Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Mol Cell Biochem ; 479(2): 337-350, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37074505

RESUMEN

Doxorubicin (DOXO) induces marked cardiotoxicity, though increased oxidative stress while there are some documents related with cardioprotective effects of some antioxidants against organ-toxicity during cancer treatment. Although magnolia bark has some antioxidant-like effects, its action in DOXO-induced heart dysfunction has not be shown clearly. Therefore, here, we aimed to investigate the cardioprotective action of a magnolia bark extract with active component magnolol and honokiol complex (MAHOC; 100 mg/kg) in DOXO-treated rat hearts. One group of adult male Wistar rats was injected with DOXO (DOXO-group; a cumulative dose of 15 mg/kg in 2-week) or saline (CON-group). One group of DOXO-treated rats was administered with MAHOC before DOXO (Pre-MAHOC group; 2-week) while another group was administered with MAHOC following the 2-week DOXO (Post-MAHOC group). MAHOC administration, before or after DOXO, provided full survival of animals during 12-14 weeks, and significant recoveries in the systemic parameters of animals such as plasma levels of manganese and zinc, total oxidant and antioxidant statuses, and also systolic and diastolic blood pressures. This treatment also significantly improved heart function including recoveries in end-diastolic volume, left ventricular end-systolic volume, heart rate, cardiac output, and prolonged P-wave duration. Furthermore, the MAHOC administrations improved the structure of left ventricles such as recoveries in loss of myofibrils, degenerative nuclear changes, fragmentation of cardiomyocytes, and interstitial edema. Biochemical analysis in the heart tissues provided the important cardioprotective effect of MAHOC on the redox regulation of the heart, such as improvements in activities of glutathione peroxidase and glutathione reductase, and oxygen radical-absorbing capacity of the heart together with recoveries in other systemic parameters of animals, while all of these benefits were observed in the Pre-MAHOC treatment group, more prominently. Overall, one can point out the beneficial antioxidant effects of MAHOC in chronic heart diseases as a supporting and complementing agent to the conventional therapies.


Asunto(s)
Compuestos Alílicos , Antioxidantes , Compuestos de Bifenilo , Cardiotoxicidad , Lignanos , Fenoles , Masculino , Ratas , Animales , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/prevención & control , Ratas Wistar , Antioxidantes/farmacología , Miocitos Cardíacos , Doxorrubicina/toxicidad , Estrés Oxidativo
2.
Am J Physiol Cell Physiol ; 324(5): C1017-C1027, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36878847

RESUMEN

Sirtuins are NAD+-dependent deacetylases with beneficial roles in conditions relevant to human health, including metabolic disease, type II diabetes, obesity, cancer, aging, neurodegenerative diseases, and cardiac ischemia. Since ATP-sensitive K+ (KATP) channels have cardioprotective roles, we investigated whether they are regulated by sirtuins. Nicotinamide mononucleotide (NMN) was used to increase cytosolic NAD+ levels and to activate sirtuins in cell lines, isolated rat and mouse cardiomyocytes or insulin-secreting INS-1 cells. KATP channels were studied with patch clamping, biochemistry techniques, and antibody uptake experiments. NMN led to an increase in intracellular NAD+ levels and an increase in the KATP channel current, without significant changes in the unitary current amplitude or open probability. An increased surface expression was confirmed using surface biotinylation approaches. The rate of KATP channel internalization was diminished by NMN, which may be a partial explanation for the increased surface expression. We show that NMN acts via sirtuins since the increased KATP channel surface expression was prevented by blockers of SIRT1 and SIRT2 (Ex527 and AGK2) and mimicked by SIRT1 activation (SRT1720). The pathophysiological relevance of this finding was studied using a cardioprotection assay with isolated ventricular myocytes, in which NMN protected against simulated ischemia or hypoxia in a KATP channel-dependent manner. Overall, our data draw a link between intracellular NAD+, sirtuin activation, KATP channel surface expression, and cardiac protection against ischemic damage.


Asunto(s)
Diabetes Mellitus Tipo 2 , Sirtuinas , Ratas , Ratones , Humanos , Animales , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo , NAD/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Miocitos Cardíacos/metabolismo , Adenosina Trifosfato/metabolismo , Canales KATP/genética , Canales KATP/metabolismo
3.
Cell Biochem Funct ; 41(8): 1526-1542, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38014767

RESUMEN

Cardiac conduction abnormalities are disorders in metabolic syndrome (MetS), however, their mechanisms are unknown. Although ventricular arrhythmia reflects the changes in QT-interval of electrocardiograms associated with the changes in cardiomyocyte action potential durations (APDs), recent studies emphasize role of intercellular crosstalk between cardiomyocytes and nonmyocytes via passive (electrotonic)-conduction. Therefore, considering the possible increase in intercellular interactions of nonmyocytes with cardiomyocytes, we hypothesized an early-cardiac-remodeling characterized by short QT-interval via contributions and modulations of changes by nonmyocytes to the ventricular APs in an early-stage MetS hearts. Following the feeding of 8-week-old rats with a high-sucrose diet (32%; MetS rats) and validation of insulin resistance, there was a significant increase in heart rate and changes in the electrical characteristics of the hearts, especially a shortening in action potential (AP) duration of the papillary muscles. The patch-clamp analysis of ventricular cardiomyocytes showed an increase in the Na+ -channel currents while there were decreases in  l-type Ca2+ -channel (LTCC) currents with unchanged K+ -channel currents. There was an increase in the phosphorylated form of connexin 43 (pCx43), mostly with lateral localization on sarcolemma, while its unphosphorylated form (Cx43) exhibited a high degree of localization within intercalated discs. A high-level positively-stained α-SMA and CD68 cells were prominently localized and distributed in interfibrillar spaces of the heart, implying the possible contributions of myofibroblasts and macrophages to both shortened APDs and abnormal electrical conduction in MetS hearts. Our data propose a previously unrecognized pathway for SQT induction in the heart. This pathway includes not only the contribution of short ventricular-APDs via ionic mechanisms but also increasing contributions of the electrotonic-cardiomyocyte depolarization, spontaneous electrical activity-associated fast heterogeneous impulse conduction in the heart via increased interactions and relocations between cardiomyocytes and nonmyocytes, which may be an explanation for the development of an SQT in early-cardiac-remodeling.


Asunto(s)
Arritmias Cardíacas , Miocitos Cardíacos , Ratas , Animales , Miocitos Cardíacos/metabolismo , Arritmias Cardíacas/metabolismo , Miocardio/metabolismo , Electrocardiografía , Potenciales de Acción
4.
Biochem Genet ; 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38091184

RESUMEN

Intracellular free Zn2+ ([Zn2+]i) is less than 1-nM in cardiomyocytes and its regulation is performed with Zn2+-transporters. However, the roles of Zn2+-transporters in cardiomyocytes are not defined exactly yet. Here, we aimed to examine the role of an overexpression and subcellular localization of a ZnT6 in insulin-resistance mimic H9c2 cardiomyoblasts (IR-cells; 50-µM palmitic acid for 24-h incubation). We used both IR-cells and ZnT6-overexpressed (ZnT6OE) cells in comparison to those of H9c2 cells (CON-cells). The IR-cells have higher ZnT6-protein levels than CON-cells while this level was similar to those of ZnT6OE-cells. The [Zn2+]i in IR-cells was increased significantly and mitochondrial localization of ZnT6 was demonstrated in these cells by using confocal microscopy visualization. Furthermore, electron microscopy analysis demonstrated abnormal morphological appearance in both IR-cells and ZnT6OE-cells characterized by irregular mitochondrion cristae and condensed and dilated cisterna in the sarcoplasmic reticulum. Mitochondria were similarly depolarized in both IR-cells and ZnT6OE-cells. The protein expression level of a mitofusin protein MFN2 in the IR-cells was decreased, significantly, whereas, it was found significantly upregulated in both ZnT6-OE-cells and IR-incubated ZnT6OE-cells, which demonstrates the role of ZnT6-overexpression but not IR. Additionally, the total protein level of a mitochondrial fission protein, dynamin-related protein 1, DRP1 was found to be increased over 1.5-fold in IR-cells while this increase was found to be higher in the ZnT6OE-cells than those of IR-cells, demonstrating an additional effect on IR-increase. ZnT6-overexpression induced also significant increases in K-acetylation, trimethylation of histone H3 lysine27, and mono-methylation of histone H3 lysine36, in a similar manner to those of IR-cells. Overall, our data point out an important contribution of ZnT6-overexpression to IR-induced cellular changes, such as alteration in mitochondria function and activation of epigenetic modifications.

5.
J Cell Physiol ; 237(2): 1353-1371, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34632595

RESUMEN

Insufficient-heart function is associated with myocardial insulin resistance in the elderly, particularly associated with long-QT, in a dependency on dysfunctional KCNQ1/KCNE1-channels. So, we aimed to examine the contribution of alterations in KCNQ1/KCNE1-current (IKs ) to the aging-related remodeling of the heart as well as the role of insulin treatment on IKs in the aged rats. Prolonged late-phase action potential (AP) repolarization of ventricular cardiomyocytes from insulin-resistant 24-month-old rats was significantly reversed by in vitro treatment of insulin or PKG inhibitor (in vivo, as well) via recovery in depressed IKs . Although the protein level of either KCNQ1 or KCNE1 in cardiomyocytes was not affected with aging, PKG level was significantly increased in those cells. The inhibited IKs in ß3 -ARs-stimulated cells could be reversed with a PKG inhibitor, indicating the correlation between PKG-activation and ß3 -ARs activation. Furthermore, in vivo treatment of aged rats, characterized by ß3 -ARs activation, with either insulin or a PKG inhibitor for 2 weeks provided significant recoveries in IKs , prolonged late phases of APs, prolonged QT-intervals, and low heart rates without no effect on insulin resistance. In vivo insulin treatment provided also significant recovery in increased PKG and decreased PIP2 level, without the insulin effect on the KCNQ1 level in ß3 -ARs overexpressed cells. The inhibition of IKs in aged-rat cardiomyocytes seems to be associated with activated ß3 -ARs dependent remodeling in the interaction between KCNQ1 and KCNE1. Significant recoveries in ventricular-repolarization of insulin-treated aged cardiomyocytes via recovery in IKs strongly emphasize two important issues: (1) IKs can be a novel target in aging-associated remodeling in the heart and insulin may be a cardioprotective agent in the maintenance of normal heart function during the aging process. (2) This study is one of the first to demonstrate insulin's benefits on long-QT in insulin-resistant aged rats by accelerating the ventricular AP repolarization through reversing the depressed IKs via affecting the ß3 -ARs signaling pathway and particularly affecting activated PKG.


Asunto(s)
Resistencia a la Insulina , Síndrome de QT Prolongado , Canales de Potasio con Entrada de Voltaje , Potenciales de Acción , Animales , Insulina/metabolismo , Insulina/farmacología , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Síndrome de QT Prolongado/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Ratas , Transducción de Señal
6.
Mol Cell Biochem ; 477(11): 2609-2625, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35598217

RESUMEN

Recent studies discuss the evidence of lesser degrees of hyperglycemia contribution to cardiovascular disease (CVD) than impaired glucose tolerance. Indeed, the biggest risk for CVD seems to shift to glucose intolerance in humans with insulin resistance. Although there is a connection between abnormal insulin signaling and heart dysfunction in diabetics, there is also a relation between cardiac insulin resistance and aging heart failure (HF). Moreover, studies have revealed that HF is associated with generalized insulin resistance. Recent clinical outcomes parallel to the experimental data undertaken with antihyperglycemic drugs have shown their beneficial effects on the cardiovascular system through a direct effect on the myocardium, beyond their ability to lower blood glucose levels and their receptor-associated actions. In this regard, several new-class drugs, such as glucagon-like peptide 1 receptor agonists (GLP-1Ra) and sodium-glucose cotransport 2 inhibitors (SGLT2i), can improve cardiac health beyond their ability to control glycemia. In recent years, great improvements have been made toward the possibility of direct heart-targeting effects including modulation of the expression of specific cardiac genes in vivo for therapeutic purposes. However, many questions remain unanswered, regarding their therapeutic effects on cardiomyocytes in heart failure, although there are various cellular levels studies with these drugs. There are also some important comparative studies on the role of SGLT2i versus GLP-1Ra in patients with and without CVD as well as with or without hyperglycemia. Here, we sought to summarize and interpret the available evidence from clinical studies focusing on the effects of either GLP-1Ra or SGLT-2i or their combinations on cardiac structure and function. Furthermore, we documented data from experimental studies, at systemic, organ, and cellular levels. Overall, one can summarize that both clinical and experimental data support that either SGLT2i or GLP-1R agonists have similar benefits as cardioprotective agents in patients with or without impaired glucose tolerance.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Insuficiencia Cardíaca , Resistencia a la Insulina , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Enfermedades Cardiovasculares/tratamiento farmacológico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptido 1 Similar al Glucagón/agonistas , Intolerancia a la Glucosa/tratamiento farmacológico , Insuficiencia Cardíaca/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
7.
BMC Cardiovasc Disord ; 22(1): 147, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35379188

RESUMEN

PURPOSE: Metabolic syndrome (MetS) became a tremendous public health burden in the last decades. Store-operated calcium entry (SOCE) is a unique mechanism that causes a calcium influx, which is triggered by calcium store depletion. MetS-induced alterations in cardiac calcium signaling, especially in SOCE are still unclear. Therefore, we aim to examine the possible role of SOCE and its components (STIM1 and Orai1) in the MetS-induced cardiac remodeling. METHODS: We used male, adult (12 weeks) Wistar albino rats (n = 20). Animals were randomly divided into two groups which were: control (C) and MetS. We gave 33% sucrose solution to animals instead of water for 24 weeks to establish MetS model. In the end, papillary muscle function was evaluated, and various electrophysiological analyses were made in isolated cardiomyocytes. Additionally, STIM1 and Orai1 protein and mRNA expressions were analyzed. RESULTS: We observed a deterioration in contractility in MetS animals and demonstrated the contribution of SOCE by applying a SOCE inhibitor (BTP2). Calcium spark frequency was increased while its amplitude was decreasing in MetS hearts, which was reversed after SOCE inhibition. The amplitude of transient calcium changes in the MetS group was decreased, and it decreased further BTP2 application. Both protein and mRNA levels of STIM1 and Orai1 were increased significantly in MetS hearts. CONCLUSION: Current data indicate the significant contribution of SOCE to cardiac calcium handling in the MetS model. We think MetS-induced SOCE activation is a compensation mechanism that is required for the continuum of proper cardiac functioning, although the activation can also cause cardiac hypertrophy.


Asunto(s)
Calcio , Insulina , Animales , Masculino , Ratas , Calcio/metabolismo , Señalización del Calcio , Proteínas de Neoplasias , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Ratas Wistar , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
8.
Clin Exp Pharmacol Physiol ; 49(1): 46-59, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34519087

RESUMEN

The pleiotropic effects of glucagon-like peptide-1 receptor (GLP-1R) agonists on the heart have been recognised in obese or diabetic patients. However, little is known regarding the molecular mechanisms of these agonists in cardioprotective actions under metabolic disturbances. We evaluated the effects of GLP-1R agonist liraglutide treatment on left ventricular cardiomyocytes from high-carbohydrate induced metabolic syndrome rats (MetS rats), characterised with insulin resistance and cardiac dysfunction with a long-QT. Liraglutide (0.3 mg/kg for 4 weeks) treatment of MetS rats significantly reversed long-QT, through a shortening the prolonged action potential duration and recovering inhibited K+ -currents. We also determined a significant recovery in the leaky sarcoplasmic reticulum (SR) and high cytosolic Ca2+ -level, which are confirmed with a full recovery in activated Na+ /Ca2+ -exchanger currents (INCX ). Moreover, the liraglutide treatment significantly reversed the depolarised mitochondrial membrane potential (MMP), increased production of oxidant markers, and cellular acidification together with the depressed ATP production. Our light microscopy analysis of isolated cardiomyocytes showed marked recoveries in the liraglutide-treated MetS group such as marked reverses in highly dilated T-tubules and SR-mitochondria junctions. Moreover, we determined a significant increase in depressed GLUT4 protein level in liraglutide-treated MetS group, possibly associated with recovery in casein kinase 2α. Overall, the study demonstrated a molecular mechanism of liraglutide-induced cardioprotection in MetS rats, at most, via its pleiotropic effects, such as alleviation in the electrical abnormalities, Ca2+ -homeostasis, and mitochondrial dysfunction in ventricular cardiomyocytes.


Asunto(s)
Calcio/metabolismo , Carbohidratos de la Dieta/efectos adversos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Síndrome Metabólico/tratamiento farmacológico , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Carbohidratos de la Dieta/administración & dosificación , Glucosa/metabolismo , Liraglutida/uso terapéutico , Síndrome Metabólico/fisiopatología , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Ratas , Ratas Wistar
9.
Mol Cell Biochem ; 476(2): 971-989, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33225416

RESUMEN

An important energy supplier of cardiomyocytes is mitochondria, similar to other mammalian cells. Studies have demonstrated that any defect in the normal processes controlled by mitochondria can lead to abnormal ROS production, thereby high oxidative stress as well as lack of ATP. Taken into consideration, the relationship between mitochondrial dysfunction and overproduction of ROS as well as the relation between increased ROS and high-level release of intracellular labile Zn2+, those bring into consideration the importance of the events related with those stimuli in cardiomyocytes responsible from cellular Zn2+-homeostasis and responsible Zn2+-transporters associated with the Zn2+-homeostasis and Zn2+-signaling. Zn2+-signaling, controlled by cellular Zn2+-homeostatic mechanisms, is regulated with intracellular labile Zn2+ levels, which are controlled, especially, with the two Zn2+-transporter families; ZIPs and ZnTs. Our experimental studies in mammalian cardiomyocytes and human heart tissue showed that Zn2+-transporters localizes to mitochondria besides sarco(endo)plasmic reticulum and Golgi under physiological condition. The protein levels as well as functions of those transporters can re-distribute under pathological conditions, therefore, they can interplay among organelles in cardiomyocytes to adjust a proper intracellular labile Zn2+ level. In the present review, we aimed to summarize the already known Zn2+-transporters localize to mitochondria and function to stabilize not only the cellular Zn2+ level but also cellular oxidative stress status. In conclusion, one can propose that a detailed understanding of cellular Zn2+-homeostasis and Zn2+-signaling through mitochondria may emphasize the importance of new mitochondria-targeting agents for prevention and/or therapy of cardiovascular dysfunction in humans.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Mitocondrias/patología , Miocitos Cardíacos/patología , Estrés Oxidativo/fisiología , Zinc/metabolismo , Animales , Humanos , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo
10.
Mol Cell Biochem ; 476(10): 3827-3844, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34114148

RESUMEN

Metabolic syndrome (MetS) is associated with additional cardiovascular risk in mammalians while there are relationships between hyperglycemia-associated cardiovascular dysfunction and increased platelet P2Y12 receptor activation. Although P2Y12 receptor antagonist ticagrelor (Tica) plays roles in reduction of cardiovascular events, its beneficial mechanism remains poorly understood. Therefore, we aimed to clarify whether Tica can exert a direct protective effect in ventricular cardiomyocytes from high-carbohydrate diet-induced MetS rats, at least, through affecting sarcoplasmic reticulum (SR)-mitochondria (Mit) miscommunication. Tica treatment of MetS rats (150 mg/kg/day for 15 days) significantly reversed the altered parameters of action potentials by reversing sarcolemmal ionic currents carried by voltage-dependent Na+ and K+ channels, and Na+/Ca2+-exchanger in the cells, expressed P2Y12 receptors. The increased basal-cytosolic Ca2+ level and depressed SR Ca2+ load were also reversed in Tica-treated cells, at most, though recoveries in the phosphorylation levels of ryanodine receptors and phospholamban. Moreover, there were marked recoveries in Mit structure and function (including increases in both autophagosomes and fragmentations) together with recoveries in Mit proteins and the factors associated with Ca2+ transfer between SR-Mit. There were further significant recoveries in markers of both ER stress and oxidative stress. Taken into consideration the Tica-induced prevention of ER stress and mitochondrial dysfunction, our data provided an important document on the pleiotropic effects of Tica in the electrical activity of the cardiomyocytes from MetS rats. This protective effect seems through recoveries in SR-Mit miscommunication besides modulation of different sarcolemmal ion-channel activities, independent of P2Y12 receptor antagonism.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Carbohidratos de la Dieta/efectos adversos , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Ticagrelor/farmacología , Animales , Carbohidratos de la Dieta/farmacología , Transporte Iónico/efectos de los fármacos , Masculino , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/patología , Ratas , Ratas Wistar , Retículo Sarcoplasmático/patología , Transducción de Señal/efectos de los fármacos
11.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202017

RESUMEN

BACKGROUND: Diabetic patients have prolonged cardiac repolarization and higher risk of arrhythmia. Besides, diabetes activates the innate immune system, resulting in higher levels of plasmatic cytokines, which are described to prolong ventricular repolarization. METHODS: We characterize a metabolic model of type 2 diabetes (T2D) with prolonged cardiac repolarization. Sprague-Dawley rats were fed on a high-fat diet (45% Kcal from fat) for 6 weeks, and a low dose of streptozotozin intraperitoneally injected at week 2. Body weight and fasting blood glucose were measured and electrocardiograms of conscious animals were recorded weekly. Plasmatic lipid profile, insulin, cytokines, and arrhythmia susceptibility were determined at the end of the experimental period. Outward K+ currents and action potentials were recorded in isolated ventricular myocytes by patch-clamp. RESULTS: T2D animals showed insulin resistance, hyperglycemia, and elevated levels of plasma cholesterol, triglycerides, TNFα, and IL-1b. They also developed bradycardia and prolonged QTc-interval duration that resulted in increased susceptibility to severe ventricular tachycardia under cardiac challenge. Action potential duration (APD) was prolonged in control cardiomyocytes incubated 24 h with plasma isolated from diabetic rats. However, adding TNFα and IL-1b receptor blockers to the serum of diabetic animals prevented the increased APD. CONCLUSIONS: The elevation of the circulating levels of TNFα and IL-1b are responsible for impaired ventricular repolarization and higher susceptibility to cardiac arrhythmia in our metabolic model of T2D.


Asunto(s)
Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Biomarcadores/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Susceptibilidad a Enfermedades , Mediadores de Inflamación/sangre , Animales , Arritmias Cardíacas/diagnóstico , Citocinas/sangre , Diabetes Mellitus Tipo 2/etiología , Modelos Animales de Enfermedad , Insulina/metabolismo , Resistencia a la Insulina , Canales de Potasio/metabolismo , Ratas , Remodelación Ventricular
12.
J Cell Mol Med ; 24(15): 8567-8578, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32652890

RESUMEN

The prevalence of death from cardiovascular disease is significantly higher in elderly populations; the underlying factors that contribute to the age-associated decline in cardiac performance are poorly understood. Herein, we identify the involvement of sodium/glucose co-transporter gene (SGLT2) in disrupted cellular Ca2+ -homeostasis, and mitochondrial dysfunction in age-associated cardiac dysfunction. In contrast to younger rats (6-month of age), older rats (24-month of age) exhibited severe cardiac ultrastructural defects, including deformed, fragmented mitochondria with high electron densities. Cardiomyocytes isolated from aged rats demonstrated increased reactive oxygen species (ROS), loss of mitochondrial membrane potential and altered mitochondrial dynamics, compared with younger controls. Moreover, mitochondrial defects were accompanied by mitochondrial and cytosolic Ca2+ ([Ca2+ ]i ) overload, indicative of disrupted cellular Ca2+ -homeostasis. Interestingly, increased [Ca2+ ]i coincided with decreased phosphorylation of phospholamban (PLB) and contractility. Aged-cardiomyocytes also displayed high Na+ /Ca2+ -exchanger (NCX) activity and blood glucose levels compared with young-controls. Interestingly, the protein level of SGLT2 was dramatically increased in the aged cardiomyocytes. Moreover, SGLT2 inhibition was sufficient to restore age-associated defects in [Ca2+ ]i -homeostasis, PLB phosphorylation, NCX activity and mitochondrial Ca2+ -loading. Hence, the present data suggest that deregulated SGLT2 during ageing disrupts mitochondrial function and cardiac contractility through a mechanism that impinges upon [Ca2+ ]i -homeostasis. Our studies support the notion that interventions that modulate SGLT2-activity can provide benefits in maintaining [Ca2+ ]i and cardiac function with advanced age.


Asunto(s)
Envejecimiento , Calcio/metabolismo , Mitocondrias Cardíacas/metabolismo , Retículo Sarcoplasmático/metabolismo , Transportador 2 de Sodio-Glucosa/genética , Disfunción Ventricular/etiología , Disfunción Ventricular/metabolismo , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Señalización del Calcio , Senescencia Celular , Susceptibilidad a Enfermedades , Homeostasis , Masculino , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Disfunción Ventricular/fisiopatología
13.
J Cell Mol Med ; 24(12): 6510-6522, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32383522

RESUMEN

Redox signalling in mitochondria plays an important role in myocardial ischaemia/reperfusion (I/R) injury and in cardioprotection. Reactive oxygen and nitrogen species (ROS/RNS) modify cellular structures and functions by means of covalent changes in proteins including among others S-nitros(yl)ation by nitric oxide (NO) and its derivatives, and S-sulphydration by hydrogen sulphide (H2 S). Many enzymes are involved in the mitochondrial formation and handling of ROS, NO and H2 S under physiological and pathological conditions. In particular, the balance between formation and removal of reactive species is impaired during I/R favouring their accumulation. Therefore, various interventions aimed at decreasing mitochondrial ROS accumulation have been developed and have shown cardioprotective effects in experimental settings. However, ROS, NO and H2 S play also a role in endogenous cardioprotection, as in the case of ischaemic pre-conditioning, so that preventing their increase might hamper self-defence mechanisms. The aim of the present review was to provide a critical analysis of formation and role of reactive species, NO and H2 S in mitochondria, with a special emphasis on mechanisms of injury and protection that determine the fate of hearts subjected to I/R. The elucidation of the signalling pathways of ROS, NO and H2 S is likely to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent I/R injury.


Asunto(s)
Cardiotónicos/uso terapéutico , Sulfuro de Hidrógeno/metabolismo , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos
14.
Mol Cell Biochem ; 469(1-2): 97-107, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32301059

RESUMEN

Ticagrelor, a P2Y12-receptor inhibitor, and a non-thienopyridine agent are used to treat diabetic patients via its effects on off-target mechanisms. However, the exact sub-cellular mechanisms by which ticagrelor exerts those effects remains to be elucidated. Accordingly, the present study aimed to examine whether ticagrelor influences directly the cardiomyocytes function under insulin resistance through affecting mitochondria-sarco(endo)plasmic reticulum (SER) cross-talk. Therefore, we analyzed the function and ultrastructure of mitochondria and SER in insulin resistance-mimicked (50-µM palmitic acid for 24-h) H9c2 cardiomyocytes in the presence or absence of ticagrelor (1-µM for 24-h). We found that ticagrelor treatment significantly prevented depolarization of mitochondrial membrane potential and increases in reactive oxygen species with a marked increase in the ATP level in insulin-resistant H9c2 cells. Ticagrelor treatment also reversed the increases in the resting level of free Ca2+ and mRNA level of P2Y12 receptors as well as preserved ER stress and apoptosis in insulin-resistant H9c2 cells. Furthermore, we determined marked repression with ticagrelor treatment in the increased number of autophagosomes and degeneration of mitochondrion, including swelling and loss of crista besides recoveries in enlargement and irregularity seen in SER in insulin-resistant H9c2 cells. Moreover, ticagrelor treatment could prevent the altered mRNA levels of Becklin-1 and type 1 equilibrative nucleoside transporter (ENT1), which are parallel to the preservation of ultrastructural ones. Our overall data demonstrated that ticagrelor can directly affect cardiomyocytes and provide marked protection against ER stress and dramatic induction of autophagosomes, and therefore, can alleviate the ER stress-induced oxidative stress increase and cell apoptosis during insulin resistance.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagosomas/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ticagrelor/farmacología , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Beclina-1/genética , Beclina-1/metabolismo , Calcio/metabolismo , Línea Celular , Tranportador Equilibrativo 1 de Nucleósido/genética , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Insulina/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Ácido Palmítico/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2Y12/genética , Receptores Purinérgicos P2Y12/metabolismo
15.
Mol Cell Biochem ; 463(1-2): 33-44, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31520233

RESUMEN

Excitation-contraction coupling in normal cardiac function is performed with well balanced and coordinated functioning but with complex dynamic interactions between functionally connected membrane ionic currents. However, their genomic investigations provide essential information on the regulation of diseases by their transcripts. Therefore, we examined the gene expression levels of the most important voltage-gated ionic channels such as Na+-channels (SCN5A), Ca2+-channels (CACNA1C and CACNA1H), and K+-channels, including transient outward (KCND2, KCNA2, KCNA5, KCNA8), inward rectifier (KCNJ2, KCNJ12, KCNJ4), and delayed rectifier (KCNB1) in left ventricular tissues from either ischemic or dilated cardiomyopathy (ICM or DCM). We also examined the mRNA levels of ATP-dependent K+-channels (KCNJ11, ABCC9) and ERG-family channels (KCNH2). We further determined the mRNA levels of ryanodine receptors (RyR2; ARVC2), phospholamban (PLB or PLN), SR Ca2+-pump (SERCA2; ATP2A1), an accessory protein FKBP12 (PPIASE), protein kinase A (PPNAD4), and Ca2+/calmodulin-dependent protein kinase II (CAMK2G). The mRNA levels of SCN5A, CACNA1C, and CACNA1H in both groups decreased markedly in the heart samples with similar significance, while KvLQT1 genes were high with depressed Kv4.2. The KCNJ11 and KCNJ12 in both groups were depressed, while the KCNJ4 level was significantly high. More importantly, the KCNA5 gene was downregulated only in the ICM, while the KCNJ2 was upregulated only in the DCM. Besides, mRNA levels of ARVC2 and PLB were significantly high compared to the controls, whereas others (ATP2A1, PPIASE, PPNAD4, and CAMK2G) were decreased. Importantly, the increases of KCNB1 and KCNJ11 were more prominent in the ICM than DCM, while the decreases in ATP2A1 and FKBP1A were more prominent in DCM compared to ICM. Overall, this study was the first to demonstrate that the different levels of changes in gene profiles via different types of cardiomyopathy are prominent particularly in some K+-channels, which provide further information about our knowledge of how remodeling processes can be differentiated in HF originated from different pathological conditions.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Canales Iónicos/biosíntesis , Miocitos Cardíacos/metabolismo , Anciano , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Femenino , Insuficiencia Cardíaca/patología , Humanos , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/patología
16.
Cardiovasc Drugs Ther ; 34(4): 487-501, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32377826

RESUMEN

BACKGROUND: Previous studies have demonstrated that a high-carbohydrate intake could induce metabolic syndrome (MetS) in male rats with marked cardiac functional abnormalities. In addition, studies mentioned some benefits of insulin application on these complications, but there are considerable disagreements among their findings. Therefore, we aimed to extend our knowledge on the in-vitro influence of insulin on left ventricular dysfunction and also in the isolated cardiomyocytes from MetS rats. RESULTS: At the organ function level, an acute insulin application (100-nM) provided an important beneficial effect on the left ventricular developed pressure in MetS rats. Furthermore, to treat the freshly isolated cardiomyocytes from MetS rats with insulin provided marked recoveries in elevated resting intracellular Ca2+-level, as well as significant prevention of prolonged action potential through an augmentation in depressed K+-channel currents. Insulin also normalized the cellular levels of increased ROS and phosphorylation of PKCα, together with normalizations of apoptotic markers in MetS cardiomyocytes through the insulin-mediated regulation of phospho-Akt. Since not only elevated PKCα-activity but also reductions in phospho-Akt are key modulators of titin-based cardiomyocyte stiffening in hyperglycemia, insulin treatment of the cardiomyocytes prevented the activation of titin via the above pathways. Furthermore, CK2α-activation and NOS-phosphorylation could be prevented with insulin treatment. Mechanistically, we found that impaired insulin signaling and elevated PKCα and CK2α activities, as well as depressed Akt phosphorylation, are key modulators of titin-based cardiomyocyte stiffening in MetS rats. CONCLUSION: We propose that restoring normal kinase activities and also increases in phospho-Akt by insulin can contribute marked recoveries in MetS heart function, indicating a promising approach to modulate titin-associated factors in heart dysfunction associated with type-2 diabetes mellitus. Graphical Abstract.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Conectina/metabolismo , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Insulina/farmacología , Síndrome Metabólico/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Disfunción Ventricular Izquierda/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Modelos Animales de Enfermedad , Preparación de Corazón Aislado , Masculino , Síndrome Metabólico/enzimología , Síndrome Metabólico/fisiopatología , Miocitos Cardíacos/enzimología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Wistar , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/fisiopatología , Presión Ventricular/efectos de los fármacos
17.
J Cell Physiol ; 234(8): 13370-13386, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30613975

RESUMEN

Role of ß3 -AR dysregulation, as either cardio-conserving or cardio-disrupting mediator, remains unknown yet. Therefore, we examined the molecular mechanism of ß3 -AR activation in depressed myocardial contractility using a specific agonist CL316243 or using ß3 -AR overexpressed cardiomyocytes. Since it has been previously shown a possible correlation between increased cellular free Zn2+ ([Zn2+ ]i ) and depressed cardiac contractility, we first demonstrated a relation between ß3 -AR activation and increased [Zn2+ ]i , parallel to the significant depolarization in mitochondrial membrane potential in rat ventricular cardiomyocytes. Furthermore, the increased [Zn2+ ]i induced a significant increase in messenger RNA (mRNA) level of ß3 -AR in cardiomyocytes. Either ß3 -AR activation or its overexpression could increase cellular reactive oxygen species (ROS) and reactive nitrogen species (RNS) levels, in line with significant changes in nitric oxide (NO)-pathway, including increases in the ratios of pNOS3/NOS3 and pGSK-3ß/GSK-3ß, and PKG expression level in cardiomyocytes. Although ß3 -AR activation induced depression in both Na+ - and Ca2+ -currents, the prolonged action potential (AP) seems to be associated with a marked depression in K+ -currents. The ß3 -AR activation caused a negative inotropic effect on the mechanical activity of the heart, through affecting the cellular Ca2+ -handling, including its effect on Ca2+ -leakage from sarcoplasmic reticulum (SR). Our cellular level data with ß3 -AR agonism were supported with the data on high [Zn2+ ]i and ß3 -AR protein-level in metabolic syndrome (MetS)-rat heart. Overall, our present data can emphasize the important deleterious effect of ß3 -AR activation in cardiac remodeling under pathological condition, at least, through a cross-link between ß3 -AR activation, NO-signaling, and [Zn2+ ]i pathways. Moreover, it is interesting to note that the recovery in ER-stress markers with ß3 -AR agonism in hyperglycemic cardiomyocytes is favored. Therefore, how long and to which level the ß3 -AR agonism would be friend or become foe remains to be mystery, yet.


Asunto(s)
Contracción Miocárdica/fisiología , Especies de Nitrógeno Reactivo/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Zinc/metabolismo , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Señalización del Calcio , Línea Celular , Dioxoles/farmacología , Masculino , Potencial de la Membrana Mitocondrial , Síndrome Metabólico/metabolismo , Modelos Cardiovasculares , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Receptores Adrenérgicos beta 3/genética
18.
Mol Cell Biochem ; 461(1-2): 65-72, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31327095

RESUMEN

Azoramide is identified as a new compound with the dual properties for the improvement of ER-folding capacity in various cells as well as for the treatment of T2DM. Although the effect of azoramide in glucose-homeostasis in mammalians is not known very well, a limited number of experimental studies showed that it could improve the insulin sensitivity in genetically obese mice. Therefore, here, we aimed to investigate the direct effect of azoramide on insulin signaling in insulin-resistant (IR) cardiomyocytes using IR-modelled ventricular cardiomyocytes. This model was established in H9c2 cells using palmitic acid incubation (50-µM for 24-h). The development of IR in cells was verified by monitoring the cellular 2-DG6P uptake assays in these treated cells. The 2-DG6P uptake was 50% less in the IR-cells compared to the control cells, while azoramide treatment (20-µM for 48-h) could prevent fully that decrease. In addition, azoramide treatment markedly preserved the IR-induced less ATP production and high-ROS production in these IR-cells. Furthermore, this treatment prevented the functional changes in mitochondria characterized by depolarized mitochondrial membrane potential and mitochondrial fusion or fusion-related protein levels as well as cellular ATP level. Moreover, this treatment provided marked protection against IR-associated changes in the insulin signaling pathway in cells, including recovery in the phosphorylation of IRS1 and Akt as well as the protein level of GLUT4 and Akt. Our present results, for the first time, demonstrated that azoramide plays an important protective role in IR-cardiomyocytes, at most, protective action on mitochondria. Therefore, one can suggest that azoramide, as a novel regulator, can provide direct cardioprotection in the IR-heart, at most, via affecting mitochondria and can be a good candidate as a new drug for the treatment of IR-associated cardiovascular disorders in mammalians with systemic IR.


Asunto(s)
Amidas/farmacología , Resistencia a la Insulina , Mitocondrias/metabolismo , Ácido Palmítico/toxicidad , Tiazoles/farmacología , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Glucosa/metabolismo , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Can J Physiol Pharmacol ; 97(6): 528-535, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30444646

RESUMEN

Sodium-glucose cotransporter 2 (SGLT2) inhibitors showed significant effects in patients with diabetes or metabolic syndrome (MetS) with high cardiovascular risk. Although the increased intracellular Zn2+ level ([Zn2+]i), oxidative stress, and altered cardiac matrix metalloproteinases (MMPs) in diabetic cardiomyopathy can intersect with different signaling pathways, the exact mechanisms are not known yet. Since either MMPs or SGLT2 have important roles in cardiac-fibrosis under hyperglycemia, we aimed to examine the role of SGLT2 inhibitor dapagliflozin (DAP) on cardiac Zn2+-transporters responsible for [Zn2+]i-regulation, comparison to insulin (INS), together with MMP levels and systemic oxidative stress status in MetS-rats. High-carbohydrated diet-induced MetS-rats received DAP or INS for 2 weeks. DAP but not INS in MetS-rats significantly decreased high blood-glucose levels, while both treatments exerted benefits on increased total oxidative status and decreased total antioxidant status in MetS-rat plasma as well as in heart tissue. Protein levels of Zn2+-transporters, responsible for Zn2+-influx into cytosol, ZIP7 and ZIP14 were increased with significant decrease in ZIP8 of MetS-rat cardiomyoctes, while Zn2+-transporters, responsible for cytosolic Zn2+-efflux, ZnT7 was decreased with no change in ZnT8. Both treatments induced significant beneficial effects on altered ZIP14, ZIP8, and ZnT7 levels. Furthermore, both treatments exerted benefits on depressed gelatin-zymography and protein expression levels of MMP-2 and MMP-9 in MetS-rat ventricular cardiomyocytes. The direct effect of DAP on heart was also confirmed with measurements of left ventricular developed pressure. Overall, we showed that DAP has important antioxidant-like cardio-protective effects in MetS-rats, similar to INS-effect, affecting Zn2+-regulation via Zn2+-transporters, MMPs, and oxidative stress. Therefore one can suggest that SGLT2 inhibitors can be new therapeutic agents for cardio-protection not only in hyperglycemia but also in failing heart.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Glucósidos/farmacología , Insulina/farmacología , Síndrome Metabólico/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transportador 2 de Sodio-Glucosa/metabolismo , Zinc/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Resistencia a la Insulina , Masculino , Metaloproteinasas de la Matriz/metabolismo , Síndrome Metabólico/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteolisis/efectos de los fármacos , Ratas , Ratas Wistar , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
20.
Can J Physiol Pharmacol ; 97(11): 1064-1072, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31299169

RESUMEN

Metabolic syndrome (MetS) is a cluster of risk factors, including insulin resistance among others, underlying the development of diabetes and (or) cardiovascular diseases. Studies show a close relationship between cardiac dysfunction and abnormal cAMP catabolism, which contributes to pathological remodelling. Stimulating the synthesis of cAMP via suppression of phosphodiesterases (PDEs) has positive therapeutic effects. Therefore, we examined the role of PDEs on cardiac dysfunction in high-carbohydrate diet-induced MetS rats. We first demonstrated significantly high expression levels of PDE3 and PDE4, the most highly expressed subtypes, together with depressed cAMP levels in heart tissue from MetS rats. Second, we demonstrated the activity of these PDEs by using either their basal or PDE inhibitor-induced intracellular levels of cAMP and Ca2+, the transient intracellular Ca2+ changes under electrical stimulation, isometric contractions in papillary muscle strips and some key signalling proteins (such as RyR2, PLN, PP1A, and PKA) are responsible for the Ca2+ homeostasis in isolated cardiomyocytes from MetS rats. The clear recovery in decreased basal cAMP levels, increased protein expression levels of PDE3 and PDE4, and positive responses in the altered Ca2+ homeostasis to PDE inhibitors as seen in our study can provide important insights about the roles of activated PDEs in depressed contractile activity in hearts from MetS rats.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Carbohidratos de la Dieta/efectos adversos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/fisiopatología , Animales , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Síndrome Metabólico/enzimología , Contracción Muscular/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Ratas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda