility and muscle weakness. The synergistic biochemical and biomechanical relationship between bone and muscle is a critical potential therapeutic target, such that muscle weakness should not be ignored. Previous studies demonstrated mitochondrial dysfunction in the skeletal muscle of oim/oim mice, which model a severe human type III OI. Here, we further characterize this mitochondrial dysfunction and evaluate several parameters of whole body and skeletal muscle metabolism. We demonstrate reduced mitochondrial respiration in female gastrocnemius muscle, but not in liver or heart mitochondria, suggesting that mitochondrial dysfunction is not global in the oim/oim mouse. Myosin heavy chain fiber type distributions were altered in the oim/oim soleus muscle with a decrease (-33 to 50%) in type I myofibers and an increase (+31%) in type IIa myofibers relative to their wildtype (WT) littermates. Additionally, altered body composition and increased energy expenditure were observed oim/oim mice relative to WT littermates. These results suggest that skeletal muscle mitochondrial dysfunction is linked to whole body metabolic alterations and to skeletal muscle weakness in the oim/oim mouse.


Asunto(s)
Metabolismo Energético/genética , Mitocondrias Cardíacas/genética , Músculo Esquelético/metabolismo , Osteogénesis Imperfecta/genética , Animales , Modelos Animales de Enfermedad , Fémur/metabolismo , Fémur/patología , Humanos , Ratones , Mitocondrias Cardíacas/fisiología , Músculo Esquelético/patología , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología , Índice de Severidad de la Enfermedad
20.
Biomed Pharmacother ; 138: 111316, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33684689

RESUMEN

BACKGROUND: Cardiovascular diseases are the leading cause of death globally, and they are causing enormous socio-economic burden to the developed and developing countries. Allyl Methyl Sulfide (AMS) is a novel cardioprotective metabolite identified in the serum of rats after raw garlic administration. The present study explored the cardioprotective effect of AMS on thoracic aortic constriction (TAC)-induced cardiac hypertrophy and heart failure model in rats. METHODS: Thoracic aortic constriction (TAC) by titanium ligating clips resulted in the development of pressure overload-induced cardiac hypertrophy and heart failure model. Four weeks prior to TAC and for 8 weeks after TAC, Sprague Dawley (SD) rats were administered with AMS (25 and 50 mg/kg/day) or Enalapril (10 mg/kg/day). RESULTS: We have observed AMS (25 and 50 mg/kg/day) intervention significantly improved structural and functional parameters of the heart. mRNA expression of fetal genes i.e., atrial natriuretic peptide (ANP), alpha skeletal actin (α-SA) and beta myosin heavy chain (ß-MHC) were reduced in AMS treated TAC hearts along with decrease in perivascular and interstitial fibrosis. AMS attenuated lipid peroxidation and improved protein expression of endogenous antioxidant enzymes i.e., catalase and manganese superoxide dismutase (MnSOD) along with electron transport chain (ETC) complex activity. AMS increased mitochondrial fusion proteins i.e., mitofusin 1 (MFN1), mitofusin 2 (MFN2) and optic atrophy protein (OPA1), and reduced fission protein i.e., dynamin-related protein 1 (DRP1). Preliminary study suggests that AMS intervention upregulated genes involved in mitochondrial bioenergetics in normal rats. Further, in-vitro studies suggest that AMS reduced mitochondrial reactive oxygen species (ROS), preserved mitochondrial membrane potential and oxygen consumption rate (OCR) in isoproterenol-treated cardiomyoblast. CONCLUSION: This study demonstrated that AMS protected cardiac remodelling, LV dysfunction and fibrosis in pressure overload-induced cardiac hypertrophy and heart failure model by improving endogenous antioxidants and mitochondrial function.


Asunto(s)
Compuestos Alílicos/uso terapéutico , Cardiotónicos/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Mitocondrias Cardíacas/efectos de los fármacos , Sulfuros/uso terapéutico , Compuestos Alílicos/farmacología , Animales , Aorta Torácica/diagnóstico por imagen , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiopatología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/fisiopatología , Cardiotónicos/farmacología , Línea Celular , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/fisiopatología , Masculino , Mitocondrias Cardíacas/fisiología , Ratas , Ratas Sprague-Dawley , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/fisiología , Sulfuros/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA