Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.853
Filtrar
1.
BMC Cardiovasc Disord ; 24(1): 415, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39123142

RESUMEN

BACKGROUND: Ischemia reperfusion (IR) causes impaired myocardial function, and autophagy activation ameliorates myocardial IR injury. Isoliquiritigenin (ISO) has been found to protect myocardial tissues via AMPK, with exerting anti-tumor property through autophagy activation. This study aims to investigate ISO capacity to attenuate myocardial IR through autophagy activation mediated by AMPK/mTOR/ULK1 signaling. METHODS: ISO effects were explored by SD rats and H9c2 cells. IR rats and IR-induced H9c2 cell models were established by ligating left anterior descending (LAD) coronary artery and hypoxia/re-oxygenation, respectively, followed by low, medium and high dosages of ISO intervention (Rats: 10, 20, and 40 mg/kg; H9c2 cells: 1, 10, and 100 µmol/L). Myocardial tissue injury in rats was assessed by myocardial function-related index, HE staining, Masson trichrome staining, TTC staining, and ELISA. Autophagy of H9c2 cells was detected by transmission electron microscopy (TEM) and immunofluorescence. Autophagy-related and AMPK/mTOR/ULK1 pathway-related protein expressions were detected with western blot. RESULTS: ISO treatment caused myocardial function improvement, and inhibition of myocardial inflammatory infiltration, fibrosis, infarct area, oxidative stress, CK-MB, cTnI, and cTnT expression in IR rats. In IR-modeled H9c2 cells, ISO treatment lowered apoptosis rate and activated autophagy and LC3 fluorescence expression. In vivo and in vitro, ISO intervention exhibited enhanced Beclin1, LC3II/LC3I, and p-AMPK/AMPK levels, whereas inhibited P62, p-mTOR/mTOR and p-ULK1(S757)/ULK1 protein expression, activating autophagy and protecting myocardial tissues from IR injury. CONCLUSION: ISO treatment may induce autophagy by regulating AMPK/mTOR/ULK1 signaling, thereby improving myocardial IR injury, as a potential candidate for treatment of myocardial IR injury.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Homólogo de la Proteína 1 Relacionada con la Autofagia , Autofagia , Chalconas , Modelos Animales de Enfermedad , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Ratas Sprague-Dawley , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/prevención & control , Autofagia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Chalconas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Línea Celular , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Masculino , Ratas , Función Ventricular Izquierda/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/enzimología , Apoptosis/efectos de los fármacos , Fibrosis
2.
Cardiovasc Toxicol ; 24(8): 766-775, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38850470

RESUMEN

Cognitive impairment is a commonly observed complication following myocardial infarction; however, the underlying mechanisms are still not well understood. The most recent research suggests that extracellular signal-regulated kinase (ERK) plays a critical role in the development and occurrence of cognitive dysfunction-related diseases. This study aims to explore whether the ERK inhibitor U0126 targets the ERK/Signal Transducer and Activator of Transcription 1 (STAT1) pathway to ameliorate cognitive impairment after myocardial infarction. To establish a mouse model of myocardial infarction, we utilized various techniques including Echocardiography, Hematoxylin-eosin (HE) staining, Elisa, Open field test, Elevated plus maze test, and Western blot analysis to assess mouse cardiac function, cognitive function, and signal transduction pathways. For further investigation into the mechanisms of cognitive function and signal transduction, we administered the ERK inhibitor U0126 via intraperitoneal injection. Reduced total distance and activity range were observed in mice subjected to myocardial infarction during the open field test, along with decreased exploration of the open arms in the elevated plus maze test. However, U0126 treatment exhibited a significant improvement in cognitive decline, indicating a protective effect through the inhibition of the ERK/STAT1 signaling pathway. Hence, this study highlights the involvement of the ERK/STAT1 pathway in regulating cognitive dysfunction following myocardial infarction and establishes U0126 as a promising therapeutic target.


Asunto(s)
Conducta Animal , Butadienos , Cognición , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular , Ratones Endogámicos C57BL , Infarto del Miocardio , Nitrilos , Factor de Transcripción STAT1 , Animales , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/patología , Infarto del Miocardio/tratamiento farmacológico , Cognición/efectos de los fármacos , Nitrilos/farmacología , Masculino , Butadienos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Conducta Animal/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/enzimología , Disfunción Cognitiva/prevención & control , Transducción de Señal/efectos de los fármacos , Prueba de Laberinto Elevado , Prueba de Campo Abierto/efectos de los fármacos , Ratones
3.
J Cardiovasc Pharmacol ; 83(5): 446-456, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38416872

RESUMEN

ABSTRACT: Myocardial fibrosis, a common complication of myocardial infarction (MI), is characterized by excessive collagen deposition and can result in impaired cardiac function. The specific role of CD137 in the development of post-MI myocardial fibrosis remains unclear. Thus, this study aimed to elucidate the effects of CD137 signaling using CD137 knockout mice and in vitro experiments. CD137 expression levels progressively increased in the heart after MI, particularly in myofibroblast, which play a key role in fibrosis. Remarkably, CD137 knockout mice exhibited improved cardiac function and reduced fibrosis compared with wild-type mice at day 28 post-MI. The use of Masson's trichrome and picrosirius red staining demonstrated a reduction in the infarct area and collagen volume fraction in CD137 knockout mice. Furthermore, the expression of alpha-smooth muscle actin and collagen I, key markers of fibrosis, was decreased in heart tissues lacking CD137. In vitro experiments supported these findings because CD137 depletion attenuated cardiac fibroblast differentiation, and migration, and collagen I synthesis. In addition, the administration of CD137L recombinant protein further promoted alpha-smooth muscle actin expression and collagen I synthesis, suggesting a profibrotic effect. Notably, the application of an inhibitor targeting the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway attenuated the profibrotic effects of CD137L. To conclude, this study provides evidence that CD137 plays a significant role in promoting myocardial fibrosis after MI. Inhibition of CD137 signaling pathways may hold therapeutic potential for mitigating pathological cardiac remodeling and improving post-MI cardiac function.


Asunto(s)
Modelos Animales de Enfermedad , Fibrosis , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Remodelación Ventricular , Animales , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Masculino , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Miofibroblastos/metabolismo , Miofibroblastos/patología , Miofibroblastos/enzimología , Sistema de Señalización de MAP Quinasas , Miocardio/patología , Miocardio/metabolismo , Miocardio/enzimología , Ligando 4-1BB/metabolismo , Ligando 4-1BB/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Actinas/metabolismo , Células Cultivadas , Transducción de Señal , Movimiento Celular , Ratones , Función Ventricular Izquierda , Diferenciación Celular , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/efectos de los fármacos
4.
Biochem Pharmacol ; 197: 114950, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35143754

RESUMEN

We previously reported that 2,5-dimethylcelecoxib (DM-C), a derivative of celecoxib, lacks cyclooxygenase-2 inhibitory effects and suppresses cardiac remodeling by activating glycogen synthase kinase-3 (GSK-3). However, it remains unclear whether DM-C attenuates fibroblast-to-myofibroblast transformation (FMT), which plays a key role in cardiac fibrosis. Therefore, we evaluated the effect of DM-C on FMT using a cryoinjury-induced myocardial infarction (CMI) mouse model. We found that DM-C attenuated the deterioration of left ventricular ejection fraction after CMI by decreasing cardiac fibrosis. Analysis of the expression level of α-smooth muscle actin (α-SMA), a marker for myofibroblasts, indicated that DM-C decreased FMT at the cardiac injury site. To investigate the mechanism by which DM-C attenuated FMT, fibroblasts obtained from the heart were stimulated with TGF-ß to induce FMT, and the effect of DM-C was analyzed. DM-C suppressed the expression of α-SMA and the phosphorylation levels of Smad 2/3 and GSK-3, indicating that DM-C suppressed α-SMA expression by inhibiting the transforming growth factor (TGF)-ß signaling pathway via activation of GSK-3. DM-C decreased the expression of collagen, connective tissue growth factor (CTGF) and Snail, which are also known to accelerate cardiac fibrosis. These results suggested that DM-C attenuated cardiac fibrosis by suppressing FMT at the injured site after CMI by inhibiting the TGF-ß signaling pathway via activation of GSK-3. Thus, DM-C has potential against cardiac disease as a novel anti-fibrotic agent.


Asunto(s)
Fibroblastos/efectos de los fármacos , Congelación/efectos adversos , Infarto del Miocardio/tratamiento farmacológico , Miofibroblastos/efectos de los fármacos , Pirazoles/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico , Animales , Células Cultivadas , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Glucógeno Sintasa Quinasa 3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/enzimología , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Miofibroblastos/enzimología , Miofibroblastos/patología , Nitrógeno/toxicidad , Pirazoles/farmacología , Ratas , Ratas Endogámicas Lew , Transducción de Señal/fisiología , Sulfonamidas/farmacología
5.
Am J Physiol Cell Physiol ; 322(2): C296-C310, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35044856

RESUMEN

Aging chronically increases endoplasmic reticulum (ER) stress that contributes to mitochondrial dysfunction. Activation of calpain 1 (CPN1) impairs mitochondrial function during acute ER stress. We proposed that aging-induced ER stress led to mitochondrial dysfunction by activating CPN1. We posit that attenuation of the ER stress or direct inhibition of CPN1 in aged hearts can decrease cardiac injury during ischemia-reperfusion by improving mitochondrial function. Male young (3 mo) and aged mice (24 mo) were used in the present study, and 4-phenylbutyrate (4-PBA) was used to decrease the ER stress in aged mice. Subsarcolemmal (SSM) and interfibrillar mitochondria (IFM) were isolated. Chronic 4-PBA treatment for 2 wk decreased CPN1 activation as shown by the decreased cleavage of spectrin in cytosol and apoptosis inducing factor (AIF) and the α1 subunit of pyruvate dehydrogenase (PDH) in mitochondria. Treatment improved oxidative phosphorylation in 24-mo-old SSM and IFM at baseline compared with vehicle. When 4-PBA-treated 24-mo-old hearts were subjected to ischemia-reperfusion, infarct size was decreased. These results support that attenuation of the ER stress decreased cardiac injury in aged hearts by improving mitochondrial function before ischemia. To challenge the role of CPN1 as an effector of the ER stress, aged mice were treated with MDL-28170 (MDL, an inhibitor of calpain 1). MDL treatment improved mitochondrial function in aged SSM and IFM. MDL-treated 24-mo-old hearts sustained less cardiac injury following ischemia-reperfusion. These results support that age-induced ER stress augments cardiac injury during ischemia-reperfusion by impairing mitochondrial function through activation of CPN1.


Asunto(s)
Calpaína/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Factores de Edad , Animales , Calpaína/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Preparación de Corazón Aislado , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/patología , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Fosforilación Oxidativa/efectos de los fármacos , Fenilbutiratos/farmacología
6.
Biochem Pharmacol ; 195: 114866, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34863976

RESUMEN

Cardiovascular and heart diseases are leading causes of morbidity and mortality. Coronary artery endothelial and vascular dysfunction, inflammation, and mitochondrial dysfunction contribute to progression of heart diseases such as arrhythmias, congestive heart failure, and heart attacks. Classes of fatty acid epoxylipids and their enzymatic regulation by soluble epoxide hydrolase (sEH) have been implicated in coronary artery dysfunction, inflammation, and mitochondrial dysfunction in heart diseases. Likewise, genetic and pharmacological manipulations of epoxylipids have been demonstrated to have therapeutic benefits for heart diseases. Increasing epoxylipids reduce cardiac hypertrophy and fibrosis and improve cardiac function. Beneficial actions for epoxylipids have been demonstrated in cardiac ischemia reperfusion injury, electrical conductance abnormalities and arrhythmias, and ventricular tachycardia. This review discusses past and recent findings on the contribution of epoxylipids in heart diseases and the potential for their manipulation to treat heart attacks, arrhythmias, ventricular tachycardia, and heart failure.


Asunto(s)
Epóxido Hidrolasas/metabolismo , Compuestos Epoxi/metabolismo , Ácidos Grasos/metabolismo , Cardiopatías/metabolismo , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/enzimología , Arritmias Cardíacas/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Epóxido Hidrolasas/antagonistas & inhibidores , Compuestos Epoxi/química , Cardiopatías/tratamiento farmacológico , Cardiopatías/enzimología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/metabolismo , Humanos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/enzimología , Infarto del Miocardio/metabolismo , Solubilidad , Taquicardia Ventricular/tratamiento farmacológico , Taquicardia Ventricular/enzimología , Taquicardia Ventricular/metabolismo
7.
Cardiovasc Res ; 118(1): 169-183, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33560342

RESUMEN

AIMS: Myocardial infarction (MI) is the most common cause of heart failure (HF) worldwide. G protein-coupled receptor kinase 5 (GRK5) is upregulated in failing human myocardium and promotes maladaptive cardiac hypertrophy in animal models. However, the role of GRK5 in ischemic heart disease is still unknown. In this study, we evaluated whether myocardial GRK5 plays a critical role post-MI in mice and included the examination of specific cardiac immune and inflammatory responses. METHODS AND RESULTS: Cardiomyocyte-specific GRK5 overexpressing transgenic mice (TgGRK5) and non-transgenic littermate control (NLC) mice as well as cardiomyocyte-specific GRK5 knockout mice (GRK5cKO) and wild type (WT) were subjected to MI and, functional as well as structural changes together with outcomes were studied. TgGRK5 post-MI mice showed decreased cardiac function, augmented left ventricular dimension and decreased survival rate compared to NLC post-MI mice. Cardiac hypertrophy and fibrosis as well as fetal gene expression were increased post-MI in TgGRK5 compared to NLC mice. In TgGRK5 mice, GRK5 elevation produced immuno-regulators that contributed to the elevated and long-lasting leukocyte recruitment into the injured heart and ultimately to chronic cardiac inflammation. We found an increased presence of pro-inflammatory neutrophils and macrophages as well as neutrophils, macrophages and T-lymphocytes at 4-days and 8-weeks respectively post-MI in TgGRK5 hearts. Conversely, GRK5cKO mice were protected from ischemic injury and showed reduced early immune cell recruitment (predominantly monocytes) to the heart, improved contractility and reduced mortality compared to WT post-MI mice. Interestingly, cardiomyocyte-specific GRK2 transgenic mice did not share the same phenotype of TgGRK5 mice and did not have increased cardiac leukocyte migration and cytokine or chemokine production post-MI. CONCLUSIONS: Our study shows that myocyte GRK5 has a crucial and GRK-selective role on the regulation of leucocyte infiltration into the heart, cardiac function and survival in a murine model of post-ischemic HF, supporting GRK5 inhibition as a therapeutic target for HF.


Asunto(s)
Quimiotaxis de Leucocito , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Insuficiencia Cardíaca/enzimología , Leucocitos/metabolismo , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Función Ventricular Izquierda , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Mediadores de Inflamación/metabolismo , Leucocitos/inmunología , Ratones Noqueados , Contracción Miocárdica , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Transducción de Señal , Volumen Sistólico , Transcriptoma , Presión Ventricular
8.
Cardiovasc Res ; 118(1): 282-294, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33386841

RESUMEN

AIMS: Genetic and pharmacological inhibition of mitochondrial fission induced by acute myocardial ischaemia/reperfusion injury (IRI) has been shown to reduce myocardial infarct size. The clinically used anti-hypertensive and heart failure medication, hydralazine, is known to have anti-oxidant and anti-apoptotic effects. Here, we investigated whether hydralazine confers acute cardioprotection by inhibiting Drp1-mediated mitochondrial fission. METHODS AND RESULTS: Pre-treatment with hydralazine was shown to inhibit both mitochondrial fission and mitochondrial membrane depolarisation induced by oxidative stress in HeLa cells. In mouse embryonic fibroblasts (MEFs), pre-treatment with hydralazine attenuated mitochondrial fission and cell death induced by oxidative stress, but this effect was absent in MEFs deficient in the mitochondrial fission protein, Drp1. Molecular docking and surface plasmon resonance studies demonstrated binding of hydralazine to the GTPase domain of the mitochondrial fission protein, Drp1 (KD 8.6±1.0 µM), and inhibition of Drp1 GTPase activity in a dose-dependent manner. In isolated adult murine cardiomyocytes subjected to simulated IRI, hydralazine inhibited mitochondrial fission, preserved mitochondrial fusion events, and reduced cardiomyocyte death (hydralazine 24.7±2.5% vs. control 34.1±1.5%, P=0.0012). In ex vivo perfused murine hearts subjected to acute IRI, pre-treatment with hydralazine reduced myocardial infarct size (as % left ventricle: hydralazine 29.6±6.5% vs. vehicle control 54.1±4.9%, P=0.0083), and in the murine heart subjected to in vivo IRI, the administration of hydralazine at reperfusion, decreased myocardial infarct size (as % area-at-risk: hydralazine 28.9±3.0% vs. vehicle control 58.2±3.8%, P<0.001). CONCLUSION: We show that, in addition to its antioxidant and anti-apoptotic effects, hydralazine, confers acute cardioprotection by inhibiting IRI-induced mitochondrial fission, raising the possibility of repurposing hydralazine as a novel cardioprotective therapy for improving post-infarction outcomes.


Asunto(s)
Dinaminas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Hidralazina/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Dinaminas/metabolismo , Femenino , Células HeLa , Humanos , Preparación de Corazón Aislado , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal
9.
Open Heart ; 8(2)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34819349

RESUMEN

BACKGROUND: Myocardial infarction (MI) is associated with mental health disorders, in which neuroinflammation and cerebral microvascular dysfunction may play a role. Previously, we have shown that the proinflammatory factors Nε-(carboxymethyl)lysine (CML) and NADPH oxidase 2 (NOX2) are increased in the human infarcted heart microvasculature. The aim of this study was to analyse the presence of CML and NOX2 in the cerebral microvasculature of patients with MI. METHODS: Brain tissue was obtained at autopsy from 24 patients with MI and nine control patients. According to their infarct age, patients with MI were divided into three groups: 3-6 hours old (phase I), 6 hours-5 days old (phase II) and 5-14 days old (phase III). CML and NOX2 in the microvasculature were studied through immunohistochemical analysis. RESULTS: We observed a 2.5-fold increase in cerebral microvascular CML in patients with phase II and phase III MI (phase II: 21.39±7.91, p=0.004; phase III: 24.21±10.37, p=0.0007) compared with non-MI controls (8.55±2.98). NOX2 was increased in microvessels in patients with phase II MI (p=0.002) and phase III MI (p=0.04) compared with controls. No correlation was found between CML and NOX2 (r=0.58, p=0.13). CONCLUSIONS: MI coincides with an increased presence of CML and NOX2 in the brain microvasculature. These data point to proinflammatory alterations in the brain microvasculature that may underlie MI-associated mental health disorders.


Asunto(s)
Arterias Cerebrales/enzimología , Lisina/análogos & derivados , Microvasos/enzimología , Infarto del Miocardio/enzimología , NADPH Oxidasa 2/biosíntesis , Enfermedades Neuroinflamatorias/enzimología , Anciano , Biomarcadores/metabolismo , Arterias Cerebrales/patología , Femenino , Humanos , Inmunohistoquímica , Lisina/biosíntesis , Masculino , Microvasos/patología , Persona de Mediana Edad , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Enfermedades Neuroinflamatorias/complicaciones , Enfermedades Neuroinflamatorias/patología
10.
Biomed Res Int ; 2021: 9924549, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34651051

RESUMEN

OBJECTIVE: To study the protective effect of trimetazidine on myocardial cells in rats with myocardial infarction and explore its effect on ERK signaling pathway. METHODS: 40 SD rats were randomly divided into the sham operation group, model group, low-dose group, and high-dose group (intra-abdominal injection of trimetazidine 5 mg/kg and 10 mg/kg, respectively), construction of rat myocardial infarction model by coronary artery left anterior descending artery ligation. 7 days after surgery, the survival rate and cardiac function of each group of rats were recorded. The myocardial infarct size was detected by TTC staining. The apoptosis level of rat cardiomyocytes was detected by TUNEL staining. The content of ROS in rat cardiomyocytes was detected by DCFH-DA. Western-blot was used to detection of Caspase-3, Bcl-2/Bax, and ERK signaling pathway-related proteins in myocardial tissue. RESULTS: Compared with the model group, the survival rate of the rats in the low-dose group and the high-dose group was significantly increased (P < 0.01), the cardiac function was significantly improved (P < 0.01), the myocardial infarct size was significantly decreased (P < 0.01), the level of apoptosis was significantly decreased (P < 0.01), the content of ROS in cardiomyocytes was significantly decreased (P < 0.01), the protein expression of Caspase-3 and NF-κB in cardiomyocytes was significantly decreased (P < 0.01), and the expression of Bcl-2/Bax and p-ERK were significantly increased (P < 0.01). CONCLUSION: Trimetazidine can activate ERK signaling pathway in cardiomyocytes of rats with myocardial infarction, increase the expression of p-ERK, decrease the content of ROS in cardiomyocytes, decrease the expression of apoptotic proteins, reduce myocardial infarct size, improve cardiac function, and increase myocardial function.


Asunto(s)
Cardiotónicos/farmacología , Sistema de Señalización de MAP Quinasas , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Miocardio/patología , Trimetazidina/farmacología , Animales , Apoptosis/efectos de los fármacos , Pruebas de Función Cardíaca , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Infarto del Miocardio/fisiopatología , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Análisis de Supervivencia
11.
Am J Physiol Heart Circ Physiol ; 321(4): H650-H662, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34448639

RESUMEN

The role of the Na+/K+-ATPase (NKA) in heart failure associated with myocardial infarction (MI) is poorly understood. The elucidation of its precise function is hampered by the existence of two catalytic NKA isoforms (NKA-α1 and NKA-α2). Our aim was to analyze the effects of an increased NKA-α2 expression on functional deterioration and remodeling during long-term MI treatment in mice and its impact on Ca2+ handling and inotropy of the failing heart. Wild-type (WT) and NKA-α2 transgenic (TG) mice (TG-α2) with a cardiac-specific overexpression of NKA-α2 were subjected to MI injury for 8 wk. As examined by echocardiography, gravimetry, and histology, TG-α2 mice were protected from functional deterioration and adverse cardiac remodeling. Contractility and Ca2+ transients (Fura 2-AM) in cardiomyocytes from MI-treated TG-α2 animals showed reduced Ca2+ amplitudes during pacing or after caffeine application. Ca2+ efflux in cardiomyocytes from TG-α2 mice was accelerated and diastolic Ca2+ levels were decreased. Based on these alterations, sarcomeres exhibited an enhanced sensitization and thus increased contractility. After the acute stimulation with the ß-adrenergic agonist isoproterenol (ISO), cardiomyocytes from MI-treated TG-α2 mice responded with increased sarcomere shortenings and Ca2+ peak amplitudes. This positive inotropic response was absent in cardiomyocytes from WT-MI animals. Cardiomyocytes with NKA-α2 as predominant isoform minimize Ca2+ cycling but respond to ß-adrenergic stimulation more efficiently during chronic cardiac stress. These mechanisms might improve the ß-adrenergic reserve and contribute to functional preservation in heart failure.NEW & NOTEWORTHY Reduced systolic and diastolic calcium levels in cardiomyocytes from NKA-α2 transgenic mice minimize the desensitization of the ß-adrenergic signaling system. These effects result in an improved ß-adrenergic reserve and prevent functional deterioration and cardiac remodeling.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Insuficiencia Cardíaca/enzimología , Contracción Miocárdica , Infarto del Miocardio/enzimología , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Receptores Adrenérgicos beta/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Remodelación Ventricular , Agonistas Adrenérgicos beta/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Receptores Adrenérgicos beta/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/genética , Remodelación Ventricular/efectos de los fármacos
12.
Cell Physiol Biochem ; 55(4): 489-504, 2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34416105

RESUMEN

BACKGROUND/AIMS: Diaphragm dysfunction with increased reactive oxygen species (ROS) occurs within 72 hrs post-myocardial infarction (MI) in mice and may contribute to loss of inspiratory maximal pressure and endurance in patients. METHODS: We used wild-type (WT) and whole-body Nox4 knockout (Nox4KO) mice to measure diaphragm bundle force in vitro with a force transducer, mitochondrial respiration in isolated fiber bundles with an O2 sensor, mitochondrial ROS by fluorescence, mRNA (RT-PCR) and protein (immunoblot), and fiber size by histology 72 hrs post-MI. RESULTS: MI decreased diaphragm fiber cross-sectional area (CSA) (~15%, p = 0.015) and maximal specific force (10%, p = 0.005), and increased actin carbonylation (5-10%, p = 0.007) in both WT and Nox4KO. Interestingly, MI did not affect diaphragm mRNA abundance of MAFbx/atrogin-1 and MuRF-1 but Nox4KO decreased it by 20-50% (p < 0.01). Regarding the mitochondria, MI and Nox4KO decreased the protein abundance of citrate synthase and subunits of electron transport system (ETS) complexes and increased mitochondrial O2 flux (JO2) and H2O2 emission (JH2O2) normalized to citrate synthase. Mitochondrial electron leak (JH2O2/JO2) in the presence of ADP was lower in Nox4KO and not changed by MI. CONCLUSION: Our study shows that the early phase post-MI causes diaphragm atrophy, contractile dysfunction, sarcomeric actin oxidation, and decreases citrate synthase and subunits of mitochondrial ETS complexes. These factors are potential causes of loss of inspiratory muscle strength and endurance in patients, which likely contribute to the pathophysiology in the early phase post-MI. Whole-body Nox4KO did not prevent the diaphragm abnormalities induced 72 hrs post-MI, suggesting that systemic pharmacological inhibition of Nox4 will not benefit patients in the early phase post-MI.


Asunto(s)
Diafragma/enzimología , Mitocondrias Musculares/enzimología , Contracción Muscular , Atrofia Muscular/enzimología , Infarto del Miocardio/enzimología , NADPH Oxidasa 4/deficiencia , Animales , Diafragma/patología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Musculares/genética , Mitocondrias Musculares/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , NADPH Oxidasa 4/metabolismo
13.
Pflugers Arch ; 473(8): 1273-1285, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34231059

RESUMEN

Myocardial infarction (MI) is one of the leading causes of death worldwide. Prognosis and mortality rate are directly related to infarct size and post-infarction pathological heart remodeling, which can lead to heart failure. Hypoxic MI-affected areas increase the expression of hypoxia-inducible factor (HIF-1), inducing infarct size reduction and improving cardiac function. Hypoxia translocates HIF-1 to the nucleus, activating carbonic anhydrase IX (CAIX) transcription. CAIX regulates myocardial intracellular pH, critical for heart performance. Our objective was to investigate CAIX participation and relation with sodium bicarbonate transporters 1 (NBC1) and HIF-1 in cardiac remodeling after MI. We analyzed this pathway in an "in vivo" rat coronary artery ligation model and isolated cardiomyocytes maintained under hypoxia. Immunohistochemical studies revealed an increase in HIF-1 levels after 2 h of infarction. Similar results were observed in 2-h infarcted cardiac tissue (immunoblotting) and in hypoxic cardiomyocytes with a nuclear distribution (confocal microscopy). Immunohistochemical studies showed an increase CAIX in the infarcted area at 2 h, mainly distributed throughout the cell and localized in the plasma membrane at 24 h. Similar results were observed in 2 h in infarcted cardiac tissue (immunoblotting) and in hypoxic cardiomyocytes (confocal microscopy). NBC1 expression increased in cardiac tissue after 2 h of infarction (immunoblotting). CAIX and NBC1 interaction increases in cardiac tissue subjected to MI for 2h when CAIX is present (immunoprecipitation). These results suggest that CAIX interacts with NBC1 in our infarct model as a mechanism to prevent acidic damage in hypoxic tissue, making it a promising therapeutic target.


Asunto(s)
Anhidrasa Carbónica IX/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/enzimología , Infarto del Miocardio/enzimología , Simportadores de Sodio-Bicarbonato/metabolismo , Animales , Masculino , Cultivo Primario de Células , Ratas Wistar , Remodelación Ventricular
14.
Cell Death Dis ; 12(7): 665, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215721

RESUMEN

It has been reported that growth differentiation factor 11 (GDF11) protects against myocardial ischemia/reperfusion (IR) injury, but the underlying mechanisms have not been fully clarified. Considering that GDF11 plays a role in the aging/rejuvenation process and that aging is associated with telomere shortening and cardiac dysfunction, we hypothesized that GDF11 might protect against IR injury by activating telomerase. Human plasma GDF11 levels were significantly lower in acute coronary syndrome patients than in chronic coronary syndrome patients. IR mice with myocardial overexpression GDF11 (oe-GDF11) exhibited a significantly smaller myocardial infarct size, less cardiac remodeling and dysfunction, fewer apoptotic cardiomyocytes, higher telomerase activity, longer telomeres, and higher ATP generation than IR mice treated with an adenovirus carrying a negative control plasmid. Furthermore, mitochondrial biogenesis-related proteins and some antiapoptotic proteins were significantly upregulated by oe-GDF11. These cardioprotective effects of oe-GDF11 were significantly antagonized by BIBR1532, a specific telomerase inhibitor. Similar effects of oe-GDF11 on apoptosis and mitochondrial energy biogenesis were observed in cultured neonatal rat cardiomyocytes, whereas GDF11 silencing elicited the opposite effects to oe-GDF11 in mice. We concluded that telomerase activation by GDF11 contributes to the alleviation of myocardial IR injury through enhancing mitochondrial biogenesis and suppressing cardiomyocyte apoptosis.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Mitocondrias Cardíacas/enzimología , Infarto del Miocardio/enzimología , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Biogénesis de Organelos , Telomerasa/metabolismo , Aminobenzoatos/farmacología , Animales , Apoptosis , Proteínas Morfogenéticas Óseas/genética , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Factores de Diferenciación de Crecimiento/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Naftalenos/farmacología , Ratas , Transducción de Señal , Telomerasa/antagonistas & inhibidores
15.
Basic Res Cardiol ; 116(1): 34, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34018053

RESUMEN

Reducing infarct size (IS) by interfering with mechanisms for cardiomyocyte death remains an elusive goal. DMX-5804, a selective inhibitor of the stress-activated kinase MAP4K4, suppresses cell death in mouse myocardial infarction (MI), human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), and 3D human engineered heart tissue, whose fidelity to human biology is hoped to strengthen the route to clinical success. Here, DMX-10001, a soluble, rapidly cleaved pro-drug of DMX-5804, was developed for i.v. testing in large-mammal MI. Following pharmacodynamic studies, a randomized, blinded efficacy study was performed in swine subjected to LAD balloon occlusion (60 min) and reperfusion (24 h). Thirty-six animals were enrolled; 12 were excluded by pre-defined criteria, death before infusion, or technical issues. DMX-10001 was begun 20 min before reperfusion (30 min, 60 mg/kg/h; 23.5 h, 17 mg/kg/h). At all times tested, beginning 30 min after the start of infusion, DMX-5804 concentrations exceeded > fivefold the levels that rescued hPSC-CMs and reduced IS in mice after oral dosing with DMX-5804 itself. No significant reduction occurred in IS or no-reflow corrected for the area at ischemic risk, even though DMX-10001 reduced IS, expressed in grams or % of LV mass, by 27%. In summary, a rapidly cleaved pro-drug of DMX-5804 failed to reduce IS in large-mammal MI, despite exceeding the concentrations for proven success in both mice and hPSC-CMs.


Asunto(s)
Células Madre Pluripotentes Inducidas/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Profármacos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Femenino , Hemodinámica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Profármacos/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Serina-Treonina Quinasas/metabolismo , Sus scrofa , Investigación Biomédica Traslacional , Función Ventricular Izquierda/efectos de los fármacos
16.
Free Radic Res ; 55(3): 267-274, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34003050

RESUMEN

Glutathione S-transferases (GSTs) are the family of enzymes involved in the second line of defense against oxidative stress (OS). The lack of GSTT1/GSTM1 enzyme quantity or activity, due to the presence of homozygous deletion compromises antioxidative defense resulting in OS. OS is the critical mechanism in the pathophysiology of atherosclerosis, coronary artery disease, and myocardial infarction (MI). The increase in reactive oxygen species together with the process of apoptosis plays a role in left ventricular remodeling (LVR) after MI. The associations of GSTT1 and GSTM1 gene polymorphisms with the risk of MI are inconsistent. The aim was to analyze the association of GSTT1/GSTM1 null genotypes with first MI and LVR 8 months after the MI. The study involved 330 controls and 438 consecutive patients with symptoms and signs of first MI. The subgroup of 150 MI patients was prospectively followed up for 6 months. Evidence of maladaptive LVR was obtained by 2D Doppler echocardiography 3-5 days and 6 months after the MI. A multiplex polymerase chain reaction was used to detect the deletion in GSTT1 and GSTM1 genes. GSTM1 null genotype was significantly and independently associated with first MI (adjusted OR = 1.45 95% CI 1.03-2.03, p = 0.03). Association of double null genotypes with maladaptive LVR in patients 6 months after the first MI was no longer significant after adjustment for factors that differed significantly between patients with and without maladaptive LVR. This study demonstrated the association of GSTM1 null genotypes with the risk of MI in the Serbian population.


Asunto(s)
Glutatión Transferasa/metabolismo , Infarto del Miocardio/enzimología , Femenino , Predisposición Genética a la Enfermedad , Glutatión Transferasa/genética , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/genética , Estudios Prospectivos , Factores de Riesgo , Eliminación de Secuencia
17.
Cardiovasc Toxicol ; 21(8): 669-682, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33999393

RESUMEN

We aimed to explore the regulatory mechanism of the axis of miR-223-3p-PIK3C2A-PI3K/Akt on cardiomyocyte apoptosis in rats with myocardial infarction. Thirty 8-week-old healthy male SD rats were used for establishing the sham group and the model group, with HE staining, TUNEL staining, and TTC staining performed. After the identification of the targeting relationship between PIK3C2A and miR-223-3p, experimental rats were randomly divided into seven groups by plasmid transfection, including the Blank group, negative control (NC) group, miR-223-3p mimic group, miR-223-3p inhibitor group, siRNA-PIK3C2A group, oe-PIK3C2A group, and miR-223-3p inhibitor + oe-PIK3C2A group. Four weeks after transfection, the expression levels of miR-223-3p and PIK3C2A in tissues as well as PI3K, Akt, Bax, and bcl-2 mRNA in cells were detected by qRT-PCR and western blot, in combination with the detection of apoptosis rate by flow cytometry. Compared with the sham group, the model group showed typical myocardial injury and abnormal staining, higher apoptotic index, and larger myocardial infarction area (all P < 0.05). PIK3C2A was the target gene of miR-223-3p. The expression level of miR-223-3p in model group was significantly lower than that in sham group, while the mRNA and protein expression levels of PIK3C2A increased significantly (all P < 0.05). In cell tests, the expression level of miR-223-3p increased significantly in miR-223-3p mimic group (P < 0.05), which, however, showed no significant change in siRNA-PIK3C2A group (P > 0.05). MiR-223-3p inhibitor group and siRNA-PIK3C2A group had obviously increased PI3K, Akt, mTOR and Bcl-2 mRNA, and protein expression, while decreased mRNA and protein expression of PIK3C2A and Bax (all P < 0.05); miR-223-3p mimic groups had the opposite trends (all P < 0.05). siRNA-PIK3C2A + miR-223-3p mimic showed no obvious change relative to the control groups (all P > 0.05). Low expression of miR-223-3p may downregulate PIK3C2A expression, resulting in the inhibition of myocardial cell apoptosis in rats with myocardial infarction via the activation of PI3K/Akt signaling pathway.


Asunto(s)
Apoptosis , MicroARNs , Infarto del Miocardio , Miocitos Cardíacos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Animales , Masculino , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Transducción de Señal
19.
BMC Cardiovasc Disord ; 21(1): 197, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879068

RESUMEN

BACKGROUND: Acute myocardial infarction (AMI) is myocardial necrosis caused by acute coronary ischemia and hypoxia. It can be complicated by arrhythmia, shock, heart failure and other symptoms that can be life-threatening. A multi-regulator driven dysfunction module for AMI was constructed. It is intended to explore the pathogenesis and functional pathways regulation of acute myocardial infarction. METHODS: Combining differential expression analysis, co-expression analysis, and the functional enrichment analysis, a set of expression disorder modules related to AMI was obtained. Hypergeometric test was performed to calculate the potential regulatory effects of multiple factors on the module, identifying a range of non-coding RNA and transcription factors. RESULTS: A total of 4551 differentially expressed genes for AMI and seven co-expression modules were obtained. These modules are primarily involved in the metabolic processes of prostaglandin transport processes, regulating DNA recombination and AMPK signal transduction. Based on this set of functional modules, 3 of 24 transcription factors (TFs) including NFKB1, MECP2 and SIRT1, and 3 of 782 non-coding RNA including miR-519D-3P, TUG1 and miR-93-5p were obtained. These core regulators are thought to be involved in the progression of AMI disease. Through the AMPK signal transduction, the critical gene stearoyl-CoA desaturase (SCD) can lead to the occurrence and development of AMI. CONCLUSIONS: In this study, a dysfunction module was used to explore the pathogenesis of multifactorial mediated AMI and provided new methods and ideas for subsequent research. It helps researchers to have a deeper understanding of its potential pathogenesis. The conclusion provides a theoretical basis for biologists to design further experiments related to AMI.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Infarto del Miocardio/enzimología , Miocardio/enzimología , Estearoil-CoA Desaturasa/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Bases de Datos de Ácidos Nucleicos , Progresión de la Enfermedad , Regulación Enzimológica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Necrosis , Mapas de Interacción de Proteínas , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal , Estearoil-CoA Desaturasa/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Basic Res Cardiol ; 116(1): 21, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33751227

RESUMEN

Myocardial connexin 43 (Cx43) forms gap junctions and hemichannels, and is also present within subsarcolemmal mitochondria. The protein is phosphorylated by several kinases including mitogen-activated protein kinase (MAPK), protein kinase C (PKC), and casein kinase 1 (CK1). A reduction in Cx43 content abrogates myocardial infarct size reduction by ischemic preconditioning (IPC). The present study characterizes the contribution of Cx43 phosphorylation towards mitochondrial function, hemichannel activity, and the cardioprotection by IPC in wild-type (WT) mice and in mice in which Cx43-phosphorylation sites targeted by above kinases are mutated to non-phosphorylatable residues (Cx43MAPKmut, Cx43PKCmut, and Cx43CK1mut mice). The amount of Cx43 in the left ventricle and in mitochondria was reduced in all mutant strains compared to WT mice and Cx43 phosphorylation was altered at residues not directly targeted by the mutations. Whereas complex 1 respiration was reduced in all strains, complex 2 respiration was decreased in Cx43CK1mut mice only. In Cx43 epitope-mutated mice, formation of reactive oxygen species and opening of the mitochondrial permeability transition pore were not affected. The hemichannel open probability was reduced in Cx43PKCmut and Cx43CK1mut but not in Cx43MAPKmut cardiomyocytes. Infarct size in isolated saline-perfused hearts after ischemia/reperfusion (45 min/120 min) was comparable between genotypes and was significantly reduced by IPC (3 × 3 min ischemia/5 min reperfusion) in WT, Cx43MAPKmut, and Cx43PKCmut, but not in Cx43CK1mut mice, an effect independent from the amount of Cx43 and the probability of hemichannel opening. Taken together, our study shows that alterations of Cx43 phosphorylation affect specific cellular functions and highlights the importance of Cx43 phosphorylation by CK1 for IPC's cardioprotection.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Conexina 43/metabolismo , Precondicionamiento Isquémico Miocárdico , Mitocondrias Cardíacas/enzimología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/enzimología , Animales , Conexina 43/genética , Modelos Animales de Enfermedad , Preparación de Corazón Aislado , Ratones Mutantes , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , 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 , Miocitos Cardíacos/patología , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...