Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 132(5): 628-644, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36744470

RESUMEN

BACKGROUND: The pathogenesis of MYBPC3-associated hypertrophic cardiomyopathy (HCM) is still unresolved. In our HCM patient cohort, a large and well-characterized population carrying the MYBPC3:c772G>A variant (p.Glu258Lys, E258K) provides the unique opportunity to study the basic mechanisms of MYBPC3-HCM with a comprehensive translational approach. METHODS: We collected clinical and genetic data from 93 HCM patients carrying the MYBPC3:c772G>A variant. Functional perturbations were investigated using different biophysical techniques in left ventricular samples from 4 patients who underwent myectomy for refractory outflow obstruction, compared with samples from non-failing non-hypertrophic surgical patients and healthy donors. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and engineered heart tissues (EHTs) were also investigated. RESULTS: Haplotype analysis revealed MYBPC3:c772G>A as a founder mutation in Tuscany. In ventricular myocardium, the mutation leads to reduced cMyBP-C (cardiac myosin binding protein-C) expression, supporting haploinsufficiency as the main primary disease mechanism. Mechanical studies in single myofibrils and permeabilized muscle strips highlighted faster cross-bridge cycling, and higher energy cost of tension generation. A novel approach based on tissue clearing and advanced optical microscopy supported the idea that the sarcomere energetics dysfunction is intrinsically related with the reduction in cMyBP-C. Studies in single cardiomyocytes (native and hiPSC-derived), intact trabeculae and hiPSC-EHTs revealed prolonged action potentials, slower Ca2+ transients and preserved twitch duration, suggesting that the slower excitation-contraction coupling counterbalanced the faster sarcomere kinetics. This conclusion was strengthened by in silico simulations. CONCLUSIONS: HCM-related MYBPC3:c772G>A mutation invariably impairs sarcomere energetics and cross-bridge cycling. Compensatory electrophysiological changes (eg, reduced potassium channel expression) appear to preserve twitch contraction parameters, but may expose patients to greater arrhythmic propensity and disease progression. Therapeutic approaches correcting the primary sarcomeric defects may prevent secondary cardiomyocyte remodeling.


Asunto(s)
Cardiomiopatía Hipertrófica , Células Madre Pluripotentes Inducidas , Humanos , Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Cardiomiopatía Hipertrófica/patología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Mutación , Calcio de la Dieta/metabolismo , Proteínas del Citoesqueleto/genética
2.
J Physiol ; 602(5): 791-808, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38348881

RESUMEN

T-tubules (TT) form a complex network of sarcolemmal membrane invaginations, essential for well-co-ordinated excitation-contraction coupling (ECC) and thus homogeneous mechanical activation of cardiomyocytes. ECC is initiated by rapid depolarization of the sarcolemmal membrane. Whether TT membrane depolarization is active (local generation of action potentials; AP) or passive (following depolarization of the outer cell surface sarcolemma; SS) has not been experimentally validated in cardiomyocytes. Based on the assessment of ion flux pathways needed for AP generation, we hypothesize that TT are excitable. We therefore explored TT excitability experimentally, using an all-optical approach to stimulate and record trans-membrane potential changes in TT that were structurally disconnected, and hence electrically insulated, from the SS membrane by transient osmotic shock. Our results establish that cardiomyocyte TT can generate AP. These AP show electrical features that differ substantially from those observed in SS, consistent with differences in the density of ion channels and transporters in the two different membrane domains. We propose that TT-generated AP represent a safety mechanism for TT AP propagation and ECC, which may be particularly relevant in pathophysiological settings where morpho-functional changes reduce the electrical connectivity between SS and TT membranes. KEY POINTS: Cardiomyocytes are characterized by a complex network of membrane invaginations (the T-tubular system) that propagate action potentials to the core of the cell, causing uniform excitation-contraction coupling across the cell. In the present study, we investigated whether the T-tubular system is able to generate action potentials autonomously, rather than following depolarization of the outer cell surface sarcolemma. For this purpose, we developed a fully optical platform to probe and manipulate the electrical dynamics of subcellular membrane domains. Our findings demonstrate that T-tubules are intrinsically excitable, revealing distinct characteristics of self-generated T-tubular action potentials. This active electrical capability would protect cells from voltage drops potentially occurring within the T-tubular network.


Asunto(s)
Miocitos Cardíacos , Optogenética , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Membrana Celular , Potenciales de la Membrana , Potenciales de Acción/fisiología
3.
J Mol Cell Cardiol ; 172: 120-131, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36058298

RESUMEN

Hypertrophic cardiomyopathy (HCM) patients often present an enhanced arrhythmogenicity that can lead to lethal arrhythmias, especially during exercise. Recent studies have indicated an abnormal response of HCM cardiomyocytes to ß-adrenergic receptor stimulation (ß-ARS), with prolongation of their action potential rather than shortening. The mechanisms underlying this aberrant response to sympathetic stimulation and its possible proarrhythmic role remain unknown. The aims of this study are to investigate the key ionic mechanisms underlying the HCM abnormal response to ß-ARS and the resultant repolarisation abnormalities using human-based experimental and computational methodologies. We integrated and calibrated the latest models of human ventricular electrophysiology and ß-ARS using experimental measurements of human adult cardiomyocytes from control and HCM patients. Our major findings include: (1) the developed in silico models of ß-ARS capture the behaviour observed in the experimental data, including the aberrant response of HCM cardiomyocytes to ß-ARS; (2) the reduced increase of potassium currents under ß-ARS was identified as the main mechanism of action potential prolongation in HCM, rather than a more sustained inward calcium current; (3) action potential duration differences between healthy and HCM cardiomyocytes were increased upon ß-ARS, while endocardial to epicardial differences in HCM cardiomyocytes were reduced; (4) models presenting repolarisation abnormalities were characterised by downregulation of the rapid delayed rectifier potassium current and the sodium­potassium pump, while inward currents were upregulated. In conclusion, our results identify causal relationships between the HCM phenotype and its arrhythmogenic response to ß-ARS through the downregulation of potassium currents.


Asunto(s)
Cardiomiopatía Hipertrófica , Potasio , Adulto , Humanos , Potenciales de Acción/fisiología , Adrenérgicos , Miocitos Cardíacos , Arritmias Cardíacas , Receptores Adrenérgicos beta
4.
J Muscle Res Cell Motil ; 42(2): 305-322, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33222034

RESUMEN

The highly organized transverse T-tubule membrane system represents the ultrastructural substrate for excitation-contraction coupling in ventricular myocytes. While the architecture and function of T-tubules have been well described in animal models, there is limited morpho-functional data on T-tubules in human myocardium. Hypertrophic cardiomyopathy (HCM) is a primary disease of the heart muscle, characterized by different clinical presentations at the various stages of its progression. Most HCM patients, indeed, show a compensated hypertrophic disease ("non-failing hypertrophic phase"), with preserved left ventricular function, and only a small subset of individuals evolves into heart failure ("end stage HCM"). In terms of T-tubule remodeling, the "end-stage" disease does not differ from other forms of heart failure. In this review we aim to recapitulate the main structural features of T-tubules during the "non-failing hypertrophic stage" of human HCM by revisiting data obtained from human myectomy samples. Moreover, by comparing pathological changes observed in myectomy samples with those introduced by acute (experimentally induced) detubulation, we discuss the role of T-tubular disruption as a part of the complex excitation-contraction coupling remodeling process that occurs during disease progression. Lastly, we highlight how T-tubule morpho-functional changes may be related to patient genotype and we discuss the possibility of a primitive remodeling of the T-tubule system in rare HCM forms associated with genes coding for proteins implicated in T-tubule structural integrity, formation and maintenance.


Asunto(s)
Cardiomiopatía Hipertrófica , Sarcolema , Animales , Cardiomiopatía Hipertrófica/genética , Acoplamiento Excitación-Contracción , Humanos , Miocardio , Miocitos Cardíacos
5.
Circ Res ; 124(8): e44-e54, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30732554

RESUMEN

RATIONALE: Despite major advances in cardiovascular medicine, heart disease remains a leading cause of death worldwide. However, the field of tissue engineering has been growing exponentially in the last decade and restoring heart functionality is now an affordable target; yet, new materials are still needed for effectively provide rapid and long-lasting interventions. Liquid crystalline elastomers (LCEs) are biocompatible polymers able to reversibly change shape in response to a given stimulus and generate movement. Once stimulated, LCEs can produce tension or movement like a muscle. However, so far their application in biology was limited by slow response times and a modest possibility to modulate tension levels during activation. OBJECTIVE: To develop suitable LCE-based materials to assist cardiac contraction. METHODS AND RESULTS: Thanks to a quick, simple, and versatile synthetic approach, a palette of biocompatible acrylate-based light-responsive LCEs with different molecular composition was prepared and mechanically characterized. Out of this, the more compliant one was selected. This material was able to contract for some weeks when activated with very low light intensity within a physiological environment. Its contraction was modulated in terms of light intensity, stimulation frequency, and ton/toff ratio to fit different contraction amplitude/time courses, including those of the human heart. Finally, LCE strips were mounted in parallel with cardiac trabeculae, and we demonstrated their ability to improve muscular systolic function, with no impact on diastolic properties. CONCLUSIONS: Our results indicated LCEs are promising in assisting cardiac mechanical function and developing a new generation of contraction assist devices.


Asunto(s)
Materiales Biocompatibles , Elastómeros , Corazón Auxiliar , Luz , Cristales Líquidos , Contracción Miocárdica , Ingeniería de Tejidos/métodos , Acrilatos , Órganos Bioartificiales , Materiales Biocompatibles/síntesis química , Fenómenos Biofísicos , Reactivos de Enlaces Cruzados/química , Elastómeros/síntesis química , Transferencia de Energía , Cristales Líquidos/química , Sistemas Microelectromecánicos/métodos , Movimientos de los Órganos , Factores de Tiempo , Andamios del Tejido/química
6.
Pharmacol Res ; 168: 105581, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33781873

RESUMEN

In-depth characterization of heart-brain communication in critically ill patients with severe acute respiratory failure is attracting significant interest in the COronaVIrus Disease 19 (COVID-19) pandemic era during intensive care unit (ICU) stay and after ICU or hospital discharge. Emerging research has provided new insights into pathogenic role of the deregulation of the heart-brain axis (HBA), a bidirectional flow of information, in leading to severe multiorgan disease syndrome (MODS) in patients with confirmed infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Noteworthy, HBA dysfunction may worsen the outcome of the COVID-19 patients. In this review, we discuss the critical role HBA plays in both promoting and limiting MODS in COVID-19. We also highlight the role of HBA as new target for novel therapeutic strategies in COVID-19 in order to open new translational frontiers of care. This is a translational perspective from the Italian Society of Cardiovascular Researches.


Asunto(s)
Encefalopatías/terapia , Encéfalo/efectos de los fármacos , COVID-19/terapia , Cardiopatías/terapia , Corazón/efectos de los fármacos , Corticoesteroides/administración & dosificación , Antiinflamatorios/administración & dosificación , Antivirales/administración & dosificación , Encéfalo/inmunología , Encéfalo/metabolismo , Encefalopatías/inmunología , Encefalopatías/metabolismo , COVID-19/inmunología , COVID-19/metabolismo , Cuidados Críticos/métodos , Enfermedad Crítica/terapia , Suplementos Dietéticos , Alimentos Funcionales , Cardiopatías/inmunología , Cardiopatías/metabolismo , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Microvasos/efectos de los fármacos , Microvasos/inmunología , Microvasos/metabolismo , Insuficiencia Multiorgánica/inmunología , Insuficiencia Multiorgánica/metabolismo , Insuficiencia Multiorgánica/terapia , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo
7.
BMC Fam Pract ; 22(1): 208, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34666689

RESUMEN

BACKGROUND: Polypharmacy is defined as the prescription of at least 5 different medicines for therapeutic or prophylactic effect and is a serious issue among elderly patients, who are frequently affected by multi-morbidity. Deprescribing is one of the proposed approaches to reduce the number of administered drugs, by eliminating those that are inappropriately prescribed. The aim of this systematic review is to provide an updated and systematic assessment of the benefit-risk profile of deprescribing of anti-hypertensive drugs, which are among the most commonly used drugs. METHODS: MEDLINE, EMBASE and The Cochrane Library were searched for studies assessing the efficacy and safety of anti-hypertensive drugs deprescribing in the period between January, 12,016 and December, 312,019. The quality of randomized clinical trials (RCTs) was assessed using the GRADE approach for the evaluation of the main outcomes. The risk of bias assessment was carried out using the Cochrane risk-of-bias tool. RESULTS: Overall, two RCTs were identified. Despite summarized evidence was in favor of anti-hypertensive deprescribing, the overall risk of bias was rated as high for each RCT included. According to the GRADE approach, the overall quality of the RCTs included was moderate regarding the following outcomes: systolic blood pressure < 150 mmHg after 12 weeks of follow-up, quality of life, frailty and cardiovascular risk. CONCLUSIONS: This updated systematic review of the efficacy and safety of anti-hypertensive treatment deprescribing found two recently published RCTs, in addition to the previous guideline of the National Institute for Health and Care Excellence (NICE). Evidence points towards non-inferiority of anti-hypertensive deprescribing as compared to treatment continuation, despite the quality of published studies is not high. High quality experimental studies are urgently needed to further assess the effect of deprescribing for this drug class in specific categories of patients.


Asunto(s)
Antihipertensivos , Deprescripciones , Anciano , Antihipertensivos/efectos adversos , Presión Sanguínea , Humanos , Polifarmacia , Calidad de Vida
8.
Curr Heart Fail Rep ; 18(4): 169-179, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34148184

RESUMEN

PURPOSE OF REVIEW: We provide a state of the art of therapeutic options in hypertrophic cardiomyopathy (HCM), focusing on recent advances in our understanding of the pathophysiology of sarcomeric disease. RECENT FINDINGS: A wealth of novel information regarding the molecular mechanisms associated with the clinical phenotype and natural history of HCM have been developed over the last two decades. Such advances have only recently led to a number of controlled randomized studies, often limited in size and fortune. Recently, however, the allosteric inhibitors of cardiac myosin adenosine triphosphatase, countering the main pathophysiological abnormality associated with HCM-causing mutations, i.e. hypercontractility, have opened new management perspectives. Mavacamten is the first drug specifically developed for HCM used in a successful phase 3 trial, with the promise to reach symptomatic obstructive patients in the near future. In addition, the fine characterization of cardiomyocyte electrophysiological remodelling has recently highlighted relevant therapeutic targets. Current therapies for HCM focus on late disease manifestations without addressing the intrinsic pathological mechanisms. However, novel evidence-based approaches have opened the way for agents targeting HCM molecular substrates. The impact of these targeted interventions will hopefully alter the natural history of the disease in the near future.


Asunto(s)
Cardiomiopatía Hipertrófica , Insuficiencia Cardíaca , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/terapia , Humanos , Mutación , Miocitos Cardíacos , Proyectos de Investigación
9.
Pharmacol Res ; 160: 105176, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32871247

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the commonest genetic cardiac disease, with a prevalence of 1/500. It is caused by over 1400 different mutations, mainly involving the genes coding for sarcomere proteins. The main pathological features of HCM are left ventricular hypertrophy, diastolic dysfunction and the increased ventricular arrhythmogenesis. Predicting the risk of heart failure and lethal arrhythmias is the most challenging clinical task for HCM patient management. Moreover, there are no disease-modifying therapies that can prevent disease progression or sudden arrhythmic death in HCM patients. In this review, we will illustrate the most advanced research models and methods that have been employed for HCM studies, including preclinical tests of novel or existing drugs, along with visionary future development based on gene editing approaches. Acknowledging the advantages and limitations of the different models, and a critical consideration of the different, often conflicting result obtained using different approaches is essential for a deep understanding of HCM pathophysiology and for obtaining meaningful information on novel treatments, in order to improve patient risk stratification and therapeutic management.


Asunto(s)
Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/genética , Desarrollo de Medicamentos , Enfermedades Genéticas Congénitas/tratamiento farmacológico , Enfermedades Genéticas Congénitas/genética , Animales , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Humanos , Modelos Biológicos
10.
Pharmacol Res ; 157: 104781, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32360273

RESUMEN

The results of trials with sodium-glucose cotransporter 2 (SGLT2) inhibitors raised the possibility that this class of drugs provides cardiovascular benefits independently from their anti-diabetic effects, although the mechanisms are unknown. Therefore, we tested the effects of SGLT2 inhibitor dapagliflozin on the progression of experimental heart disease in a non-diabetic model of heart failure with preserved ejection fraction. Dahl salt-sensitive rats were fed a high-salt diet to induce hypertension and diastolic dysfunction and were then treated with dapagliflozin for six weeks. Dapagliflozin ameliorated diastolic function as documented by echo-Doppler and heart catheterization, while blood pressure remained markedly elevated. Chronic in vivo treatment with dapagliflozin reduced diastolic Ca2+ and Na+ overload and increased Ca2+ transient amplitude in ventricular cardiomyocytes, although no direct action of dapagliflozin on isolated cardiomyocytes was observed. Dapagliflozin reversed endothelial activation and endothelial nitric oxide synthase deficit, with reduced cardiac inflammation and consequent attenuation of pro-fibrotic signaling. The potential involvement of coronary endothelium was supported by the endothelial upregulation of Na+/H+ exchanger 1in vivo and direct effects on dapagliflozin on the activity of this exchanger in endothelial cells in vitro. In conclusions, several mechanisms may cumulatively play a significant role in the dapagliflozin-associated cardioprotection. Dapagliflozin ameliorates diastolic function and exerts a positive effect on the myocardium, possibly targeting coronary endothelium. The lower degree of endothelial dysfunction, inflammation and fibrosis translate into improved myocardial performance.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Vasos Coronarios/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Glucósidos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Animales , Señalización del Calcio , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Diástole , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas Endogámicas Dahl , Sodio/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
11.
Proc Natl Acad Sci U S A ; 114(43): E9006-E9015, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073097

RESUMEN

The mitochondrial Ca2+ uniporter complex (MCUC) is a multimeric ion channel which, by tuning Ca2+ influx into the mitochondrial matrix, finely regulates metabolic energy production. In the heart, this dynamic control of mitochondrial Ca2+ uptake is fundamental for cardiomyocytes to adapt to either physiologic or pathologic stresses. Mitochondrial calcium uniporter (MCU), which is the core channel subunit of MCUC, has been shown to play a critical role in the response to ß-adrenoreceptor stimulation occurring during acute exercise. The molecular mechanisms underlying the regulation of MCU, in conditions requiring chronic increase in energy production, such as physiologic or pathologic cardiac growth, remain elusive. Here, we show that microRNA-1 (miR-1), a member of the muscle-specific microRNA (myomiR) family, is responsible for direct and selective targeting of MCU and inhibition of its translation, thereby affecting the capacity of the mitochondrial Ca2+ uptake machinery. Consistent with the role of miR-1 in heart development and cardiomyocyte hypertrophic remodeling, we additionally found that MCU levels are inversely related with the myomiR content, in murine and, remarkably, human hearts from both physiologic (i.e., postnatal development and exercise) and pathologic (i.e., pressure overload) myocardial hypertrophy. Interestingly, the persistent activation of ß-adrenoreceptors is likely one of the upstream repressors of miR-1 as treatment with ß-blockers in pressure-overloaded mouse hearts prevented its down-regulation and the consequent increase in MCU content. Altogether, these findings identify the miR-1/MCU axis as a factor in the dynamic adaptation of cardiac cells to hypertrophy.


Asunto(s)
Canales de Calcio/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Aorta/citología , Canales de Calcio/genética , Cardiomegalia/metabolismo , Metabolismo Energético , Humanos , Ratones , MicroARNs/genética , Condicionamiento Físico Animal , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/metabolismo
12.
Genet Med ; 21(2): 284-292, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29875424

RESUMEN

PURPOSE: Genetic testing in hypertrophic cardiomyopathy (HCM) has long relied on Sanger sequencing of sarcomeric genes. The advent of next-generation sequencing (NGS) has catalyzed routine testing of additional genes of dubious HCM-causing potential. We used 19 years of genetic testing results to define a reliable set of genes implicated in Mendelian HCM and assess the value of expanded NGS panels. METHODS: We dissected genetic testing results from 1,198 single-center HCM probands and devised a widely applicable score to identify which genes yield effective results in the diagnostic setting. RESULTS: Compared with early panels targeting only fully validated sarcomeric HCM genes, expanded NGS panels allow the prompt recognition of probands with HCM-mimicking diseases. Scoring by "diagnostic effectiveness" highlighted that PLN should also be routinely screened besides historically validated genes for HCM and its mimics. CONCLUSION: The additive value of expanded panels in HCM genetic testing lies in the systematic screening of genes associated with HCM mimics, requiring different patient management. Only variants in a limited set of genes are highly actionable and interpretable in the clinic, suggesting that larger panels offer limited additional sensitivity. A score estimating the relative effectiveness of a given gene's inclusion in diagnostic panels is proposed.


Asunto(s)
Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/genética , Pruebas Genéticas , Adulto , Anciano , Estudios de Cohortes , Femenino , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Sarcómeros/genética , Adulto Joven
13.
Int J Mol Sci ; 20(15)2019 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-31382622

RESUMEN

Cardiomyocytes from human induced pluripotent stem cells (hiPSC-CMs) are the most promising human source with preserved genetic background of healthy individuals or patients. This study aimed to establish a systematic procedure for exploring development of hiPSC-CM functional output to predict genetic cardiomyopathy outcomes and identify molecular targets for therapy. Biomimetic substrates with microtopography and physiological stiffness can overcome the immaturity of hiPSC-CM function. We have developed a custom-made apparatus for simultaneous optical measurements of hiPSC-CM action potential and calcium transients to correlate these parameters at specific time points (day 60, 75 and 90 post differentiation) and under inotropic interventions. In later-stages, single hiPSC-CMs revealed prolonged action potential duration, increased calcium transient amplitude and shorter duration that closely resembled those of human adult cardiomyocytes from fresh ventricular tissue of patients. Thus, the major contribution of sarcoplasmic reticulum and positive inotropic response to ß-adrenergic stimulation are time-dependent events underlying excitation contraction coupling (ECC) maturation of hiPSC-CM; biomimetic substrates can promote calcium-handling regulation towards adult-like kinetics. Simultaneous optical recordings of long-term cultured hiPSC-CMs on biomimetic substrates favor high-throughput electrophysiological analysis aimed at testing (mechanistic hypothesis on) disease progression and pharmacological interventions in patient-derived hiPSC-CMs.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Calcio/metabolismo , Cardiomiopatías/tratamiento farmacológico , Células Madre Pluripotentes Inducidas/metabolismo , Potenciales de Acción/efectos de los fármacos , Biomimética , Cardiomiopatías/genética , Cardiomiopatías/patología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Acoplamiento Excitación-Contracción/efectos de los fármacos , Humanos , Hidrogeles/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Especificidad por Sustrato
14.
Heart Fail Clin ; 14(2): 161-178, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29525645

RESUMEN

Cardiomyopathies are diseases of the myocardium, often genetically determined, associated with heterogeneous phenotypes and clinical manifestations. Despite significant progress in the understanding of these conditions, available treatments mostly target late complications, whereas approaches that promise to interfere with the primary mechanisms and natural history are just beginning to surface. The last decade has witnessed the establishment of large international cardiomyopathy registries, paralleled by advances in cardiac imaging and genetic testing, deeper understanding of the pathophysiology and growing involvement by the pharmaceutical industry. As a result, the number of molecular interventions under scrutiny is increasing sharply.


Asunto(s)
Cardiomiopatías , Pruebas Genéticas/métodos , Terapia Genética/métodos , Miocardio/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/terapia , Ensayos Clínicos como Asunto/métodos , Humanos
15.
J Physiol ; 595(12): 3815-3822, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27981580

RESUMEN

Electrical excitability is an essential feature of cardiomyocytes and the homogenous propagation of the action potential is guaranteed by a complex network of membrane invaginations called the transverse-axial tubular system (TATS). TATS structural remodelling is a hallmark of cardiac diseases and we demonstrated that this can be accompanied by electrical defects at single T-tubular level. Using a random-access multi-photon (RAMP) microscope, we found that pathological T-tubules can fail to conduct action potentials, which delays local Ca2+ release. Although the underlying causes for T-tubular electrical failure are still unknown, our findings suggest that they are likely to be related to local ultrastructural alterations. Here, we first review the experimental approach that allowed us to observe and dissect the consequences of TATS electrical dysfunction and then propose two different strategies to unveil the reasons for T-tubular electrical failures. The first strategy consists in a correlative approach, in which the failing T-tubule identified with the RAMP microscope is then imaged with electron microscopy. The second approach exploits the diffusion of molecules within TATS to gain insights into the local TATS structure, even without a thorough reconstruction of the tubular network. Although challenging, the local electrical failure occurring at single T-tubules is a fundamental question that needs to be addressed and could provide novel insights in cardiac pathophysiology.


Asunto(s)
Cardiopatías/fisiopatología , Corazón/fisiopatología , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Cardiopatías/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología
16.
Small ; 13(46)2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29045016

RESUMEN

The communication reports the use of liquid crystalline networks (LCNs) for engineering tissue cultures with human cells. Their ability as cell scaffolds for different cell lines is demonstrated. Preliminary assessments of the material biocompatibility are performed on human dermal fibroblasts and murine muscle cells (C2C12), demonstrating that coatings or other treatments are not needed to use the acrylate-based materials as support. Moreover, it is found that adherent C2C12 cells undergo differentiation, forming multinucleated myotubes, which show the typical elongated shape, and contain bundles of stress fibers. Once biocompatibility is demonstrated, the same LCN films are used as a substrate for culturing human induced pluripotent stem cell-derived cardiomyocites (hiPSC-CMs) proving that LCNs are capable to develop adult-like dimensions and a more mature cell function in a short period of culture in respect to standard supports. The demonstrated biocompatibility together with the extraordinary features of LCNs opens to preparation of complex cell scaffolds, both patterned and stimulated, for dynamic cell culturing. The ability of these materials to improve cell maturation and differentiation will be developed toward engineered heart and skeletal muscular tissues exploring regenerative medicine toward bioartificial muscles for injured sites replacement.


Asunto(s)
Cristales Líquidos/química , Medicina Regenerativa , Cicatrización de Heridas , Animales , Adhesión Celular , Diferenciación Celular , Línea Celular , Proliferación Celular , Fibroblastos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Miocitos Cardíacos/citología
17.
Proc Natl Acad Sci U S A ; 111(42): 15196-201, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288764

RESUMEN

Action potentials (APs), via the transverse axial tubular system (TATS), synchronously trigger uniform Ca(2+) release throughout the cardiomyocyte. In heart failure (HF), TATS structural remodeling occurs, leading to asynchronous Ca(2+) release across the myocyte and contributing to contractile dysfunction. In cardiomyocytes from failing rat hearts, we previously documented the presence of TATS elements which failed to propagate AP and displayed spontaneous electrical activity; the consequence for Ca(2+) release remained, however, unsolved. Here, we develop an imaging method to simultaneously assess TATS electrical activity and local Ca(2+) release. In HF cardiomyocytes, sites where T-tubules fail to conduct AP show a slower and reduced local Ca(2+) transient compared with regions with electrically coupled elements. It is concluded that TATS electrical remodeling is a major determinant of altered kinetics, amplitude, and homogeneity of Ca(2+) release in HF. Moreover, spontaneous depolarization events occurring in failing T-tubules can trigger local Ca(2+) release, resulting in Ca(2+) sparks. The occurrence of tubule-driven depolarizations and Ca(2+) sparks may contribute to the arrhythmic burden in heart failure.


Asunto(s)
Calcio/metabolismo , Insuficiencia Cardíaca/metabolismo , Células Musculares/citología , Miocitos Cardíacos/metabolismo , Potenciales de Acción/fisiología , Animales , Señalización del Calcio/fisiología , Células Cultivadas , Proteínas Fluorescentes Verdes/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Masculino , Contracción Miocárdica/fisiología , Ratas , Ratas Wistar , Retículo Sarcoplasmático/metabolismo
18.
J Mol Cell Cardiol ; 96: 72-81, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26385634

RESUMEN

INTRODUCTION: Hypertrophic cardiomyopathy (HCM) is a cause of sudden arrhythmic death, but the understanding of its pro-arrhythmic mechanisms and an effective pharmacological treatment are lacking. HCM electrophysiological remodelling includes both increased inward and reduced outward currents, but their role in promoting repolarisation abnormalities remains unknown. The goal of this study is to identify key ionic mechanisms driving repolarisation abnormalities in human HCM, and to evaluate anti-arrhythmic effects of single and multichannel inward current blocks. METHODS: Experimental ionic current, action potential (AP) and Ca(2+)-transient (CaT) recordings were used to construct populations of human non-diseased and HCM AP models (n=9118), accounting for inter-subject variability. Simulations were conducted for several degrees of selective and combined inward current block. RESULTS: Simulated HCM cardiomyocytes exhibited prolonged AP and CaT, diastolic Ca(2+) overload and decreased CaT amplitude, in agreement with experiments. Repolarisation abnormalities in HCM models were consistently driven by L-type Ca(2+) current (ICaL) re-activation, and ICaL block was the most effective intervention to normalise repolarisation and diastolic Ca(2+), but compromised CaT amplitude. Late Na(+) current (INaL) block partially abolished repolarisation abnormalities, with small impact on CaT. Na(+)/Ca(2+) exchanger (INCX) block effectively restored repolarisation and CaT amplitude, but increased Ca(2+) overload. Multichannel block increased efficacy in normalising repolarisation, AP biomarkers and CaT amplitude compared to selective block. CONCLUSIONS: Experimentally-calibrated populations of human AP models identify ICaL re-activation as the key mechanism for repolarisation abnormalities in HCM, and combined INCX, INaL and ICaL block as effective anti-arrhythmic therapies also able to partially reverse the HCM electrophysiological phenotype.


Asunto(s)
Potenciales de Acción , Antiarrítmicos/farmacología , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/metabolismo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Potenciales de Acción/efectos de los fármacos , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo L/metabolismo , Fenómenos Electrofisiológicos/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
19.
Eur J Nucl Med Mol Imaging ; 43(13): 2413-2422, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27527796

RESUMEN

AIMS: Myocardial blood flow <1.1 mL/min/g following dipyridamole (Dip-MBF) assessed by positron emission tomography (PET) was identified in 2003 as an important outcome predictor in hypertrophic cardiomyopathy (HCM), based on scans performed in the 90s. However, such extreme Dip-MBF impairment is rarely observed in contemporary cohorts. We, therefore, reassessed the Dip-MBF threshold defining high-risk HCM patients. METHODS: Dip-MBF was measured using 13N-ammonia in 100 HCM consecutive patients, prospectively enrolled and followed for 4.0 ± 2.2 years. Outcome was assessed based on tertiles of Dip-MBF. The study end-point was a combination of cardiovascular death, progression to severe functional limitation, cardioembolic stroke, life-threatening ventricular arrhythmias. RESULTS: Global Dip-MBF was 1.95 ± 0.85, ranging from 0.7 to 5.9 mL/min/g. Dip-MBF tertile cut-off values were: 0.73 to 1.53 mL/min/g (lowest), 1.54 to 2.13 mL/min/g (middle), and 2.14 to 5.89 mL/min/g (highest). During follow-up, lowest tertile Dip-MBF was associated with sevenfold independent risk of unfavorable outcome compared to the other two tertiles. Dip-MBF 1.35 mL/min/g was identified as the best threshold for outcome prediction. Regional perfusion analysis showed that all cardiac deaths (n = 4) occurred in patients in the lowest tertile of lateral wall Dip-MBF (≤1.72 mL/min/g); septal Dip-MBF was not predictive. CONCLUSIONS: Dip-MBF confirms its role as potent predictor of outcome in HCM. However, the threshold for prediction in a contemporary cohort is higher than that reported in earlier studies. Dip-MBF impairment in the lateral wall, possibly reflecting diffuse disease extending to non-hypertrophic regions, is a sensitive predictor of mortality in HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/mortalidad , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Enfermedad de la Arteria Coronaria/mortalidad , Imagen de Perfusión/métodos , Tomografía de Emisión de Positrones/métodos , Adulto , Causalidad , Comorbilidad , Dipiridamol/administración & dosificación , Femenino , Humanos , Italia/epidemiología , Estudios Longitudinales , Masculino , Prevalencia , Reproducibilidad de los Resultados , Factores de Riesgo , Sensibilidad y Especificidad , Tasa de Supervivencia , Vasodilatadores/administración & dosificación
20.
Pharmacol Res ; 108: 80-87, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27142783

RESUMEN

To date, up to 65% of drugs used in neonates and infants are off-label or unlicensed, as they were implemented in clinical care without the usual regulatory phases of pharmacological drug development. Pharmacotherapy in this age group is still mainly based on the individual clinical expertise of specialized pediatricians. Pharmacological trials involving neonates are indeed more difficult to perform: appropriate dosing is hampered by the rapid physiological changes occurring at this stage of development, and the selection of proper end-points and biomarkers is complicated by the limited knowledge of the pathophysiology of the specific diseases of infancy. Moreover, there are many ethical challenges in planning and conducting drug studies in pediatric patients (especially in newborns and infants). In the current review, we address some challenges and discuss possible perspectives to stimulate scientific and clinical pharmacological research in neonates and infants. We hereby aim to illustrate the add on value of the regulatory framework for model-based neonatal medicinal development currently used in Europe and the United States. We provide several examples of successful recent pharmacological trials performed in neonates and infants. In these examples, success was ensured by the implementation of specific pharmacokinetic assessments, thanks to accurate drug dosing achieved with a combination of dose validation, population pharmacokinetics and mathematical models of drug clearance and distribution; moreover, age-specific pharmacodynamics was considered via appropriate evaluations of drug efficacy with end-points adapted to the peculiar pathophysiology of diseases in this age group. These "pharmacological" challenges add to the ethical challenges that are always present in planning and conducting clinical studies in neonates and infants and support the opinion that clinical research in pediatrics should be evaluated by ad hoc ethical committees with specific expertise.


Asunto(s)
Descubrimiento de Drogas/métodos , Pediatría/métodos , Investigación Farmacéutica/métodos , Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/uso terapéutico , Relación Dosis-Respuesta a Droga , Humanos , Lactante , Recién Nacido , Taquicardia Supraventricular/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA