Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Basic Res Cardiol ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724619

RESUMEN

Succinate accumulates during myocardial ischemia and is rapidly oxidized during reperfusion, leading to reactive oxygen species (ROS) production through reverse electron transfer (RET) from mitochondrial complex II to complex I, and favoring cell death. Given that connexin 43 (Cx43) modulates mitochondrial ROS production, we investigated whether Cx43 influences RET using inducible knock-out Cx43Cre-ER(T)/fl mice. Oxygen consumption, ROS production, membrane potential and coenzyme Q (CoQ) pool were analyzed in subsarcolemmal (SSM, expressing Cx43) and interfibrillar (IFM) cardiac mitochondria isolated from wild-type Cx43fl/fl mice and Cx43Cre-ER(T)/fl knock-out animals treated with 4-hydroxytamoxifen (4OHT). In addition, infarct size was assessed in isolated hearts from these animals submitted to ischemia-reperfusion (IR), and treated or not with malonate, a complex II inhibitor attenuating RET. Succinate-dependent ROS production and RET were significantly lower in SSM, but not IFM, from Cx43-deficient animals. Mitochondrial membrane potential, a RET driver, was similar between groups, whereas CoQ pool (2.165 ± 0.338 vs. 4.18 ± 0.55 nmol/mg protein, p < 0.05) and its reduction state were significantly lower in Cx43-deficient animals. Isolated hearts from Cx43Cre-ER(T)/fl mice treated with 4OHT had a smaller infarct size after IR compared to Cx43fl/fl, despite similar concentration of succinate at the end of ischemia, and no additional protection by malonate. Cx43 deficiency attenuates ROS production by RET in SSM, but not IFM, and was associated with a decrease in CoQ levels and a change in its redox state. These results may partially explain the reduced infarct size observed in these animals and their lack of protection by malonate.

2.
Int J Mol Sci ; 25(3)2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38338818

RESUMEN

TRPV4 channels, which respond to mechanical activation by permeating Ca2+ into the cell, may play a pivotal role in cardiac remodeling during cardiac overload. Our study aimed to investigate TRPV4 involvement in pathological and physiological remodeling through Ca2+-dependent signaling. TRPV4 expression was assessed in heart failure (HF) models, induced by isoproterenol infusion or transverse aortic constriction, and in exercise-induced adaptive remodeling models. The impact of genetic TRPV4 inhibition on HF was studied by echocardiography, histology, gene and protein analysis, arrhythmia inducibility, Ca2+ dynamics, calcineurin (CN) activity, and NFAT nuclear translocation. TRPV4 expression exclusively increased in HF models, strongly correlating with fibrosis. Isoproterenol-administered transgenic TRPV4-/- mice did not exhibit HF features. Cardiac fibroblasts (CFb) from TRPV4+/+ animals, compared to TRPV4-/-, displayed significant TRPV4 overexpression, elevated Ca2+ influx, and enhanced CN/NFATc3 pathway activation. TRPC6 expression paralleled that of TRPV4 in all models, with no increase in TRPV4-/- mice. In cultured CFb, the activation of TRPV4 by GSK1016790A increased TRPC6 expression, which led to enhanced CN/NFATc3 activation through synergistic action of both channels. In conclusion, TRPV4 channels contribute to pathological remodeling by promoting fibrosis and inducing TRPC6 upregulation through the activation of Ca2+-dependent CN/NFATc3 signaling. These results pose TRPV4 as a primary mediator of the pathological response.


Asunto(s)
Calcineurina , Insuficiencia Cardíaca , Canales Catiónicos TRPV , Remodelación Ventricular , Animales , Ratones , Calcineurina/metabolismo , Células Cultivadas , Fibrosis , Insuficiencia Cardíaca/metabolismo , Isoproterenol , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Remodelación Ventricular/genética
3.
Int J Mol Sci ; 25(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38673951

RESUMEN

Succinate dehydrogenase inhibition with malonate during initial reperfusion reduces myocardial infarct size in both isolated mouse hearts subjected to global ischemia and in in situ pig hearts subjected to transient coronary ligature. However, the long-term effects of acute malonate treatment are unknown. Here, we investigated whether the protective effects of succinate dehydrogenase inhibition extend to a reduction in scar size and adverse left ventricular remodeling 28 days after myocardial infarction. Initially, ten wild-type mice were subjected to 45 min of left anterior descending coronary artery (LAD) occlusion, followed by 24 h of reperfusion, and were infused during the first 15 min of reperfusion with saline with or without disodium malonate (10 mg/kg/min, 120 µL/kg/min). Malonate-treated mice depicted a significant reduction in infarct size (15.47 ± 3.40% of area at risk vs. 29.34 ± 4.44% in control animals, p < 0.05), assessed using triphenyltetrazolium chloride. Additional animals were then subjected to a 45 min LAD ligature, followed by 28 days of reperfusion. Treatment with a single dose of malonate during the first 15 min of reperfusion induced a significant reduction in scar area, measured using Picrosirius Red staining (11.94 ± 1.70% of left ventricular area (n = 5) vs. 23.25 ± 2.67% (n = 9), p < 0.05), an effect associated with improved ejection fraction 28 days after infarction, as determined using echocardiography, and an attenuated enhancement in expression of the pro-inflammatory and fibrotic markers NF-κB and Smad2/3 in remote myocardium. In conclusion, a reversible inhibition of succinate dehydrogenase with a single dose of malonate at the onset of reperfusion has long-term protective effects in mice subjected to transient coronary occlusion.


Asunto(s)
Malonatos , Infarto del Miocardio , Daño por Reperfusión Miocárdica , Succinato Deshidrogenasa , Remodelación Ventricular , Animales , Malonatos/farmacología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Ratones , Succinato Deshidrogenasa/metabolismo , Succinato Deshidrogenasa/antagonistas & inhibidores , Masculino , Remodelación Ventricular/efectos de los fármacos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Cicatriz/patología , Cicatriz/tratamiento farmacológico , Ratones Endogámicos C57BL
4.
Crit Care ; 27(1): 374, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773186

RESUMEN

BACKGROUND AND AIMS: The triggering factors of sepsis-induced myocardial dysfunction (SIMD) are poorly understood and are not addressed by current treatments. S100A8/A9 is a pro-inflammatory alarmin abundantly secreted by activated neutrophils during infection and inflammation. We investigated the efficacy of S100A8/A9 blockade as a potential new treatment in SIMD. METHODS: The relationship between plasma S100A8/A9 and cardiac dysfunction was assessed in a cohort of 62 patients with severe sepsis admitted to the intensive care unit of Linköping University Hospital, Sweden. We used S100A8/A9 blockade with the small-molecule inhibitor ABR-238901 and S100A9-/- mice for therapeutic and mechanistic studies on endotoxemia-induced cardiac dysfunction in mice. RESULTS: In sepsis patients, elevated plasma S100A8/A9 was associated with left-ventricular (LV) systolic dysfunction and increased SOFA score. In wild-type mice, 5 mg/kg of bacterial lipopolysaccharide (LPS) induced rapid plasma S100A8/A9 increase and acute LV dysfunction. Two ABR-238901 doses (30 mg/kg) administered intraperitoneally with a 6 h interval, starting directly after LPS or at a later time-point when LV dysfunction is fully established, efficiently prevented and reversed the phenotype, respectively. In contrast, dexamethasone did not improve cardiac function compared to PBS-treated endotoxemic controls. S100A8/A9 inhibition potently reduced systemic levels of inflammatory mediators, prevented upregulation of inflammatory genes and restored mitochondrial function in the myocardium. The S100A9-/- mice were protected against LPS-induced LV dysfunction to an extent comparable with pharmacologic S100A8/A9 blockade. The ABR-238901 treatment did not induce an additional improvement of LV function in the S100A9-/- mice, confirming target specificity. CONCLUSION: Elevated S100A8/A9 is associated with the development of LV dysfunction in severe sepsis patients and in a mouse model of endotoxemia. Pharmacological blockade of S100A8/A9 with ABR-238901 has potent anti-inflammatory effects, mitigates myocardial dysfunction and might represent a novel therapeutic strategy for patients with severe sepsis.


Asunto(s)
Endotoxemia , Cardiopatías , Disfunción Ventricular Izquierda , Humanos , Ratones , Animales , Endotoxemia/complicaciones , Endotoxemia/tratamiento farmacológico , Lipopolisacáridos , Calgranulina A/fisiología , Calgranulina B/genética , Miocardio , Inflamación/tratamiento farmacológico
5.
Basic Res Cardiol ; 116(1): 4, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495853

RESUMEN

Remote ischemic conditioning (RIC) and the GLP-1 analog exenatide activate different cardioprotective pathways and may have additive effects on infarct size (IS). Here, we aimed to assess the efficacy of RIC as compared with sham procedure, and of exenatide, as compared with placebo, and the interaction between both, to reduce IS in humans. We designed a two-by-two factorial, randomized controlled, blinded, multicenter, clinical trial. Patients with ST-segment elevation myocardial infarction receiving primary percutaneous coronary intervention (PPCI) within 6 h of symptoms were randomized to RIC or sham procedure and exenatide or matching placebo. The primary outcome was IS measured by late gadolinium enhancement in cardiac magnetic resonance performed 3-7 days after PPCI. The secondary outcomes were myocardial salvage index, transmurality index, left ventricular ejection fraction and relative microvascular obstruction volume. A total of 378 patients were randomly allocated, and after applying exclusion criteria, 222 patients were available for analysis. There were no significant interactions between the two randomization factors on the primary or secondary outcomes. IS was similar between groups for the RIC (24 ± 11.8% in the RIC group vs 23.7 ± 10.9% in the sham group, P = 0.827) and the exenatide hypotheses (25.1 ± 11.5% in the exenatide group vs 22.5 ± 10.9% in the placebo group, P = 0.092). There were no effects with either RIC or exenatide on the secondary outcomes. Unexpected adverse events or side effects of RIC and exenatide were not observed. In conclusion, neither RIC nor exenatide, or its combination, were able to reduce IS in STEMI patients when administered as an adjunct to PPCI.


Asunto(s)
Brazo/irrigación sanguínea , Exenatida/uso terapéutico , Incretinas/uso terapéutico , Precondicionamiento Isquémico , Miocardio/patología , Intervención Coronaria Percutánea , Infarto del Miocardio con Elevación del ST/terapia , Anciano , Terapia Combinada , Método Doble Ciego , Exenatida/efectos adversos , Femenino , Humanos , Incretinas/efectos adversos , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Intervención Coronaria Percutánea/efectos adversos , Estudios Prospectivos , Flujo Sanguíneo Regional , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Infarto del Miocardio con Elevación del ST/patología , Infarto del Miocardio con Elevación del ST/fisiopatología , España , Factores de Tiempo , Resultado del Tratamiento , Función Ventricular Izquierda
6.
Int J Mol Sci ; 22(9)2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33922534

RESUMEN

Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.


Asunto(s)
Conexinas/metabolismo , Cardiopatías/fisiopatología , Animales , Cardiopatías/metabolismo , Humanos
7.
Int J Mol Sci ; 22(21)2021 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-34768801

RESUMEN

The mechanisms committed in the activation and response of vascular and inflammatory immune cells play a major role in tissue remodeling in cardiovascular diseases (CVDs) such as atherosclerosis, pulmonary arterial hypertension, and abdominal aortic aneurysm. Cardiovascular remodeling entails interrelated cellular processes (proliferation, survival/apoptosis, inflammation, extracellular matrix (ECM) synthesis/degradation, redox homeostasis, etc.) coordinately regulated by a reduced number of transcription factors. Nuclear receptors of the subfamily 4 group A (NR4A) have recently emerged as key master genes in multiple cellular processes and vital functions of different organs, and have been involved in a variety of high-incidence human pathologies including atherosclerosis and other CVDs. This paper reviews the major findings involving NR4A3 (Neuron-derived Orphan Receptor 1, NOR-1) in the cardiovascular remodeling operating in these diseases.


Asunto(s)
Enfermedades Cardiovasculares/patología , Sistema Cardiovascular/patología , Proteínas de Unión al ADN/metabolismo , Inflamación , Proteínas del Tejido Nervioso/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Animales , Aterosclerosis , Remodelación Atrial , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Proteínas de Unión al ADN/fisiología , Humanos , Proteínas del Tejido Nervioso/fisiología , Hipertensión Arterial Pulmonar , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología
8.
Int J Mol Sci ; 22(8)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923786

RESUMEN

Succinate dehydrogenase (SDH) inhibition with malonate during reperfusion reduced myocardial infarction in animals, whereas its endogenous substrate, succinate, is detected in plasma from STEMI patients. We investigated whether protection by SDH inhibition is additive to that of remote ischemic perconditioning (RIC) in pigs submitted to transient coronary artery occlusion, and whether protective maneuvers influence plasma levels of citric acid cycle metabolites. Forty pigs were submitted to 40 min coronary occlusion and reperfusion, and allocated to four groups (controls, sodium malonate 10 mmol/L, RIC, and malonate + RIC). Plasma was obtained from femoral and great cardiac veins and analyzed by LC-MS/MS. Malonate, RIC, and malonate + RIC reduced infarct size (24.67 ± 5.98, 25.29 ± 3.92 and 29.83 ± 4.62% vs. 46.47 ± 4.49% in controls, p < 0.05), but no additive effects were detected. Enhanced concentrations of succinate, fumarate, malate and citrate were observed in controls during initial reperfusion in the great cardiac vein, and most were reduced by cardioprotective maneuvers. Concentrations of succinate, fumarate, and malate significantly correlated with infarct size. In conclusion, despite the combination of SDH inhibition during reperfusion and RIC did not result in additive protection, plasma concentrations of selected citric acid cycle metabolites are attenuated by protective maneuvers, correlate with irreversible injury, and might become a prognosis tool in STEMI patients.


Asunto(s)
Ciclo del Ácido Cítrico , Oclusión Coronaria/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Precondicionamiento Isquémico/métodos , Infarto del Miocardio/metabolismo , Succinato Deshidrogenasa/antagonistas & inhibidores , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Oclusión Coronaria/patología , Oclusión Coronaria/terapia , Ácidos Dicarboxílicos/sangre , Ácidos Dicarboxílicos/metabolismo , Inhibidores Enzimáticos/farmacología , Corazón/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Porcinos
9.
Int J Mol Sci ; 23(1)2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-35008601

RESUMEN

Information about heart failure with reduced ejection fraction (HFrEF) in women and the potential effects of aging in the female heart is scarce. We investigated the vulnerability to develop HFrEF in female elderly mice compared to young animals, as well as potential differences in reverse remodeling. First, HF was induced by isoproterenol infusion (30 mg/kg/day, 28 days) in young (10-week-old) and elderly (22-month-old) female mice. In a second set of animals, mice underwent isoproterenol infusion followed by no treatment during 28 additional days. Cardiac remodeling was assessed by echocardiography, histology and gene expression of collagen-I and collagen-III. Following isoproterenol infusion, elderly mice developed similar HFrEF features compared to young animals, except for greater cell hypertrophy and tissue fibrosis. After beta-adrenergic withdrawal, young female mice experienced complete reversal of the HFrEF phenotype. Conversely, reversed remodeling was impaired in elderly animals, with no significant recovery of LV ejection fraction, cardiomyocyte hypertrophy and collagen deposition. In conclusion, chronic isoproterenol infusion is a valid HF model for elderly and young female mice and induces a similar HF phenotype in both. Elderly animals, unlike young, show impaired reverse remodeling, with persistent tissue fibrosis and cardiac dysfunction even after beta-adrenergic withdrawal.


Asunto(s)
Envejecimiento , Modelos Animales de Enfermedad , Fibrosis , Insuficiencia Cardíaca/inducido químicamente , Isoproterenol/toxicidad , Animales , Cardiomiopatías , Colágeno/genética , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/fisiopatología , Ratones , Ratones Endogámicos C57BL , Volumen Sistólico , Función Ventricular Izquierda , Remodelación Ventricular
10.
Int J Mol Sci ; 20(23)2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31775376

RESUMEN

Ischemic postconditioning (IPoC) reduces reperfusion arrhythmias but the antiarrhythmic mechanisms remain unknown. The aim of this study was to analyze IPoC electrophysiological effects and the role played by adenosine A1, A2A and A3 receptors, protein kinase C, ATP-dependent potassium (KATP) channels, and connexin 43. IPoC reduced reperfusion arrhythmias (mainly sustained ventricular fibrillation) in isolated rat hearts, an effect associated with a transient delay in epicardial electrical activation, and with action potential shortening. Electrical impedance measurements and Lucifer-Yellow diffusion assays agreed with such activation delay. However, this delay persisted during IPoC in isolated mouse hearts in which connexin 43 was replaced by connexin 32 and in mice with conditional deletion of connexin 43. Adenosine A1, A2A and A3 receptor blockade antagonized the antiarrhythmic effect of IPoC and the associated action potential shortening, whereas exogenous adenosine reduced reperfusion arrhythmias and shortened action potential duration. Protein kinase C inhibition by chelerythrine abolished the protective effect of IPoC but did not modify the effects on action potential duration. On the other hand, glibenclamide, a KATP inhibitor, antagonized the action potential shortening but did not interfere with the antiarrhythmic effect. The antiarrhythmic mechanisms of IPoC involve adenosine receptor activation and are associated with action potential shortening. However, this action potential shortening is not essential for protection, as it persisted during protein kinase C inhibition, a maneuver that abolished IPoC protection. Furthermore, glibenclamide induced the opposite effects. In addition, IPoC delays electrical activation and electrical impedance recovery during reperfusion, but these effects are independent of connexin 43.


Asunto(s)
Arritmias Cardíacas/prevención & control , Conexina 43/fisiología , Poscondicionamiento Isquémico/métodos , Canales KATP/metabolismo , Isquemia Miocárdica/complicaciones , Proteína Quinasa C/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Canales KATP/genética , Ratones , Ratones Transgénicos , Proteína Quinasa C/genética , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P1/genética
11.
Biochim Biophys Acta Biomembr ; 1860(1): 174-181, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28642043

RESUMEN

Connexin 43 (Cx43) forms gap junction channels that are essential for the propagation of electrical depolarization in cardiomyocytes, but also with important roles in the pathophysiology of reperfusion injury. However, more recent studies have shown that Cx43 has also important functions independent from intercellular communication between adjacent cardiomyocytes. Some of these actions have been related to the presence of Cx43 in the mitochondria of these cells (mitoCx43). The functions of mitoCx43 have not been completely elucidated, but there is strong evidence indicating that mitoCx43 modulates mitochondrial respiration at respiratory complex I, production of radical oxygen species and ATP synthesis. These functions of mitoCx43 modulate mitochondrial and cellular tolerance to reperfusion after prolonged ischemia and are necessary for the cardioprotective effect of ischemic preconditioning. In the present review article we discuss available knowledge on these functions of mitoCx43 in relation to reperfusion injury, the molecular mechanisms involved and explore the possibility that mitoCx43 may constitute a new pharmacological target in patients with ST-segment elevation myocardial infarction (STEMI). This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.


Asunto(s)
Conexina 43/metabolismo , Precondicionamiento Isquémico Miocárdico , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Infarto del Miocardio con Elevación del ST/metabolismo , Animales , Complejo I de Transporte de Electrón/metabolismo , Humanos , Mitocondrias Cardíacas/patología , Daño por Reperfusión Miocárdica/patología , Infarto del Miocardio con Elevación del ST/patología
12.
FASEB J ; 31(9): 3787-3799, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28522596

RESUMEN

Lysyl oxidase (LOX) controls matrix remodeling, a key process that underlies cardiovascular diseases and heart failure; however, a lack of suitable animal models has limited our knowledge with regard to the contribution of LOX to cardiac dysfunction. Here, we assessed the impact of LOX overexpression on ventricular function and cardiac hypertrophy in a transgenic LOX (TgLOX) mouse model with a strong cardiac expression of human LOX. TgLOX mice exhibited high expression of the transgene in cardiomyocytes and cardiofibroblasts, which are associated with enhanced LOX activity and H2O2 production and with cardiofibroblast reprogramming. LOX overexpression promoted an age-associated concentric remodeling of the left ventricle and impaired diastolic function. Furthermore, LOX transgenesis aggravated angiotensin II (Ang II)-induced cardiac hypertrophy and dysfunction, which triggered a greater fibrotic response that was characterized by stronger collagen deposition and cross-linking and high expression of fibrotic markers. In addition, LOX transgenesis increased the Ang II-induced myocardial inflammatory infiltrate, exacerbated expression of proinflammatory markers, and decreased that of cardioprotective factors. Mechanistically, LOX overexpression enhanced oxidative stress and potentiated the Ang II-mediated cardiac activation of p38 MAPK while reducing AMPK activation. Our findings suggest that LOX induces an age-dependent disturbance of diastolic function and aggravates Ang II-induced hypertrophy, which provides novel insights into the role of LOX in cardiac performance.-Galán, M., Varona, S., Guadall, A., Orriols, M., Navas, M., Aguiló, S., de Diego, A., Navarro, M. A., García-Dorado, D., Rodríguez-Sinovas, A., Martínez-González, J., Rodriguez, C. Lysyl oxidase overexpression accelerates cardiac remodeling and aggravates angiotensin II-induced hypertrophy.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/inducido químicamente , Regulación Enzimológica de la Expresión Génica/fisiología , Proteína-Lisina 6-Oxidasa/metabolismo , Remodelación Ventricular/fisiología , Animales , Cardiomegalia/enzimología , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Inflamación , Ratones , Ratones Transgénicos , Miocardio/citología , Proteína-Lisina 6-Oxidasa/genética , Transducción de Señal
13.
Int J Mol Sci ; 18(10)2017 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-29019935

RESUMEN

The use of Doxorubicin (DOXO), a potent antineoplastic agent, is limited by the development of cardiotoxicity. DOXO-induced cardiotoxicity is multifactorial, although alterations in calcium homeostasis, seem to be involved. Since even the Connexin43 (Cx43) plays a pivotal role in these two phenomena, in this study we have analyzed the effects of DOXO on Cx43 expression and localization. Damage caused by anthracyclines on cardiomyocytes is immediate after each injection, in the present study we used a short-term model of DOXO-induced cardiomyopathy. C57BL/6j female mice were randomly divided in groups and injected with DOXO (2 or 10 mg/kg i.p.) for 1-3 or 7 days once every other day. Cardiac function was assessed by Echocardiography. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCAII) and phospholamban (PLB) expression were assessed by Western blot analysis, intracellular [Ca2+] were detected spectrofluorometrically by means of Fura-2 pentakis (acetoxymethyl) ester (FURA-2AM), and Cx43 and pCx43 expression and localization was analyzed by Western blot and confirmed by immunofluorescence analysis. DOXO induces impairment in Ca2+ homeostasis, already evident after a single administration, and affects Cx43 expression and localization. Our data suggest that DOXO-induced alterations in Ca2+ homeostasis causes in the cells the induction of compensatory mechanisms until a certain threshold, above which cardiac injury is triggered.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Calcio/metabolismo , Cardiomiopatías/inducido químicamente , Conexina 43/metabolismo , Doxorrubicina/efectos adversos , Miocitos Cardíacos/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/administración & dosificación , Calcio/análisis , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/metabolismo , Cardiotoxicidad/etiología , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Femenino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Cultivo Primario de Células , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores de Tiempo
14.
J Mol Cell Cardiol ; 84: 154-61, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25953257

RESUMEN

Obesity is a major risk factor for cardiovascular morbidity and mortality. However, some studies suggest that among patients with established cardiovascular disease, obesity is associated with better prognosis, a phenomenon described as the obesity paradox. In this study we tested the hypothesis that obesity with hyperinsulinemia and without hyperglycemia attenuates the impact of transient coronary occlusion on left ventricular remodeling and function. B6D2F1 mice from both genders fed with a high fat diet (HFD) or control diet for 6 months were subjected to 45 min of coronary occlusion and 28 days of reperfusion. Left ventricular dimensions and function were assessed by serial echocardiography, and infarct size was determined by Picrosirius red staining. HFD mice developed obesity with hypercholesterolemia and hyperinsulinemia in the absence of hyperglycemia or hypertension. During the period of feeding, no changes were observed in ventricular mass, volume or function, or in vascular reactivity. HFD attenuated the consequences of transient coronary occlusion as shown by a marked reduction in infarct size (51%, P = 0.021) and cardiac dilation, as well as improved left ventricular function as compared to control diet animals. These effects were associated with enhanced reperfusion injury salvage kinases (RISK) pathway function in HFD hearts shown as increased Akt and GSK3ß phosphorylation. These results demonstrate that dietary obesity without hyperglycemia or hypertension attenuates the impact of ischemia/reperfusion injury in association with increased insulin signaling and RISK activation. This study provides experimental support to the controversial concept of the obesity paradox in humans.


Asunto(s)
Infarto del Miocardio/complicaciones , Infarto del Miocardio/fisiopatología , Obesidad/complicaciones , Obesidad/fisiopatología , Función Ventricular Izquierda , Remodelación Ventricular , Envejecimiento , Animales , Dieta Alta en Grasa , Electrocardiografía , Activación Enzimática , Ratones , Infarto del Miocardio/diagnóstico por imagen , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Proteínas Quinasas/metabolismo , Transducción de Señal , Ultrasonografía
15.
J Mol Cell Cardiol ; 80: 34-44, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25536180

RESUMEN

Recent work has highlighted the role of NR4A receptors in atherosclerosis and inflammation. In vascular smooth muscle cell (VSMC) proliferation, however, NOR-1 (neuron-derived orphan receptor-1) exerts antagonistic effects to Nur77 and Nurr1. The aim of this study was to analyse the effect of NOR-1 in VSMC inflammatory response. We assessed the consequence of a gain-of-function of this receptor on the response of VSMC to inflammatory stimuli. In human VSMC, lentiviral over-expression of NOR-1 reduced lipopolysaccharide (LPS)-induced up-regulation of cytokines (IL-1ß, IL-6 and IL-8) and chemokines (MCP-1 and CCL20). Similar effects were obtained in cells stimulated with TNFα or oxLDL. Conversely, siRNA-mediated NOR-1 inhibition significantly increased the expression of pro-inflammatory mediators. Interestingly, in the aortas from transgenic mice that over-express human NOR-1 in VSMC (TgNOR-1), the up-regulation of cytokine/chemokine by LPS was lower compared to wild-type littermates. Similar results were obtained in VSMC from transgenic animals. NOR-1 reduced the transcriptional activity of NFκB sensitive promoters (in transient transfections), and the binding of NFκB to its responsive element (in electrophoretic mobility shift assays). Furthermore, NOR-1 prevented the activation of NFκB pathway by decreasing IκBα phosphorylation/degradation and inhibiting the phosphorylation and subsequent translocation of p65 to the nucleus (assessed by Western blot and immunocytochemistry). These effects were associated with an attenuated phosphorylation of ERK1/2, p38 MAPK and Jun N-terminal kinase, pathways involved in the activation of NFκB. In mouse challenged with LPS, the activation of the NFκB signalling was also attenuated in the aorta from TgNOR-1. Our data support a role for NOR-1 as a negative modulator of the acute response elicited by pro-inflammatory stimuli in the vasculature.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Inflamación/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Animales , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Mediadores de Inflamación/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Unión Proteica , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Transducción de Señal , Activación Transcripcional
16.
Exp Physiol ; 100(1): 23-34, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25557728

RESUMEN

NEW FINDINGS: What is the central question of this study? The microtubule network is disrupted during myocardial ischaemia-reperfusion injury. It was suggested that prevention of microtubule disruption with paclitaxel might reduce cardiac infarct size; however, the effects on infarction have not been studied. What is the main finding and its importance? Paclitaxel caused a reduction in microtubule disruption and cardiomyocyte hypercontracture during ischaemia-reperfusion. However, it induced a greater increase in cytosolic calcium, which may explain the lack of effect against infarction that we have seen in isolated rat hearts. The large increase in perfusion pressure induced by paclitaxel in this model may have clinical implications, because detrimental effects of the drug were reported after its clinical application. Microtubules play a major role in the transmission of mechanical forces within the myocardium and in maintenance of organelle function. However, this intracellular network is disrupted during myocardial ischaemia-reperfusion. We assessed the effects of prevention of microtubule disruption with paclitaxel on ischaemia-reperfusion injury in isolated rat cardiomyocytes and hearts. Isolated rat cardiomyocytes were submitted to normoxia (1 h) or 45 min of simulated ischaemia (pH 6.4, 0% O2 , 37 °C) and reoxygenation, without or with treatment with the microtubule stabilizer, paclitaxel (10(-5) M), or the inhibitor of microtubule polymerization, colchicine (5 × 10(-6) M). Simulated ischaemia leads to microtubule disruption before the onset of ischaemic contracture. Paclitaxel attenuated both microtubule disruption and the incidence of hypercontracture, whereas treatment with colchicine mimicked the effects of simulated ischaemia and reoxygenation. In isolated normoxic rat hearts, treatment with paclitaxel induced concentration-dependent decreases in heart rate and left ventricular developed pressure and increases in perfusion pressure. Despite protection against hypercontracture, paclitaxel pretreatment did not modify infarct size (60.37 ± 2.27% in control hearts versus 58.75 ± 10.25, 55.44 ± 10.32 and 50.06 ± 10.14% after treatment with 10(-6) , 3 × 10(-6) and 10(-5) m of paclitaxel) after 60 min of global ischaemia and reperfusion in isolated rat hearts. Lack of protection was correlated with a higher increase in cytosolic calcium levels during simulated ischaemia in cardiomyocytes treated with paclitaxel (2.32 ± 0.15 versus 1.13 ± 0.16 a.u. in the presence or absence of 10(-6) m paclitaxel, respectively, P < 0.05), but not with changes in aortic reactivity. In conclusion, microtubule stabilization with paclitaxel reduces hypercontracture in isolated rat cardiomyocytes but does not protect against infarction in isolated rat hearts.


Asunto(s)
Microtúbulos/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Paclitaxel/farmacología , Moduladores de Tubulina/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Colchicina/farmacología , Citoprotección , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Técnicas In Vitro , Masculino , Mecanotransducción Celular/efectos de los fármacos , Microtúbulos/metabolismo , Microtúbulos/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Estrés Mecánico , Función Ventricular Izquierda/efectos de los fármacos , Presión Ventricular/efectos de los fármacos
17.
J Mol Cell Cardiol ; 68: 79-88, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24434643

RESUMEN

Mitochondria play a central role in the protection conferred by ischemic preconditioning (IP) by not fully elucidated mechanisms. We investigated whether IP protects mitochondria against ischemia-reperfusion (IR) injury through mechanisms independent of cytosolic signaling. In isolated rat hearts, sublethal IR increased superoxide production and reduced complex-I- and II-mediated respiration in subsarcolemmal (SS), but not interfibrillar (IF) mitochondria. This effect of IR on mitochondrial respiration was significantly attenuated by IP. Similar results were obtained in isolated cardiac mitochondria subjected to in vitro IR. The reduction in SS mitochondrial respiration in the heart and in vitro model was paralleled by an increase in oxidized cysteine residues, which was also prevented by IP. IP was also protective in mitochondria submitted to lethal IR. The protective effect of IP against respiratory failure was unaffected by inhibition of mitochondrial KATP channels or mitochondrial permeability transition. However, IP protection was lost in mitochondria from genetically-modified animals in which connexin-43, a protein present in SS but not IF mitochondria, was replaced by connexin-32. Our results demonstrate the existence of a protective mitochondrial mechanism or "mitochondrial preconditioning" independent of cytosol that confers protection against IR-induced respiratory failure and oxidative damage, and requires connexin-43.


Asunto(s)
Precondicionamiento Isquémico , Mitocondrias Cardíacas/fisiología , Daño por Reperfusión Miocárdica/prevención & control , Animales , Respiración de la Célula , Conexina 43/metabolismo , Circulación Coronaria , Citosol/metabolismo , Técnicas In Vitro , Activación del Canal Iónico , Masculino , Ratones , Ratones Noqueados , Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/patología , Oxidación-Reducción , Fosforilación Oxidativa , Estrés Oxidativo , Ácido Peroxinitroso/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Superóxidos/metabolismo
18.
Biochim Biophys Acta ; 1818(8): 1962-70, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21839721

RESUMEN

Multicellular organisms have developed a variety of mechanisms that allow communication between their cells. Whereas some of these systems, as neurotransmission or hormones, make possible communication between remote areas, direct cell-to-cell communication through specific membrane channels keep in contact neighboring cells. Direct communication between the cytoplasm of adjacent cells is achieved in vertebrates by membrane channels formed by connexins. However, in addition to allowing exchange of ions and small metabolites between the cytoplasms of adjacent cells, connexin channels also communicate the cytosol with the extracellular space, thus enabling a completely different communication system, involving activation of extracellular receptors. Recently, the demonstration of connexin at the inner mitochondrial membrane of cardiomyocytes, probably forming hemichannels, has enlarged the list of actions of connexins. Some of these mechanisms are also shared by a different family of proteins, termed pannexins. Importantly, these systems allow not only communication between healthy cells, but also play an important role during different types of injury. The aim of this review is to discuss the role played by both connexin hemichannels and pannexin channels in cell communication and injury. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/fisiología , Lesiones Cardíacas/metabolismo , Miocardio/patología , Proteínas del Tejido Nervioso/metabolismo , Animales , Comunicación Celular , Muerte Celular , Citoplasma/metabolismo , Cardiopatías/metabolismo , Humanos , Modelos Biológicos , Daño por Reperfusión , Transducción de Señal
19.
Basic Res Cardiol ; 108(3): 351, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23595215

RESUMEN

Connexin 43 (Cx43) deficiency increases myocardial tolerance to ischemia-reperfusion injury and abolishes preconditioning protection. It is not known whether modifications in baseline signaling through protective RISK or SAFE pathways or in response to preconditioning may contribute to these effects. To answer this question we used Cx43(Cre-ER(T)/fl) mice, in which Cx43 expression is abolished after 4-hydroxytamoxifen (4-OHT) administration. Isolated hearts from Cx43(Cre-ER(T)/fl) mice, or from Cx43(fl/fl) controls, treated with vehicle or 4-OHT, were submitted to global ischemia (40 min) and reperfusion. Cx43 deficiency was associated with reduced infarct size after ischemia-reperfusion (11.17 ± 3.25 % vs. 65.04 ± 3.79, 59.31 ± 5.36 and 65.40 ± 4.91, in Cx43(fl/fl) animals treated with vehicle, Cx43(fl/fl) mice treated with 4-OHT, and Cx43(Cre-ER(T)/fl) mice treated with vehicle, respectively, n = 8-9, p < 0.001). However, the ratio phosphorylated/total protein expression for Akt, ERK-1/2, GSK3ß and STAT3 was not increased in normoxic samples from animals lacking Cx43. Instead, a reduction in the phosphorylation state of GSK3ß was observed in Cx43-deficient mice (ratio: 0.15 ± 0.02 vs. 0.56 ± 0.11, 0.77 ± 0.15, and 0.46 ± 0.14, respectively, n = 5-6, p < 0.01). Furthermore, ischemic preconditioning (IPC, 4 cycles of 3.5 min of ischemia and 5 min of reperfusion) increased phosphorylation of ERK-1/2, GSK3ß, and STAT3 in all hearts without differences between groups (n = 5-6, p < 0.05), although Cx43 deficient mice were not protected by either IPC or pharmacological preconditioning with diazoxide. Our data demonstrate that modification of RISK and SAFE signaling does not contribute to the role of Cx43 in the increased tolerance to myocardial ischemia-reperfusion injury and in preconditioning protection.


Asunto(s)
Conexina 43/deficiencia , Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/enzimología , Transducción de Señal , Adenosina Trifosfato/metabolismo , Animales , Muerte Celular , Conexina 43/genética , Modelos Animales de Enfermedad , Metabolismo Energético , Activación Enzimática , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Fosfocreatina/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Función Ventricular Izquierda , Presión Ventricular
20.
Biomed Pharmacother ; 167: 115469, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37729730

RESUMEN

Extracellular matrix (ECM) is an active player in cardiovascular calcification (CVC), a major public health issue with an unmet need for effective therapies. Lysyl oxidase (LOX) conditions ECM biomechanical properties; thus, we hypothesized that LOX might impact on mineral deposition in calcific aortic valve disease (CAVD) and atherosclerosis. LOX was upregulated in calcified valves from two cohorts of CAVD patients. Strong LOX immunostaining was detected surrounding calcified foci in calcified human valves and atherosclerotic lesions colocalizing with RUNX2 on valvular interstitial cells (VICs) or vascular smooth muscle cells (VSMCs). Both LOX secretion and organized collagen deposition were enhanced in calcifying VICs exposed to osteogenic media. ß-aminopropionitrile (BAPN), an inhibitor of LOX, attenuated collagen deposition and calcification. VICs seeded onto decellularized matrices from BAPN-treated VICs calcified less than cells cultured onto control scaffolds; instead, VICs exposed to conditioned media from cells over-expressing LOX or cultured onto LOX-crosslinked matrices calcified more. Atherosclerosis was induced in WT and transgenic mice that overexpress LOX in VSMC (TgLOXVSMC) by AAV-PCSK9D374Y injection and high-fat feeding. In atherosclerosis-challenged TgLOXVSMC mice both atherosclerosis burden and calcification assessed by near-infrared fluorescence (NIRF) imaging were higher than in WT mice. These animals also exhibited larger calcified areas in atherosclerotic lesions from aortic arches and brachiocephalic arteries. Moreover, LOX transgenesis exacerbated plaque inflammation, and increased VSMC cellularity, the rate of RUNX2-positive cells and both connective tissue content and collagen cross-linking. Our findings highlight the relevance of LOX in CVC and postulate this enzyme as a potential therapeutic target for CVC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA