Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 596(2): 181-196, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29193078

RESUMO

KEY POINTS: Optogenetics has emerged as a potential alternative to electrotherapy for treating heart rhythm disorders, but its applicability for terminating atrial arrhythmias remains largely unexplored. We used computational models reconstructed from clinical MRI scans of fibrotic patient atria to explore the feasibility of optogenetic termination of atrial tachycardia (AT), comparing two different illumination strategies: distributed vs. targeted. We show that targeted optogenetic stimulation based on automated, non-invasive flow-network analysis of patient-specific re-entry morphology may be a reliable approach for identifying the optimal illumination target in each individual (i.e. the critical AT isthmus). The above-described approach yields very high success rates (up to 100%) and requires dramatically less input power than distributed illumination We conclude that simulations in patient-specific models show that targeted light pulses lasting longer than the AT cycle length can efficiently and reliably terminate AT if the human atria can be successfully light-sensitized via gene delivery of ChR2. ABSTRACT: Optogenetics has emerged as a potential alternative to electrotherapy for treating arrhythmia, but feasibility studies have been limited to ventricular defibrillation via epicardial light application. Here, we assess the efficacy of optogenetic atrial tachycardia (AT) termination in human hearts using a strategy that targets for illumination specific regions identified in an automated manner. In three patient-specific models reconstructed from late gadolinium-enhanced MRI scans, we simulated channelrhodopsin-2 (ChR2) expression via gene delivery. In all three models, we attempted to terminate re-entrant AT (induced via rapid pacing) via optogenetic stimulation. We compared two strategies: (1) distributed illumination of the endocardium by multi-optrode grids (number of optrodes, Nopt  = 64, 128, 256) and (2) targeted illumination of the critical isthmus, which was identified via analysis of simulated activation patterns using an algorithm based on flow networks. The illuminated area and input power were smaller for the targeted approach (19-57.8 mm2 ; 0.6-1.8 W) compared to the sparsest distributed arrays (Nopt  = 64; 124.9 ± 6.3 mm2 ; 3.9 ± 0.2 W). AT termination rates for distributed illumination were low, ranging from <5% for short pulses (1/10 ms long) to ∼20% for longer stimuli (100/1000 ms). When we attempted to terminate the same AT episodes with targeted illumination, outcomes were similar for short pulses (1/10 ms long: 0% success) but improved for longer stimuli (100 ms: 54% success; 1000 ms: 90% success). We conclude that simulations in patient-specific models show that light pulses lasting longer than the AT cycle length can efficiently and reliably terminate AT in atria light-sensitized via gene delivery. We show that targeted optogenetic stimulation based on analysis of AT morphology may be a reliable approach for defibrillation and requires less power than distributed illumination.


Assuntos
Potenciais de Ação , Simulação por Computador , Átrios do Coração/citologia , Optogenética/métodos , Taquicardia/terapia , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Átrios do Coração/fisiopatologia , Átrios do Coração/efeitos da radiação , Humanos
2.
J Physiol ; 594(23): 6879-6891, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-26941055

RESUMO

KEY POINTS: Optogenetics-based defibrillation, a theoretical alternative to electrotherapy, involves expression of light-sensitive ion channels in the heart (via gene or cell therapy) and illumination of the cardiac surfaces (via implanted LED arrays) to elicit light-induced activations. We used a biophysically detailed human ventricular model to determine whether such a therapy could terminate fibrillation (VF) and identify which combinations of light-sensitive ion channel properties and illumination configurations would be effective. Defibrillation was successful when a large proportion (> 16.6%) of ventricular tissue was directly stimulated by light that was bright enough to induce an action potential in an uncoupled cell. While illumination with blue light never successfully terminated VF, illumination of red light-sensitive ion channels with dense arrays of implanted red light sources resulted in successful defibrillation. Our results suggest that cardiac expression of red light-sensitive ion channels is necessary for the development of effective optogenetics-based defibrillation therapy using LED arrays. ABSTRACT: Optogenetics-based defibrillation has been proposed as a novel and potentially pain-free approach to enable cardiomyocyte-selective defibrillation in humans, but the feasibility of such a therapy remains unknown. This study aimed to (1) assess the feasibility of terminating sustained ventricular fibrillation (VF) via light-induced excitation of opsins expressed throughout the myocardium and (2) identify the ideal (theoretically possible) opsin properties and light source configurations that would maximise therapeutic efficacy. We conducted electrophysiological simulations in an MRI-based human ventricular model with VF induced by rapid pacing; light sensitisation via systemic, cardiac-specific gene transfer of channelrhodopsin-2 (ChR2) was simulated. In addition to the widely used blue light-sensitive ChR2-H134R, we also modelled theoretical ChR2 variants with augmented light sensitivity (ChR2+), red-shifted spectral sensitivity (ChR2-RED) or both (ChR2-RED+). Light sources were modelled as synchronously activating LED arrays (LED radius: 1 mm; optical power: 10 mW mm-2 ; array density: 1.15-4.61 cm-2 ). For each unique optogenetic configuration, defibrillation was attempted with two different optical pulse durations (25 and 500 ms). VF termination was only successful for configurations involving ChR2-RED and ChR2-RED+ (for LED arrays with density ≥ 2.30 cm-2 ), suggesting that opsin spectral sensitivity was the most important determinant of optogenetic defibrillation efficacy. This was due to the deeper penetration of red light in cardiac tissue compared with blue light, which resulted in more widespread light-induced propagating wavefronts. Longer pulse duration and higher LED array density were associated with increased optogenetic defibrillation efficacy. In all cases observed, the defibrillation mechanism was light-induced depolarisation of the excitable gap, which led to block of reentrant wavefronts.


Assuntos
Coração/efeitos da radiação , Fibrilação Ventricular/terapia , Channelrhodopsins , Simulação por Computador , Humanos , Luz , Optogenética , Modelagem Computacional Específica para o Paciente
3.
Europace ; 16 Suppl 4: iv69-iv76, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25362173

RESUMO

AIMS: Diseases that abbreviate the cardiac action potential (AP) by increasing the strength of repolarizing transmembrane currents are highly arrhythmogenic. It has been proposed that optogenetic tools could be used to restore normal AP duration (APD) in the heart under such disease conditions. This study aims to evaluate the efficacy of an optogenetic treatment modality for prolonging pathologically shortened APs in a detailed computational model of short QT syndrome (SQTS) in the human atria, and compare it to drug treatment. METHODS AND RESULTS: We used a human atrial myocyte model with faster repolarization caused by SQTS; light sensitivity was inscribed via the presence of channelrhodopsin-2 (ChR2). We conducted simulations in single cells and in a magnetic resonance imaging-based model of the human left atrium (LA). Application of an appropriate optical stimulus to a diseased cell dynamically increased APD, producing an excellent match to control AP (<1.5 mV deviation); treatment of a diseased cell with an AP-prolonging drug (chloroquine) also increased APD, but the match to control AP was worse (>5 mV deviation). Under idealized conditions in the LA (uniform ChR2-expressing cell distribution, no light attenuation), optogenetics-based therapy outperformed chloroquine treatment (APD increased to 87% and 81% of control). However, when non-uniform ChR2-expressing cell distribution and light attenuation were incorporated, optogenetics-based treatment was less effective (APD only increased to 55%). CONCLUSION: This study demonstrates proof of concept for optogenetics-based treatment of diseases that alter atrial AP shape. We identified key practical obstacles intrinsic to the optogenetic approach that must be overcome before such treatments can be realized.


Assuntos
Antiarrítmicos/uso terapêutico , Cloroquina/uso terapêutico , Átrios do Coração/efeitos dos fármacos , Optogenética , Potenciais de Ação , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/terapia , Simulação por Computador , Técnicas Eletrofisiológicas Cardíacas , Estudos de Viabilidade , Átrios do Coração/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Modelos Cardiovasculares , Rodopsina/genética , Rodopsina/metabolismo , Fatores de Tempo
4.
Front Physiol ; 12: 718622, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34526912

RESUMO

Optogenetic defibrillation of hearts expressing light-sensitive cation channels (e.g., ChR2) has been proposed as an alternative to conventional electrotherapy. Past modeling work has shown that ChR2 stimulation can depolarize enough myocardium to interrupt arrhythmia, but its efficacy is limited by light attenuation and high energy needs. These shortcomings may be mitigated by using new optogenetic proteins like Guillardia theta Anion Channelrhodopsin (GtACR1), which produces a repolarizing outward current upon illumination. Accordingly, we designed a study to assess the feasibility of GtACR1-based optogenetic arrhythmia termination in human hearts. We conducted electrophysiological simulations in MRI-based atrial or ventricular models (n = 3 each), with pathological remodeling from atrial fibrillation or ischemic cardiomyopathy, respectively. We simulated light sensitization via viral gene delivery of three different opsins (ChR2, red-shifted ChR2, GtACR1) and uniform endocardial illumination at the appropriate wavelengths (blue, red, or green light, respectively). To analyze consistency of arrhythmia termination, we varied pulse timing (three evenly spaced intervals spanning the reentrant cycle) and intensity (atrial: 0.001-1 mW/mm2; ventricular: 0.001-10 mW/mm2). In atrial models, GtACR1 stimulation with 0.005 mW/mm2 green light consistently terminated reentry; this was 10-100x weaker than the threshold levels for ChR2-mediated defibrillation. In ventricular models, defibrillation was observed in 2/3 models for GtACR1 stimulation at 0.005 mW/mm2 (100-200x weaker than ChR2 cases). In the third ventricular model, defibrillation failed in nearly all cases, suggesting that attenuation issues and patient-specific organ/scar geometry may thwart termination in some cases. Across all models, the mechanism of GtACR1-mediated defibrillation was voltage forcing of illuminated tissue toward the modeled channel reversal potential of -40 mV, which made propagation through affected regions impossible. Thus, our findings suggest GtACR1-based optogenetic defibrillation of the human heart may be feasible with ≈2-3 orders of magnitude less energy than ChR2.

5.
JACC Clin Electrophysiol ; 4(2): 155-167, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29749932

RESUMO

Cardiac optogenetics is an emergent research area involving the delivery of light-sensitive proteins (opsins) to excitable heart tissue to enable optical modulation of cardiac electrical function. Optogenetic stimulation has many noteworthy advantages over conventional electrical methods, including selective electrophysiological modulation in specifically targeted cell subpopulations, high-resolution spatiotemporal control via patterned illumination, and use of different opsins to elicit inward or outward transmembrane current. This review summarizes developments achieved since the inception of cardiac optogenetics research, which has spanned nearly a decade. The authors first provide an overview of recent methodological advances in opsin engineering, light sensitization of cardiac tissue, strategies for illuminating the heart, and frameworks for simulating optogenetics in realistic computational models of patient hearts. They then review recent cardiac optogenetics applications, including: 1) all-optical, high-throughput, contactless assays for quantification of electrophysiological properties; 2) optogenetic perturbation of cardiac tissue to unveil mechanistic insights on the initiation, perpetuation, and termination of arrhythmia; and 3) disruptive translational innovations such as light-based pacemaking and defibrillation.


Assuntos
Eletrofisiologia Cardíaca , Optogenética , Humanos
6.
Nat Commun ; 8(1): 106, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28740174

RESUMO

Plakophilin-2 (PKP2) is a component of the desmosome and known for its role in cell-cell adhesion. Mutations in human PKP2 associate with a life-threatening arrhythmogenic cardiomyopathy, often of right ventricular predominance. Here, we use a range of state-of-the-art methods and a cardiomyocyte-specific, tamoxifen-activated, PKP2 knockout mouse to demonstrate that in addition to its role in cell adhesion, PKP2 is necessary to maintain transcription of genes that control intracellular calcium cycling. Lack of PKP2 reduces expression of Ryr2 (coding for Ryanodine Receptor 2), Ank2 (coding for Ankyrin-B), Cacna1c (coding for CaV1.2) and Trdn (coding for triadin), and protein levels of calsequestrin-2 (Casq2). These factors combined lead to disruption of intracellular calcium homeostasis and isoproterenol-induced arrhythmias that are prevented by flecainide treatment. We propose a previously unrecognized arrhythmogenic mechanism related to PKP2 expression and suggest that mutations in PKP2 in humans may cause life-threatening arrhythmias even in the absence of structural disease.It is believed that mutations in desmosomal adhesion complex protein plakophilin 2 (PKP2) cause arrhythmia due to loss of cell-cell communication. Here the authors show that PKP2 controls the expression of proteins involved in calcium cycling in adult mouse hearts, and that lack of PKP2 can cause arrhythmia in a structurally normal heart.


Assuntos
Cálcio/metabolismo , Coração/fisiologia , Miocárdio/metabolismo , Placofilinas/genética , Transcrição Gênica , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Western Blotting , Expressão Gênica , Coração/fisiopatologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Placofilinas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Clin Med Insights Cardiol ; 10(Suppl 1): 47-60, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27840581

RESUMO

Light has long been used to image the heart, but now it can be used to modulate its electrophysiological function. Imaging modalities and techniques have long constituted an indispensable part of arrhythmia research and treatment. Recently, advances in the fields of optogenetics and photodynamic therapy have provided scientists with more effective approaches for probing, studying and potentially devising new treatments for cardiac arrhythmias. This article is a review of research toward the application of these techniques. It contains (a) an overview of advancements in technology and research that have contributed to light-based cardiac applications and (b) a summary of current and potential future applications of light-based control of cardiac cells, including modulation of heart rhythm, manipulation of cardiac action potential morphology, quantitative analysis of arrhythmias, defibrillation and cardiac ablation.

8.
J Clin Invest ; 126(10): 3894-3904, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27617859

RESUMO

Ventricular arrhythmias are among the most severe complications of heart disease and can result in sudden cardiac death. Patients at risk currently receive implantable defibrillators that deliver electrical shocks to terminate arrhythmias on demand. However, strong electrical shocks can damage the heart and cause severe pain. Therefore, we have tested optogenetic defibrillation using expression of the light-sensitive channel channelrhodopsin-2 (ChR2) in cardiac tissue. Epicardial illumination effectively terminated ventricular arrhythmias in hearts from transgenic mice and from WT mice after adeno-associated virus-based gene transfer of ChR2. We also explored optogenetic defibrillation for human hearts, taking advantage of a recently developed, clinically validated in silico approach for simulating infarct-related ventricular tachycardia (VT). Our analysis revealed that illumination with red light effectively terminates VT in diseased, ChR2-expressing human hearts. Mechanistically, we determined that the observed VT termination is due to ChR2-mediated transmural depolarization of the myocardium, which causes a block of voltage-dependent Na+ channels throughout the myocardial wall and interrupts wavefront propagation into illuminated tissue. Thus, our results demonstrate that optogenetic defibrillation is highly effective in the mouse heart and could potentially be translated into humans to achieve nondamaging and pain-free termination of ventricular arrhythmia.


Assuntos
Miocárdio/metabolismo , Fibrilação Ventricular/terapia , Animais , Channelrhodopsins , Simulação por Computador , Feminino , Terapia Genética , Humanos , Masculino , Camundongos Transgênicos , Modelos Biológicos , Infarto do Miocárdio , Miocárdio/patologia , Optogenética , Ativação Transcricional/efeitos da radiação
9.
Trends Cardiovasc Med ; 25(2): 73-81, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25453984

RESUMO

Optogenetics is an exciting new technology in which viral gene or cell delivery is used to inscribe light sensitivity in excitable tissue to enable optical control of bioelectric behavior. Initial progress in the fledgling domain of cardiac optogenetics has included in vitro expression of various light-sensitive proteins in cell monolayers and transgenic animals to demonstrate an array of potentially useful applications, including light-based pacing, silencing of spontaneous activity, and spiral wave termination. In parallel to these developments, the cardiac modeling community has developed a versatile computational framework capable of realistically simulating optogenetics in biophysically detailed, patient-specific representations of the human heart, enabling the exploration of potential clinical applications in a predictive virtual platform. Toward the ultimate goal of assessing the feasibility and potential impact of optogenetics-based therapies in cardiovascular medicine, this review provides (1) a detailed synopsis of in vivo, in vitro, and in silico developments in the field and (2) a critical assessment of how existing clinical technology for gene/cell delivery and intra-cardiac illumination could be harnessed to achieve such lofty goals as light-based arrhythmia termination.


Assuntos
Doenças Cardiovasculares/terapia , Optogenética/métodos , Simulação por Computador , Humanos
10.
Comput Biol Med ; 65: 200-8, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26002074

RESUMO

Cardiac optogenetics is emerging as an exciting new potential avenue to enable spatiotemporally precise control of excitable cells and tissue in the heart with low-energy optical stimuli. This approach involves the expression of exogenous light-sensitive proteins (opsins) in target heart tissue via viral gene or cell delivery. Preliminary experiments in optogenetically-modified cells, tissue, and organisms have made great strides towards demonstrating the feasibility of basic applications, including the use of light stimuli to pace or disrupt reentrant activity. However, it remains unknown whether techniques based on this intriguing technology could be scaled up and used in humans for novel clinical applications, such as pain-free optical defibrillation or dynamic modulation of action potential shape. A key step towards answering such questions is to explore potential optogenetics-based therapies using sophisticated computer simulation tools capable of realistically representing opsin delivery and light stimulation in biophysically detailed, patient-specific models of the human heart. This review provides (1) a detailed overview of the methodological developments necessary to represent optogenetics-based solutions in existing virtual heart platforms and (2) a survey of findings that have been derived from such simulations and a critical assessment of their significance with respect to the progress of the field.


Assuntos
Simulação por Computador , Cardiopatias/fisiopatologia , Coração/fisiopatologia , Modelos Cardiovasculares , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA