Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Basic Res Cardiol ; 118(1): 43, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37801130

RESUMEN

Altered autonomic balance is a hallmark of numerous cardiovascular diseases, including myocardial infarction (MI). Although device-based vagal stimulation is cardioprotective during chronic disease, a non-invasive approach to selectively stimulate the cardiac parasympathetic system immediately after an infarction does not exist and is desperately needed. Cardiac vagal neurons (CVNs) in the brainstem receive powerful excitation from a population of neurons in the paraventricular nucleus (PVN) of the hypothalamus that co-release oxytocin (OXT) and glutamate to excite CVNs. We tested if chemogenetic activation of PVN-OXT neurons following MI would be cardioprotective. The PVN of neonatal rats was transfected with vectors to selectively express DREADDs within OXT neurons. At 6 weeks of age, an MI was induced and DREADDs were activated with clozapine-N-oxide. Seven days following MI, patch-clamp electrophysiology confirmed the augmented excitatory neurotransmission from PVN-OXT neurons to downstream nuclei critical for parasympathetic activity with treatment (43.7 ± 10 vs 86.9 ± 9 pA; MI vs. treatment), resulting in stark improvements in survival (85% vs. 95%; MI vs. treatment), inflammation, fibrosis assessed by trichrome blue staining, mitochondrial function assessed by Seahorse assays, and reduced incidence of arrhythmias (50% vs. 10% cumulative incidence of ventricular fibrillation; MI vs. treatment). Myocardial transcriptomic analysis provided molecular insight into potential cardioprotective mechanisms, which revealed the preservation of beneficial signaling pathways, including muscarinic receptor activation, in treated animals. These comprehensive results demonstrate that the PVN-OXT network could be a promising therapeutic target to quickly activate beneficial parasympathetic-mediated cellular pathways within the heart during the early stages of infarction.


Asunto(s)
Infarto del Miocardio , Oxitocina , Ratas , Animales , Oxitocina/farmacología , Oxitocina/metabolismo , Ratas Sprague-Dawley , Hipotálamo , Infarto del Miocardio/metabolismo , Neuronas/metabolismo , Arritmias Cardíacas/metabolismo
2.
J Biomed Opt ; 28(1): 016001, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36636698

RESUMEN

Significance: All-optical cardiac electrophysiology enables the visualization and control of key parameters relevant to the detection of cardiac arrhythmias. Mapping such responses in human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) is of great interest for cardiotoxicity and personalized medicine applications. Aim: We introduce and validate a very low-cost compact mapping system for macroscopic all-optical electrophysiology in layers of hiPSC-CMs. Approach: The system uses oblique transillumination, low-cost cameras, light-emitting diodes, and off-the-shelf components (total < $ 15 , 000 ) to capture voltage, calcium, and mechanical waves under electrical or optical stimulation. Results: Our results corroborate the equivalency of electrical and optogenetic stimulation of hiPSC-CMs, and V m - [ Ca 2 + ] i similarity in conduction under pacing. Green-excitable optical sensors are combinable with blue optogenetic actuators (chanelrhodopsin2) only under very low green light ( < 0.05 mW / mm 2 ). Measurements in warmer culture medium yield larger spread of action potential duration and higher conduction velocities compared to Tyrode's solution at room temperature. Conclusions: As multiple optical sensors and actuators are combined, our results can help handle the "spectral congestion" and avoid parameter distortion. We illustrate the utility of the system for uncovering the action of cellular uncoupling agents and show extensibility to an epi-illumination mode for future imaging of thicker native or engineered tissues.


Asunto(s)
Técnicas Electrofisiológicas Cardíacas , Células Madre Pluripotentes Inducidas , Humanos , Miocitos Cardíacos/fisiología , Arritmias Cardíacas , Potenciales de Acción
3.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 6060-6063, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30441718

RESUMEN

Therapeutic ultrasound presents a potential novel treatment for type 2 diabetes mellitus that utilizes the non-invasive application of ultrasound energy to treat secretory defects in the earlier stages of the disease. Our previous studies have shown that ultrasound is capable of stimulating insulin release from pancreatic beta cells, safely and effectively. This study aims to both examine the calcium-dependent mechanisms of ultrasound-mediated insulin release from pancreatic beta cells using three complementary modalities - carbon fiber amperometry, ELISA studies, and Ca2+ fluorescence imaging - and to study the translational potential of therapeutic ultrasound using transgenic hyperglycemic mice for safety and efficacy studies.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Calcio , Glucosa , Insulina , Secreción de Insulina , Ratones
4.
Nat Commun ; 7: 11542, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27161419

RESUMEN

The improvement of preclinical cardiotoxicity testing, discovery of new ion-channel-targeted drugs, and phenotyping and use of stem cell-derived cardiomyocytes and other biologics all necessitate high-throughput (HT), cellular-level electrophysiological interrogation tools. Optical techniques for actuation and sensing provide instant parallelism, enabling contactless dynamic HT testing of cells and small-tissue constructs, not affordable by other means. Here we show, computationally and experimentally, the limits of all-optical electrophysiology when applied to drug testing, then implement and validate OptoDyCE, a fully automated system for all-optical cardiac electrophysiology. We validate optical actuation by virally introducing optogenetic drivers in rat and human cardiomyocytes or through the modular use of dedicated light-sensitive somatic 'spark' cells. We show that this automated all-optical approach provides HT means of cellular interrogation, that is, allows for dynamic testing of >600 multicellular samples or compounds per hour, and yields high-content information about the action of a drug over time, space and doses.


Asunto(s)
Técnicas Electrofisiológicas Cardíacas/métodos , Optogenética/métodos , Animales , Automatización , Cardiotoxinas/toxicidad , Células Cultivadas , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Nifedipino/administración & dosificación , Nifedipino/toxicidad , Ratas
5.
J Physiol ; 594(9): 2503-10, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26857427

RESUMEN

Cardiac tissue is an excitable system that can support complex spatiotemporal dynamics, including instabilities (arrhythmias) with lethal consequences. While over the last two decades optical mapping of excitation (voltage and calcium dynamics) has facilitated the detailed characterization of such arrhythmia events, until recently, no precise tools existed to actively interrogate cardiac dynamics in space and time. In this work, we discuss the combined use of new methods for space- and time-resolved optogenetic actuation and simultaneous fast, high resolution optical imaging of cardiac excitation waves. First, the mechanisms, limitations and unique features of optically induced responses in cardiomyocytes are outlined. These include the ability to bidirectionally control the membrane potential using depolarizing and hyperpolarizing opsins; the ability to induce prolonged sustained voltage changes; and the ability to control refractoriness and the shape of the cardiac action potential. At the syncytial tissue level, we discuss optogenetically enabled experimentation on cell-cell coupling, alteration of conduction properties and termination of propagating waves by light. Specific attention is given to space- and time-resolved application of optical stimulation using dynamic light patterns to perturb ongoing activation and to probe electrophysiological properties at desired tissue locations. The combined use of optical methods to perturb and to observe the system can offer new tools for precise feedback control of cardiac electrical activity, not available previously with pharmacological and electrical stimulation. These new experimental tools for all-optical electrophysiology allow for a level of precise manipulation and quantification of cardiac dynamics comparable in robustness to the computational setting, and can provide new insights into pacemaking, arrhythmogenesis and suppression or cardioversion.


Asunto(s)
Técnicas Electrofisiológicas Cardíacas , Corazón/fisiología , Optogenética , Animales , Humanos
6.
Sci Rep ; 5: 16542, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26568132

RESUMEN

All-trans-Retinal (ATR) is a photosensitizer, serving as the chromophore for depolarizing and hyperpolarizing light-sensitive ion channels and pumps (opsins), recently employed as fast optical actuators. In mammalian optogenetic applications (in brain and heart), endogenous ATR availability is not considered a limiting factor, yet it is unclear how ATR modulation may affect the response to optical stimulation. We hypothesized that exogenous ATR may improve light responsiveness of cardiac cells modified by Channelrhodopsin2 (ChR2), hence lowering the optical pacing energy. In virally-transduced (Ad-ChR2(H134R)-eYFP) light-sensitive cardiac syncytium in vitro, ATR supplements ≤2 µM improved cardiomyocyte viability and augmented ChR2 membrane expression several-fold, while >4 µM was toxic. Employing integrated optical actuation (470 nm) and optical mapping, we found that 1-2 µM ATR dramatically reduced optical pacing energy (over 30 times) to several µW/mm(2), lowest values reported to date, but also caused action potential prolongation, minor changes in calcium transients and no change in conduction. Theoretical analysis helped explain ATR-caused reduction of optical excitation threshold in cardiomyocytes. We conclude that cardiomyocytes operate at non-saturating retinal levels, and carefully-dosed exogenous ATR can enhance the performance of ChR2 in cardiac cells and yield energy benefits over orders of magnitude for optogenetic stimulation.


Asunto(s)
Miocitos Cardíacos/fisiología , Optogenética , Fármacos Fotosensibilizantes/farmacología , Retinaldehído/farmacología , Potenciales de Acción , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Channelrhodopsins , Células Gigantes/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ratas Sprague-Dawley
7.
Prog Biophys Mol Biol ; 115(2-3): 294-304, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25035999

RESUMEN

In complex multicellular systems, such as the brain or the heart, the ability to selectively perturb and observe the response of individual components at the cellular level and with millisecond resolution in time, is essential for mechanistic understanding of function. Optogenetics uses genetic encoding of light sensitivity (by the expression of microbial opsins) to provide such capabilities for manipulation, recording, and control by light with cell specificity and high spatiotemporal resolution. As an optical approach, it is inherently scalable for remote and parallel interrogation of biological function at the tissue level; with implantable miniaturized devices, the technique is uniquely suitable for in vivo tracking of function, as illustrated by numerous applications in the brain. Its expansion into the cardiac area has been slow. Here, using examples from published research and original data, we focus on optogenetics applications to cardiac electrophysiology, specifically dealing with the ability to manipulate membrane voltage by light with implications for cardiac pacing, cardioversion, cell communication, and arrhythmia research, in general. We discuss gene and cell delivery methods of inscribing light sensitivity in cardiac tissue, functionality of the light-sensitive ion channels within different types of cardiac cells, utility in probing electrical coupling between different cell types, approaches and design solutions to all-optical electrophysiology by the combination of optogenetic sensors and actuators, and specific challenges in moving towards in vivo cardiac optogenetics.


Asunto(s)
Potenciales de Acción/fisiología , Técnicas Electrofisiológicas Cardíacas/métodos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Optogenética/métodos , Imagen de Colorante Sensible al Voltaje/métodos , Animales , Técnicas Electrofisiológicas Cardíacas/instrumentación , Humanos , Optogenética/instrumentación , Imagen de Colorante Sensible al Voltaje/instrumentación
8.
Expert Rev Cardiovasc Ther ; 12(1): 17-20, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24308809

RESUMEN

Cardiac optogenetics is an exciting new methodology in which light-sensitive ion channels are expressed in heart tissue to enable optical control of bioelectricity. This technology has the potential to open new avenues for safely and effectively treating rhythm disorders in the heart with gentle beams of light. Recently, we developed a comprehensive framework for modeling cardiac optogenetics. Simulations conducted in this platform will provide insights to guide in vitro investigation and steer the development of therapeutic applications - these are the first steps toward clinical translation. In this editorial, we review literature relevant to light-sensitive protein delivery and intracardiac illumination to provide a holistic feasibility assessment for optogenetics-based arrhythmia termination therapy. We then draw on examples from computational work to show that the optical control paradigm has undeniable advantages that cannot be attained with conventional electrotherapy. Hence, we argue that cardiac optogenetics is more than a flashy substitute for current approaches.


Asunto(s)
Cardiopatías/terapia , Corazón , Canales Iónicos/fisiología , Luz , Optogenética , Corazón/fisiología , Corazón/fisiopatología , Cardiopatías/fisiopatología , Humanos , Proteínas
9.
Circ Arrhythm Electrophysiol ; 5(4): 831-40, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22722661

RESUMEN

BACKGROUND: In depolarized myocardial infarct epicardial border zones, the cardiac sodium channel is largely inactivated, contributing to slow conduction and reentry. We have demonstrated that adenoviral delivery of the skeletal muscle Na(+) channel (SkM1) to epicardial border zones normalizes conduction and reduces induction of ventricular tachycardia/ventricular fibrillation. We now studied the impact of canine mesenchymal stem cells (cMSCs) in delivering SkM1. METHODS AND RESULTS: cMSCs were isolated and transfected with SkM1. Coculture experiments showed cMSC/SkM1 but not cMSC alone and maintained fast conduction at depolarized potentials. We studied 3 groups in the canine 7d infarct: sham, cMSC, and cMSC/SkM1. In vivo epicardial border zones electrograms were broad and fragmented in sham, narrower in cMSCs, and narrow and unfragmented in cMSC/SkM1 (P<0.05). During programmed electrical stimulation of epicardial border zones, QRS duration in cMSC/SkM1 was shorter than in cMSC and sham (P<0.05). Programmed electrical stimulation-induced ventricular tachycardia/ventricular fibrillation was equivalent in all groups (P>0.05). CONCLUSION: cMSCs provide efficient delivery of SkM1 current. The interventions performed (cMSCs or cMSC/SkM1) were neither antiarrhythmic nor proarrhythmic. Comparing outcomes with cMSC/SkM1 and viral gene delivery highlights the criticality of the delivery platform to SkM1 antiarrhythmic efficacy.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Proteínas Musculares/metabolismo , Infarto del Miocardio/cirugía , Miocitos Cardíacos/metabolismo , Canales de Sodio/metabolismo , Sodio/metabolismo , Taquicardia Ventricular/prevención & control , Fibrilación Ventricular/prevención & control , Potenciales de Acción , Animales , Animales Recién Nacidos , Estimulación Cardíaca Artificial , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Perros , Técnicas Electrofisiológicas Cardíacas , Humanos , Proteínas Musculares/genética , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.5 , Ratas , Ratas Sprague-Dawley , Canales de Sodio/genética , Taquicardia Ventricular/etiología , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Transfección , Fibrilación Ventricular/etiología , Fibrilación Ventricular/genética , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA