Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Intervalo de año de publicación
1.
Genome Med ; 16(1): 97, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39135118

RESUMEN

BACKGROUND: Congenital heart disease (CHD) is the most prevalent congenital anomaly, but its underlying causes are still not fully understood. It is believed that multiple rare genetic mutations may contribute to the development of CHD. METHODS: In this study, we aimed to identify novel genetic risk factors for CHD using an ENU-based dominant genetic screen in mice. We analyzed fetuses with malformed hearts and compared them to control littermates by whole exome or whole genome sequencing (WES/WGS). The differences in mutation rates between observed and expected values were tested using the Poisson and Binomial distribution. Additionally, we compared WES data from human CHD probands obtained from the Pediatric Cardiac Genomics Consortium with control subjects from the 1000 Genomes Project using Fisher's exact test to evaluate the burden of rare inherited damaging mutations in patients. RESULTS: By screening 10,285 fetuses, we identified 1109 cases with various heart defects, with ventricular septal defects and bicuspid aortic valves being the most common types. WES/WGS analysis of 598 cases and 532 control littermates revealed a higher number of ENU-induced damaging mutations in cases compared to controls. GO term and KEGG pathway enrichment analysis showed that pathways related to cardiac contraction and neuronal development and functions were enriched in cases. Further analysis of 1457 human CHD probands and 2675 control subjects also revealed an enrichment of genes associated with muscle and nervous system development in patients. By combining the mice and human data, we identified a list of 101 candidate digenic genesets, from which each geneset was co-mutated in at least one mouse and two human probands with CHD but not in control mouse and control human subjects. CONCLUSIONS: Our findings suggest that gene mutations affecting early hemodynamic perturbations in the developing heart may play a significant role as a genetic risk factor for CHD. Further validation of the candidate gene set identified in this study could enhance our understanding of the complex genetics underlying CHD and potentially lead to the development of new diagnostic and therapeutic approaches.


Asunto(s)
Cardiopatías Congénitas , Mutación , Cardiopatías Congénitas/genética , Animales , Humanos , Ratones , Pruebas Genéticas , Femenino , Masculino , Predisposición Genética a la Enfermedad , Secuenciación del Exoma , Neuronas/metabolismo , Proteínas Contráctiles/genética
2.
J Biol Chem ; 300(7): 107470, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38879012

RESUMEN

Resistance to inhibitors of cholinesterases (ric-8 proteins) are involved in modulating G-protein function, but little is known of their potential physiological importance in the heart. In the present study, we assessed the role of resistance to inhibitors of cholinesterase 8b (Ric-8b) in determining cardiac contractile function. We developed a murine model in which it was possible to conditionally delete ric-8b in cardiac tissue in the adult animal after the addition of tamoxifen. Deletion of ric-8b led to severely reduced contractility as measured using echocardiography days after administration of tamoxifen. Histological analysis of the ventricular tissue showed highly variable myocyte size, prominent fibrosis, and an increase in cellular apoptosis. RNA sequencing revealed transcriptional remodeling in response to cardiac ric-8b deletion involving the extracellular matrix and inflammation. Phosphoproteomic analysis revealed substantial downregulation of phosphopeptides related to myosin light chain 2. At the cellular level, the deletion of ric-8b led to loss of activation of the L-type calcium channel through the ß-adrenergic pathways. Using fluorescence resonance energy transfer-based assays, we showed ric-8b protein selectively interacts with the stimulatory G-protein, Gαs. We explored if deletion of Gnas (the gene encoding Gαs) in cardiac tissue using a similar approach in the mouse led to an equivalent phenotype. The conditional deletion of the Gαs gene in the ventricle led to comparable effects on contractile function and cardiac histology. We conclude that ric-8b is essential to preserve cardiac contractile function likely through an interaction with the stimulatory G-protein and downstream phosphorylation of myosin light chain 2.


Asunto(s)
Contracción Miocárdica , Animales , Ratones , Contracción Miocárdica/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Cadenas Ligeras de Miosina/genética , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/genética , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/genética , Miocardio/metabolismo , Miocardio/patología , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Humanos , Inhibidores de la Colinesterasa/farmacología , Masculino , Apoptosis/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido
3.
Biosensors (Basel) ; 14(5)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38785693

RESUMEN

Zebrafish larvae have emerged as a valuable model for studying heart physiology and pathophysiology, as well as for drug discovery, in part thanks to its transparency, which simplifies microscopy. However, in fluorescence-based optical mapping, the beating of the heart results in motion artifacts. Two approaches have been employed to eliminate heart motion during calcium or voltage mapping in zebrafish larvae: the knockdown of cardiac troponin T2A and the use of myosin inhibitors. However, these methods disrupt the mechano-electric and mechano-mechanic coupling mechanisms. We have used ratiometric genetically encoded biosensors to image calcium in the beating heart of intact zebrafish larvae because ratiometric quantification corrects for motion artifacts. In this study, we found that halting heart motion by genetic means (injection of tnnt2a morpholino) or chemical tools (incubation with para-aminoblebbistatin) leads to bradycardia, and increases calcium levels and the size of the calcium transients, likely by abolishing a feedback mechanism that connects contraction with calcium regulation. These outcomes were not influenced by the calcium-binding domain of the gene-encoded biosensors employed, as biosensors with a modified troponin C (Twitch-4), calmodulin (mCyRFP1-GCaMP6f), or the photoprotein aequorin (GFP-aequorin) all yielded similar results. Cardiac contraction appears to be an important regulator of systolic and diastolic Ca2+ levels, and of the heart rate.


Asunto(s)
Técnicas Biosensibles , Calcio , Larva , Contracción Miocárdica , Pez Cebra , Animales , Calcio/metabolismo , Contracción Miocárdica/fisiología , Corazón/fisiología , Troponina T/metabolismo , Proteínas de Pez Cebra/metabolismo , Troponina C/metabolismo
4.
Biochem Biophys Res Commun ; 681: 200-211, 2023 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-37783118

RESUMEN

Human heart tissues grown as three-dimensional spheroids and consisting of different cardiac cell types derived from pluripotent stem cells (hiPSCs) recapitulate aspects of human physiology better than standard two-dimensional models in vitro. They typically consist of less than 5000 cells and are used to measure contraction kinetics although not contraction force. By contrast, engineered heart tissues (EHTs) formed around two flexible pillars, can measure contraction force but conventional EHTs often require between 0.5 and 2 million cells. This makes large-scale screening of many EHTs costly. Our goals here were (i) to create a physiologically relevant model that required fewer cells than standard EHTs making them less expensive, and (ii) to ensure that this miniaturized model retained correct functionality. We demonstrated that fully functional EHTs could be generated from physiologically relevant combinations of hiPSC-derived cardiomyocytes (70%), cardiac fibroblasts (15%) and cardiac endothelial cells (15%), using as few as 1.6 × 104 cells. Our results showed that these EHTs were viable and functional up to 14 days after formation. The EHTs could be electrically paced in the frequency range between 0.6 and 3 Hz, with the optimum between 0.6 and 2 Hz. This was consistent across three downscaled EHT sizes tested. These findings suggest that miniaturized EHTs could represent a cost-effective microphysiological system for disease modelling and examining drug responses particularly in secondary screens for drug discovery.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Células Endoteliales , Técnicas de Cocultivo , Miocitos Cardíacos/metabolismo , Contracción Miocárdica , Ingeniería de Tejidos/métodos
5.
J Mol Cell Biol ; 2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37771085

RESUMEN

Mitochondrial aldehyde dehydrogenase (ALDH2) offers proven cardiovascular benefit although its impact in diabetes remains elusive. This study examined the effect of ALDH2 overexpression (OE) and knockout (KO) on diabetic cardiomyopathy and mechanism involved with a focus on mitochondrial integrity. ALDH2 OE and KO mice were challenged with streptozotocin (STZ, 200 mg/kg. i.p.) to establish diabetes. Diabetic patients displayed reduced plasma ALDH2 activity, cardiac remodeling and diastolic dysfunction. STZ challenge prompted reduced respiratory exchange ratio (RER), dampened fractional shortening, ejection fraction, increased LV end systolic and diastolic diameters, cardiac remodeling, cardiomyocyte contractile and intracellular Ca2+ defects (depressed peak shortening and maximal velocity of shortening/relengthening, prolonged relengthening, dampened intracellular Ca2+ rise and clearance), myocardial ultrastructural injury, oxidative stress, apoptosis and mitochondrial damage, the effects of which were overtly attenuated and accentuated by ALDH2 OE and KO, respectively. Immunoblotting revealed downregulated mitochondrial proteins PPARγ coactivator 1α (PGC-1α) and UCP-2, Ca2+ regulatory proteins including SERCA and Na+-Ca2+ exchanger, elevated phospholamban, dampened autophagy and mitophagy (LC3B ratio, TOM20, Parkin, FUNDC1 and BNIP3), disrupted phosphorylation of Akt, GSK3ß and Foxo3a, and elevated PTEN phosphorylation, the effect of which was reversed and worsened by ALDH2 OE and KO, respectively (except FUNDC1 and BNIP3). In vivo and in vitro data revealed that novel ALDH2 activator torezolid/Alda-1 protected against STZ or high glucose-induced cardiac anomalies, the effect was nullified by inhibition of Akt, GSK3ß, Parkin and mitochondrial coupling. Our data discerned a vital role for ALDH2 in diabetic cardiomyopathy possibly through regulation of Akt, GSK3ß activation, parkin mitophagy and mitochondrial function.

6.
Pharmacol Res ; 196: 106931, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37722519

RESUMEN

Evidence exists that heart failure (HF) has an overall impact of 1-2 % in the global population being often associated with comorbidities that contribute to increased disease prevalence, hospitalization, and mortality. Recent advances in pharmacological approaches have significantly improved clinical outcomes for patients with vascular injury and HF. Nevertheless, there remains an unmet need to clarify the crucial role of nitric oxide/cyclic guanosine 3',5'-monophosphate (NO/cGMP) signalling in cardiac contraction and relaxation, to better identify the key mechanisms involved in the pathophysiology of myocardial dysfunction both with reduced (HFrEF) as well as preserved ejection fraction (HFpEF). Indeed, NO signalling plays a crucial role in cardiovascular homeostasis and its dysregulation induces a significant increase in oxidative and nitrosative stress, producing anatomical and physiological cardiac alterations that can lead to heart failure. The present review aims to examine the molecular mechanisms involved in the bioavailability of NO and its modulation of downstream pathways. In particular, we focus on the main therapeutic targets and emphasize the recent evidence of preclinical and clinical studies, describing the different emerging therapeutic strategies developed to counteract NO impaired signalling and cardiovascular disease (CVD) development.


Asunto(s)
Insuficiencia Cardíaca , Humanos , Insuficiencia Cardíaca/tratamiento farmacológico , Óxido Nítrico/metabolismo , Volumen Sistólico , Corazón , GMP Cíclico/metabolismo
7.
Elife ; 122023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37589544

RESUMEN

G-protein-coupled receptors (GPCRs) transmit signals into cells depending on the G protein type. To analyze the functions of GPCR signaling, we assessed the effectiveness of animal G-protein-coupled bistable rhodopsins that can be controlled into active and inactive states by light application using zebrafish. We expressed Gq- and Gi/o-coupled bistable rhodopsins in hindbrain reticulospinal V2a neurons, which are involved in locomotion, or in cardiomyocytes. Light stimulation of the reticulospinal V2a neurons expressing Gq-coupled spider Rh1 resulted in an increase in the intracellular Ca2+ level and evoked swimming behavior. Light stimulation of cardiomyocytes expressing the Gi/o-coupled mosquito Opn3, pufferfish TMT opsin, or lamprey parapinopsin induced cardiac arrest, and the effect was suppressed by treatment with pertussis toxin or barium, suggesting that Gi/o-dependent regulation of inward-rectifier K+ channels controls cardiac function. These data indicate that these rhodopsins are useful for optogenetic control of GPCR-mediated signaling in zebrafish neurons and cardiomyocytes.


Asunto(s)
Miocitos Cardíacos , Canales de Potasio de Rectificación Interna , Animales , Pez Cebra , Optogenética , Neuronas , Rodopsina
8.
Elife ; 122023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37589546

RESUMEN

Even though microbial photosensitive proteins have been used for optogenetics, their use should be optimized to precisely control cell and tissue functions in vivo. We exploited GtCCR4 and KnChR, cation channelrhodopsins from algae, BeGC1, a guanylyl cyclase rhodopsin from a fungus, and photoactivated adenylyl cyclases (PACs) from cyanobacteria (OaPAC) or bacteria (bPAC), to control cell functions in zebrafish. Optical activation of GtCCR4 and KnChR in the hindbrain reticulospinal V2a neurons, which are involved in locomotion, induced swimming behavior at relatively short latencies, whereas activation of BeGC1 or PACs achieved it at long latencies. Activation of GtCCR4 and KnChR in cardiomyocytes induced cardiac arrest, whereas activation of bPAC gradually induced bradycardia. KnChR activation led to an increase in intracellular Ca2+ in the heart, suggesting that depolarization caused cardiac arrest. These data suggest that these optogenetic tools can be used to reveal the function and regulation of zebrafish neurons and cardiomyocytes.


Asunto(s)
Paro Cardíaco , Miocitos Cardíacos , Animales , Adenilil Ciclasas/genética , Pez Cebra , Rodopsinas Microbianas , Optogenética , Neuronas
9.
Basic Res Cardiol ; 117(1): 37, 2022 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-35842861

RESUMEN

We have recently identified a pool of intracellular ß1 adrenergic receptors (ß1ARs) at the sarcoplasmic reticulum (SR) crucial for cardiac function. Here, we aim to characterize the integrative control of intracellular catecholamine for subcellular ß1AR signaling and cardiac function. Using anchored Förster resonance energy transfer (FRET) biosensors and transgenic mice, we determined the regulation of compartmentalized ß1AR-PKA signaling at the SR and plasma membrane (PM) microdomains by organic cation transporter 3 (OCT3) and monoamine oxidase A (MAO-A), two critical modulators of catecholamine uptake and homeostasis. Additionally, we examined local PKA substrate phosphorylation and excitation-contraction coupling in cardiomyocyte. Cardiac-specific deletion of MAO-A (MAO-A-CKO) elevates catecholamines and cAMP levels in the myocardium, baseline cardiac function, and adrenergic responses. Both MAO-A deletion and inhibitor (MAOi) selectively enhance the local ß1AR-PKA activity at the SR but not PM, and augment phosphorylation of phospholamban, Ca2+ cycling, and myocyte contractile response. Overexpression of MAO-A suppresses the SR-ß1AR-PKA activity and PKA phosphorylation. However, deletion or inhibition of OCT3 by corticosterone prevents the effects induced by MAOi and MAO-A deletion in cardiomyocytes. Deletion or inhibition of OCT3 also negates the effects of MAOi and MAO-A deficiency in cardiac function and adrenergic responses in vivo. Our data show that MAO-A and OCT3 act in concert to fine-tune the intracellular SR-ß1AR-PKA signaling and cardiac fight-or-flight response. We reveal a drug contraindication between anti-inflammatory corticosterone and anti-depressant MAOi in modulating adrenergic regulation in the heart, providing novel perspectives of these drugs with cardiac implications.


Asunto(s)
Corticosterona , Proteínas Quinasas Dependientes de AMP Cíclico , Adrenérgicos/metabolismo , Adrenérgicos/farmacología , Animales , Calcio/metabolismo , Catecolaminas/metabolismo , Catecolaminas/farmacología , Cationes/metabolismo , Cationes/farmacología , Corticosterona/metabolismo , Corticosterona/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Ratones , Monoaminooxidasa/metabolismo , Monoaminooxidasa/farmacología , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Fosforilación , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Retículo Sarcoplasmático
10.
Int J Mol Sci ; 22(21)2021 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-34768802

RESUMEN

Cardiac development is a complex process that is strictly controlled by various factors, including PcG protein complexes. Several studies have reported the critical role of PRC2 in cardiogenesis. However, little is known about the regulation mechanism of PRC1 in embryonic heart development. To gain more insight into the mechanistic role of PRC1 in cardiogenesis, we generated a PRC1 loss-of-function zebrafish line by using the CRISPR/Cas9 system targeting rnf2, a gene encoding the core subunit shared by all PRC1 subfamilies. Our results revealed that Rnf2 is not involved in cardiomyocyte differentiation and heart tube formation, but that it is crucial to maintaining regular cardiac contraction. Further analysis suggested that Rnf2 loss-of-function disrupted cardiac sarcomere assembly through the ectopic activation of non-cardiac sarcomere genes in the developing heart. Meanwhile, Rnf2 deficiency disrupts the construction of the atrioventricular canal and the sinoatrial node by modulating the expression of bmp4 and other atrioventricular canal marker genes, leading to an impaired cardiac conduction system. The disorganized cardiac sarcomere and defective cardiac conduction system together contribute to defective cardiac contraction. Our results emphasize the critical role of PRC1 in the cardiac development.


Asunto(s)
Corazón/crecimiento & desarrollo , Contracción Muscular , Miocardio/metabolismo , Sarcómeros/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Desarrollo Embrionario , Corazón/fisiología , Mutación con Pérdida de Función , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 1/fisiología , Sarcómeros/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
11.
Toxicol Appl Pharmacol ; 428: 115696, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34419494

RESUMEN

Bisphenol A (BPA) is a well-known endocrine-disrupting chemical that is widely used in a variety of products, including plastics, medical equipment and receipts. Hence, most people are exposed to BPA through the skin, via inhalation and via the digestive system, and such exposure has been linked to cardiovascular diseases including coronary artery disease, hypertension, atherosclerosis, and myocardial infarction. However, the underlying mechanisms of cardiac dysfunction caused by BPA remain poorly understood. In this study, we found that BPA exposure altered cardiac function in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Acute BPA exposure in hiPSC-CMs resulted in reduced field potential, as measured by multielectrode array (MEA). Furthermore, we observed that BPA dose-dependently inhibited ICa, INa or IKr channels. In addition, BPA exposure dose-dependently inhibited calcium transients and contraction in hiPSC-CMs. Our findings suggest that BPA exposure leads to cardiac dysfunction and cardiac risk factors such as arrhythmia.


Asunto(s)
Contaminantes Ocupacionales del Aire/toxicidad , Compuestos de Bencidrilo/toxicidad , Cardiotoxinas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Fenoles/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
12.
J Mol Biol ; 433(19): 167178, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34329643

RESUMEN

Cardiac muscle contraction depends on interactions between thick (myosin) and thin (actin) filaments (TFs). TFs are regulated by intracellular Ca2+ levels. Under activating conditions Ca2+ binds to the troponin complex and displaces tropomyosin from myosin binding sites on the TF surface to allow actomyosin interactions. Recent studies have shown that in addition to Ca2+, the first four N-terminal domains (NTDs) of cardiac myosin binding protein C (cMyBP-C) (e.g. C0, C1, M and C2), are potent modulators of the TF activity, but the mechanism of their collective action is poorly understood. Previously, we showed that C1 activates the TF at low Ca2+ and C0 stabilizes binding of C1 to the TF, but the ability of C2 to bind and/or affect the TF remains unknown. Here we obtained 7.5 Å resolution cryo-EM reconstruction of C2-decorated actin filaments to demonstrate that C2 binds to actin in a single structural mode that does not activate the TF unlike the polymorphic binding of C0 and C1 to actin. Comparison of amino acid sequences of C2 with either C0 or C1 shows low levels of identity between the residues involved in interactions with the TF but high levels of conservation for residues involved in Ig fold stabilization. This provides a structural basis for strikingly different interactions of structurally homologous C0, C1 and C2 with the TF. Our detailed analysis of the interaction of C2 with the actin filament provides crucial information required to model the collective action of cMyBP-C NTDs on the cardiac TF.


Asunto(s)
Actinas/química , Actinas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Sitios de Unión , Calcio/metabolismo , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
13.
Cell Signal ; 85: 110059, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34062239

RESUMEN

Protein phosphatase 1(PP1) is a key regulator of cardiac function through dephosphorylating serine/threonine residues within target proteins to oppose the function of protein kinases. Studies from failing hearts of animal models and human patients have demonstrated significant increase of PP1 activity in myocardium, while elevated PP1 activity in transgenic mice leads to cardiac dysfunction, suggesting that PP1 might be a therapeutic target to ameliorate cardiac dysfunction in failing hearts. In fact, cardiac overexpression of inhibitor 1, the endogenous inhibitor of PP1, increases cardiac contractility and suppresses heart failure progression. However, this notion of PP1 inhibition for heart failure treatment has been challenged by recent studies on the isoform-specific roles of PP1 in the heart. PP1 is a holoenzyme composed of catalytic subunits (PP1α, PP1ß, or PP1γ) and regulatory proteins that target them to distinct subcellular locations for functional specificity. This review will summarize how PP1 regulates phosphorylation of some of the key cardiac proteins involved in Ca2+ handling and cardiac contraction, and the potential role of PP1 isoforms in controlling cardiac physiology and pathophysiology.


Asunto(s)
Corazón , Miocardio , Animales , Humanos , Ratones , Contracción Miocárdica , Miocardio/metabolismo , Fosforilación , Isoformas de Proteínas/metabolismo , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/metabolismo
14.
Cell Rep ; 35(5): 109097, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33951436

RESUMEN

Cardiac cells generate and amplify force in the context of cardiac load, yet the membranous sheath enclosing the muscle fibers-the sarcolemma-does not experience displacement. That the sarcolemma sustains beat-to-beat pressure changes without experiencing significant distortion is a muscle-contraction paradox. Here, we report that an elastic element-the motor protein prestin (Slc26a5)-serves to amplify actin-myosin force generation in mouse and human cardiac myocytes, accounting partly for the nonlinear capacitance of cardiomyocytes. The functional significance of prestin is underpinned by significant alterations of cardiac contractility in Prestin-knockout mice. Prestin was previously considered exclusive to the inner ear's outer hair cells; however, our results show that prestin serves a broader cellular motor function.


Asunto(s)
Corazón/fisiología , Proteínas Motoras Moleculares/metabolismo , Transportadores de Sulfato/metabolismo , Animales , Humanos , Ratones
15.
Prog Biophys Mol Biol ; 159: 58-74, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32710902

RESUMEN

Human-based computational modelling and simulation are powerful tools to accelerate the mechanistic understanding of cardiac patho-physiology, and to develop and evaluate therapeutic interventions. The aim of this study is to calibrate and evaluate human ventricular electro-mechanical models for investigations on the effect of the electro-mechanical coupling and pharmacological action on human ventricular electrophysiology, calcium dynamics, and active contraction. The most recent models of human ventricular electrophysiology, excitation-contraction coupling, and active contraction were integrated, and the coupled models were calibrated using human experimental data. Simulations were then conducted using the coupled models to quantify the effects of electro-mechanical coupling and drug exposure on electrophysiology and force generation in virtual human ventricular cardiomyocytes and tissue. The resulting calibrated human electro-mechanical models yielded active tension, action potential, and calcium transient metrics that are in agreement with experiments for endocardial, epicardial, and mid-myocardial human samples. Simulation results correctly predicted the inotropic response of different multichannel action reference compounds and demonstrated that the electro-mechanical coupling improves the robustness of repolarisation under drug exposure compared to electrophysiology-only models. They also generated additional evidence to explain the partial mismatch between in-silico and in-vitro experiments on drug-induced electrophysiology changes. The human calibrated and evaluated modelling and simulation framework constructed in this study opens new avenues for future investigations into the complex interplay between the electrical and mechanical cardiac substrates, its modulation by pharmacological action, and its translation to tissue and organ models of cardiac patho-physiology.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Simulación por Computador , Ventrículos Cardíacos/metabolismo , Modelos Cardiovasculares , Miocitos Cardíacos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Fenómenos Biomecánicos , Calcio/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Fenómenos Electrofisiológicos , Humanos , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Medición de Riesgo
16.
ACS Appl Bio Mater ; 4(1): 267-276, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35014283

RESUMEN

The development of cardiac models that faithfully recapitulate heart conditions is the goal of cardiac biomedical research. Among the numerous limitations of current models, replication of the cardiac microenvironment is one of the key challenges, and the effect of mechanical cues remains obscure in cardiac tissue. In this paper, different topological structures in the engineered cardiac models are summarized, and mechanical regulation of myocyte morphology and functional responses are discussed. Microenvironmental cues in vivo are influencing cardiac functions from cellular to tissue levels, and replications of these micro and macro features in the in vitro cardiac model shed light on cardiac research from a mechanistic point of view. With simple manipulation of topology, both physiological and pathological cardiac constructs can be remodeled to investigate the origin of abnormal cell phenotypes and functional responses in cardiac diseases. The integration of topological guidance with heart-on-a-chip devices is covered briefly and limitations of the current cardiac constructs are also addressed for future advancements in personalized medicine.


Asunto(s)
Modelos Biológicos , Miocitos Cardíacos/fisiología , Cardiomiopatías/patología , Técnicas de Cultivo Tridimensional de Células , Matriz Extracelular/metabolismo , Humanos , Dispositivos Laboratorio en un Chip , Mecanotransducción Celular , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología
17.
Rev. cuba. invest. bioméd ; 40(supl.1): e1506, 2021. tab, graf
Artículo en Español | LILACS, CUMED | ID: biblio-1289474

RESUMEN

Introducción: El paracetamol es uno de los antiinflamatorios no esteroideos con efecto analgésico y antipirético más utilizados a nivel mundial. Pocos estudios se enfocan en esclarecer los mecanismos de acción a nivel cardiovascular. Objetivos: Evaluar la acción del paracetamol sobre la fuerza de contracción de anillos de aorta torácica y sobre la actividad eléctrica y contráctil de corazones aislados y perfundidos de ratas Wistar. Métodos: Se midieron los efectos del paracetamol sobre anillos de aorta de rata denudados de su endotelio vascular. Se estudiaron las acciones del fármaco sobre los corazones aislados y perfundidos de las ratas por el método de Langendorff. Se evaluó la amplitud de la fuerza de contracción cardiaca y los intervalos QT, QTc, QRS y RR del electrocardiograma. Las condiciones (control y presencia de paracetamol) fueron comparadas con una prueba t de Student para muestras pareadas (p < 0,05), previa comprobación de la normalidad de los datos. Resultados: El paracetamol no tuvo efectos sobre el músculo liso vascular de los anillos aórticos ni sobre los intervalos QT, QTc, QRS y RR del electrocardiograma bajo ninguna de las concentraciones empleadas. Por otra parte, mostró efecto inotrópico negativo estadísticamente significativo en los corazones aislados, de forma dependiente de la concentración del fármaco. La IC50 estimada para la inhibición de la fuerza de contracción cardiaca fue de 17,15 ± 5,33 µmol/L. Conclusiones: Las acciones cardiovasculares directas del paracetamol son modestas, lo cual contribuye al buen margen de seguridad para su uso en clínica, en pacientes sin enfermedad cardiovascular(AU)


Introduction: Paracetamol is among the non-steroidal anti-inflammatory, analgesic and antipyretic drugs most commonly used worldwide. Few studies have focused on clarifying its mechanisms of action on a cardiovascular level. Objectives: Evaluate the action of paracetamol on the force of contraction of thoracic aortic rings and on the electrical and contractile activity of isolated perfused Wistar rat hearts. Methods: Measurements were taken of the effects of paracetamol on rat aortic rings denuded of their vascular endothelium. Analysis was performed of the actions of the drug on the isolated perfused rat hearts using the Langendorff method. Evaluation was conducted of the amplitude of the force of cardiac contraction and of intervals QT, QTc, QRS and RR of the electrocardiogram. The conditions (control and presence of paracetamol) were compared with a paired samples Student's t-test (p < 0.05) upon verification of the normality of the data. Results: Paracetamol had no effects on the vascular smooth muscle of aortic rings or on intervals QT, QTc, QRS and RR of the electrocardiogram at none of the concentrations used. On the other hand, it displayed a statistically significant negative inotropic effect on the isolated hearts dependent on drug concentration. The IC50 estimated for inhibition of the force of cardiac contraction was 17.15 ± 5.33 µmol/L. Conclusions: The direct cardiovascular actions of paracetamol are modest, which contributes to a good safety margin for its clinical use in patients without cardiovascular disease(AU)


Asunto(s)
Humanos , Masculino , Femenino , Enfermedades Cardiovasculares/prevención & control , Antiinflamatorios no Esteroideos , Corazón , Acetaminofén/análisis
18.
Acta Physiol (Oxf) ; 228(3): e13429, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31837094

RESUMEN

Heart failure (HF) is the end-stage syndrome for most cardiac diseases, and the 5-year morbidity and mortality of HF remain high. Malignant arrhythmia is the main cause of sudden death in the progression of HF. Recently, bridging integrator 1 (BIN1) was discovered as a regulator of transverse tubule function and calcium signalling in cardiomyocytes. BIN1 downregulation is linked to abnormal cardiac contraction, and it increases the possibility of malignant arrhythmias preceding HF. Because of the detectability of cardiac BIN1 in peripheral blood, BIN1 may serve as a predictor of HF and may be useful in therapy development. However, the mechanism of BIN1 downregulation in HF and how BIN1 regulates normal cardiac function under physiological conditions remain unclear. In this review, recent progress in the biological studies of BIN1-related cardiomyocytes and the effect of cardiac dysfunction and malignant arrhythmia will be discussed.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Arritmias Cardíacas/metabolismo , Insuficiencia Cardíaca/metabolismo , Proteínas Nucleares/metabolismo , Animales , Arritmias Cardíacas/patología , Insuficiencia Cardíaca/patología , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Supresoras de Tumor/metabolismo
19.
J Vet Med Sci ; 81(10): 1509-1514, 2019 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-31484844

RESUMEN

Eukaryotic elongation factor 2 (eEF2) kinase (eEF2K) acts to inhibit protein translation through phosphorylating a specific substrate, eEF2. We previously found that the increased eEF2K expression in mesenteric artery mediates hypertension development in spontaneously hypertensive rats. More recently, we have revealed that a selective eEF2K inhibitor, A484954 induced vasorelaxation via opening inward rectifier K+ channel and activating ß2-adrenergic receptor in smooth muscle of rat isolated mesenteric artery, which contributes to prevent noradrenaline-induced acute increase in blood pressure (BP). In this study, we further explored acute effects of A484954 on BP in rats, especially focusing the action on ß-adrenergic receptor. We also examined whether A484954 affects contraction and heart rate (HR) of isolated heart. BP and HR were measured by a carotid cannulation method in rats. Isometric contraction and HR in rat isolated atria were also measured pharmacologically. A484954 potentiated adrenaline-induced decrease in diastolic BP (DBP) but not increase in systolic BP (SBP). A484954 potentiated isoproterenol-induced decrease in DBP but not SBP. Contrastingly, A484954 prevented a non-ß-adrenergic receptor agonist, angiotensin II-induced increase in both SBP and DBP. In isolated left atria, A484954 caused contraction, which was prevented by a ß-adrenergic receptor antagonist, propranolol. In isolated right atria, A484954 increased HR. In conclusion, we for the first time demonstrated that A484954 potentiates ß-adrenergic receptor agonist-induced decrease in DBP possibly through vasorelaxation mediated via activating ß2-adrenergic receptor. It was also demonstrated that A484954 causes contraction of rat isolated heart via activating ß1-adrenergic receptor.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Presión Sanguínea/efectos de los fármacos , Ciclopropanos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirrolidinas/farmacología , Vasodilatación/efectos de los fármacos , Antagonistas Adrenérgicos beta/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Determinación de la Presión Sanguínea , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Contracción Miocárdica/efectos de los fármacos , Propranolol/farmacología , Piridinas , Ratas , Ratas Wistar , Receptores Adrenérgicos beta/metabolismo
20.
Basic Res Cardiol ; 114(5): 39, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31463567

RESUMEN

Enhanced reactive oxygen species (ROS) at the beginning of reperfusion activated signal transducer and activator of transcription 3 (STAT3) in intermittent hypobaric hypoxia (IHH)-afforded cardioprotection against ischemia/reperfusion (I/R). However, its mechanism remains largely unknown. This study aimed to investigate the role and the downstream of STAT3 in exogenous enhanced post-ischemic ROS-induced cardioprotection using the model of moderate hydrogen peroxide postconditioning (H2O2PoC) mimicking endogenous ROS in IHH. Moderate H2O2PoC not only improved the post-ischemic myocardial contractile recovery and reduced the infarct size in isolated rat I/R hearts, but also alleviated mitochondrial calcium overload and ameliorated Ca2+ transients, cell contraction, and mitochondrial membrane potential in rat I/R cardiomyocytes. However, the cardioprotective effects of moderate H2O2PoC were abrogated by Janus kinase 2 (JAK2)/STAT3 inhibitor AG490 in rat hearts as well as adenovirus-delivered short hairpin RNA specific for STAT3 and the opener of mitochondrial calcium uniporter (MCU) spermine in rat cardiomyocytes. Notably, the moderate H2O2PoC-afforded cardioprotection abrogated by spermine could be rescued by STAT3 over-expression with adenovirus in rat I/R cardiomyocytes. Besides, moderate H2O2PoC enhanced mitochondrial STAT3 expression during I/R. A co-localization/interaction of STAT3 or phospho-STAT3ser727 and MCU was observed in rat cardiomyocytes with moderate H2O2PoC at 5 and 30 min of reperfusion but not in rat I/R cardiomyocytes. Further, STAT3 interacted with the N-terminal domain (NTD) of MCU in rat cardiomyocytes with moderate H2O2PoC. These findings indicated that post-ischemic moderate ROS activate STAT3 against cardiac I/R by inhibiting MCU opening via its interaction with the NTD of MCU to alleviate mitochondrial calcium overload.


Asunto(s)
Canales de Calcio/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Precondicionamiento Isquémico Miocárdico , Masculino , Mitocondrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA