Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Biophys J ; 116(3): 530-539, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30686487

RESUMO

Ryanodine receptors (RyRs) mediate calcium (Ca)-induced Ca release and intracellular Ca homeostasis. In a cardiac myocyte, RyRs group into clusters of variable size from a few to several hundred RyRs, creating a spatially nonuniform intracellular distribution. It is unclear how heterogeneity of RyR cluster size alters spontaneous sarcoplasmic reticulum (SR) Ca releases (Ca sparks) and arrhythmogenic Ca waves. Here, we tested the impact of heterogeneous RyR cluster size on the initiation of Ca waves. Experimentally, we measured RyR cluster sizes at Ca spark sites in rat ventricular myocytes and further tested functional impacts using a physiologically detailed computational model with spatial and stochastic intracellular Ca dynamics. We found that the spark frequency and amplitude increase nonlinearly with the size of RyR clusters. Larger RyR clusters have lower SR Ca release threshold for local Ca spark initiation and exhibit steeper SR Ca release versus SR Ca load relationship. However, larger RyR clusters tend to lower SR Ca load because of the higher Ca leak rate. Conversely, smaller clusters have a higher threshold and a lower leak, which tends to increase SR Ca load. At the myocyte level, homogeneously large or small RyR clusters limit Ca waves (because of low load for large clusters but low excitability for small clusters). Mixtures of large and small RyR clusters potentiates Ca waves because the enhanced SR Ca load driven by smaller clusters enables Ca wave initiation and propagation from larger RyR clusters. Our study suggests that a spatially heterogeneous distribution of RyR cluster size under pathological conditions may potentiate Ca waves and thus afterdepolarizations and triggered arrhythmias.


Assuntos
Sinalização do Cálcio , Modelos Biológicos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Diástole , Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Probabilidade
2.
Biophys J ; 106(6): 1391-7, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24655514

RESUMO

Cardiac action potential alternans and early afterdepolarizations (EADs) are linked to cardiac arrhythmias. Periodic action potentials (period 1) in healthy conditions bifurcate to other states such as period 2 or chaos when alternans or EADs occur in pathological conditions. The mechanisms of alternans and EADs have been extensively studied under steady-state conditions, but lethal arrhythmias often occur during the transition between steady states. Why arrhythmias tend to develop during the transition is unclear. We used low-dimensional mathematical models to analyze dynamical mechanisms of transient alternans and EADs. We show that depending on the route from one state to another, action potential alternans and EADs may occur during the transition between two periodic steady states. The route taken depends on the time course of external perturbations or intrinsic signaling, such as ß-adrenergic stimulation, which regulate cardiac calcium and potassium currents with differential kinetics.


Assuntos
Potenciais de Ação , Arritmias Cardíacas/metabolismo , Modelos Cardiovasculares , Miócitos Cardíacos/fisiologia , Animais , Humanos , Canais Iônicos/metabolismo , Miócitos Cardíacos/metabolismo
3.
Europace ; 16(3): 452-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24569900

RESUMO

AIMS: Ventricular tachycardia (VT) and fibrillation (VF) are the most lethal cardiac arrhythmias. The degeneration of VT into VF is associated with the breakup of a spiral wave of the action potential in cardiac tissue. ß-Adrenergic (ßAR) signalling potentiates the L-type Ca current (ICaL) faster than the slow delayed rectifier potassium current (IKs), which transiently prolongs the action potential duration (APD) and promotes early after depolarizations. In this study, we aimed at investigating how ßAR signalling affects the transition from VT to VF. METHODS AND RESULTS: We used a physiologically detailed computer model of the rabbit ventricular myocyte in a two-dimensional tissue to determine how spiral waves respond to ßAR activation following administration of isoproterenol. A simplified mathematical model was also used to investigate the underlying dynamics. We found that the spatiotemporal behaviour of spiral waves strongly depends on the kinetics of ßAR activation. When ßAR activation is rapid, a stable spiral wave turns into small fragments and its electrocardiogram reveals the transition from VT to VF. This is due to the transiently steepened APD restitution induced by the faster activation of ICaL vs. IKs upon sudden ßAR activation. The spiral wave may also disappear if its transient wavelength is too large to be supported by the tissue size upon sudden strong ßAR activation that prolongs APD transiently. When ßAR activation is gradual, a stable spiral wave remains such, because of more limited increase in both APD and slope of APD restitution due to more contemporaneous ICaL and IKs activation. CONCLUSION: Changes in APD restitution during ßAR activation revealed a novel transient spiral wave dynamics; this spatiotemporal characteristic strongly depends on the protocol of isoproterenol application.


Assuntos
Potenciais de Ação , Sistema de Condução Cardíaco/fisiopatologia , Ventrículos do Coração/fisiopatologia , Modelos Cardiovasculares , Receptores Adrenérgicos beta/metabolismo , Taquicardia Ventricular/fisiopatologia , Fibrilação Ventricular/fisiopatologia , Animais , Simulação por Computador , Progressão da Doença , Coelhos , Transdução de Sinais , Taquicardia Ventricular/complicações , Fibrilação Ventricular/etiologia
4.
J Mol Cell Cardiol ; 58: 153-61, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23481579

RESUMO

Sympathetic stimulation regulates cardiac excitation-contraction coupling in hearts but can also trigger ventricular arrhythmias caused by early afterdepolarizations (EADs) in pathological conditions. Isoproterenol (ISO) stimulation can transiently cause EADs which could result from differential kinetics of L-type Ca current (ICaL) vs. delayed rectifier potassium current (IKs) effects, but multiple PKA targets complicate mechanistic analysis. Utilizing a biophysically detailed model integrating Ca and ß-adrenergic signaling, we investigate how different phosphorylation kinetics and targets influence ß-adrenergic-induced transient EADs. We found that: 1) The faster time course of ICaL vs. IKs increases recapitulates experimentally observed ISO-induced transient EADs (which are due to ICaL reactivation). These EADs disappear at steady state ISO and do not occur during more gradual ISO application. 2) This ICaL vs. IKs kinetic mismatch with ISO can also induce transient EADs due to spontaneous sarcoplasmic reticulum (SR) Ca release and Na/Ca exchange current. The increased ICaL, SR Ca uptake and action potential duration (APD) raise SR Ca to cause spontaneous SR Ca release, but eventual IKs activation and APD shortening abolish these EADs. 3) Phospholemman (PLM) phosphorylation decreases both types of EADs by increasing outward Na/K-ATPase current (INaK) for ICaL-mediated EADs, and reducing intracellular Na and Ca loading for SR Ca-release-mediated EADs. Slowing PLM phosphorylation kinetics abolishes this protective effect. 4) Blocking phospholamban (PLB) phosphorylation has little effect on ICaL-mediated transient EADs, but abolishes SR Ca-release-mediated transient EADs by limiting SR Ca loading. 5) RyR phosphorylation has little effect on either transient EAD type. Our study emphasizes the importance of understanding non-steady state kinetics of several systems in mediating ß-adrenergic-induced EADs and arrhythmias.


Assuntos
Arritmias Cardíacas/metabolismo , Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animais , Arritmias Cardíacas/patologia , Sinalização do Cálcio/efeitos dos fármacos , Acoplamento Excitação-Contração/fisiologia , Humanos , Isoproterenol/administração & dosagem , Cinética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação/efeitos dos fármacos , Coelhos
5.
J Mol Cell Cardiol ; 58: 118-24, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23220129

RESUMO

Ca(2+) waves were probably first observed in the early 1940s. Since then Ca(2+) waves have captured the attention of an eclectic mixture of mathematicians, neuroscientists, muscle physiologists, developmental biologists, and clinical cardiologists. This review discusses the current state of mathematical models of Ca(2+) waves, the normal physiological functions Ca(2+) waves might serve in cardiac cells, as well as how the spatial arrangement of Ca(2+) release channels shape Ca(2+) waves, and we introduce the idea of Ca(2+) phase waves that might provide a useful framework for understanding triggered arrhythmias.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Modelos Teóricos , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Humanos , Miocárdio/metabolismo , Miocárdio/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina
6.
Biotechnol Genet Eng Rev ; : 1-16, 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36951554

RESUMO

Myasthenia gravis is a major disease in the context of an ageing society, and the discovery of effective herbal compound and herbal active ingredients is a highly promising direction for the treatment of myasthenia gravis. In this study, we selected shujiao, dried ginger and ginseng from the compound ingredients through a network pathology approach. The three ingredients were used to obtain drug targets in Traditional Chinese Medicine Systems Pharmacology (TCMSP), HERB and BATMAN-TCM data and intersected with the disease targets of myasthenia gravis. The resulting regulatory network maps were then used to identify core genes through the String database, and finally the core genes were molecularly aligned with the corresponding active ingredients using Autodock vina software. The 'herbal-component-target' regulatory network of the Chinese herbal formulae was constructed, which is important for finding the potential molecular mechanism for the treatment of myasthenia gravis. It will provide a theoretical basis for the therapeutic and clinical research of myasthenia gravis.

7.
Biophys J ; 103(2): 365-73, 2012 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-22853915

RESUMO

Early afterdepolarizations (EADs) are linked to both triggered arrhythmias and reentrant arrhythmias by causing premature ventricular complexes (PVCs), focal excitations, or heterogeneous tissue substrates for reentry formation. However, a critical number of cells that synchronously exhibit EADs are needed to result in arrhythmia triggers and substrates in tissue. In this study, we use mathematical modeling and computer simulations to investigate EAD synchronization and arrhythmia induction in tissue models with random cell-to-cell variations. Our major observations are as follows. Random cell-to-cell variations in action potential duration without EAD presence do not cause large dispersion of refractoriness in well-coupled tissue. In the presence of phase-2 EADs, the cells may synchronously exhibit the same number of EADs or no EADs with a very small dispersion of refractoriness, or synchronize regionally to result in large dispersion of refractoriness. In the presence of phase-3 EADs, regional synchronization leads to propagating EADs, forming PVCs in tissue. Interestingly, even though the uncoupled cells exhibit either no EAD or only a single EAD, when these cells are coupled to form a tissue, more than one PVC can occur. When the PVCs occur at different locations and time, multifocal arrhythmias are triggered, with the foci shifting in space and time in an irregular manner. The focal arrhythmias either spontaneously terminate or degenerate into reentrant arrhythmias due to heterogeneities and spatiotemporal chaotic dynamics of the foci.


Assuntos
Potenciais de Ação/fisiologia , Arritmias Cardíacas/fisiopatologia , Coração/fisiopatologia , Modelos Cardiovasculares , Animais , Miocárdio/patologia , Coelhos , Complexos Ventriculares Prematuros/fisiopatologia
8.
J Physiol ; 589(Pt 24): 6081-92, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22025660

RESUMO

Sudden cardiac death (SCD) due to ventricular fibrillation (VF) is a major world-wide health problem. A common trigger of VF involves abnormal repolarization of the cardiac action potential causing early afterdepolarizations (EADs). Here we used a hybrid biological-computational approach to investigate the dependence of EADs on the biophysical properties of the L-type Ca(2+) current (I(Ca,L)) and to explore how modifications of these properties could be designed to suppress EADs. EADs were induced in isolated rabbit ventricular myocytes by exposure to 600 µmol l(-1) H(2)O(2) (oxidative stress) or lowering the external [K(+)] from 5.4 to 2.0-2.7 mmol l(-1) (hypokalaemia). The role of I(Ca,L) in EAD formation was directly assessed using the dynamic clamp technique: the paced myocyte's V(m) was input to a myocyte model with tunable biophysical parameters, which computed a virtual I(Ca,L), which was injected into the myocyte in real time. This virtual current replaced the endogenous I(Ca,L), which was suppressed with nifedipine. Injecting a current with the biophysical properties of the native I(Ca,L) restored EAD occurrence in myocytes challenged by H(2)O(2) or hypokalaemia. A mere 5 mV depolarizing shift in the voltage dependence of activation or a hyperpolarizing shift in the steady-state inactivation curve completely abolished EADs in myocytes while maintaining a normal Ca(i) transient. We propose that modifying the biophysical properties of I(Ca,L) has potential as a powerful therapeutic strategy for suppressing EADs and EAD-mediated arrhythmias.


Assuntos
Potenciais de Ação/fisiologia , Arritmias Cardíacas/fisiopatologia , Cálcio/fisiologia , Miócitos Cardíacos/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Peróxido de Hidrogênio/farmacologia , Hipopotassemia/fisiopatologia , Nifedipino/farmacologia , Oxidantes/farmacologia , Estresse Oxidativo , Técnicas de Patch-Clamp , Coelhos
9.
Am J Physiol Heart Circ Physiol ; 300(1): H271-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21037233

RESUMO

Anisotropy can lead to unidirectional conduction block that initiates reentry. We analyzed the mechanisms in patterned anisotropic neonatal rat ventricular myocyte monolayers. Voltage and intracellular Ca (Ca(i)) were optically mapped under the following conditions: extrastimulus (S1S2) testing and/or tetrodotoxin (TTX) to suppress Na current availability; heptanol to reduce gap junction conductance; and incremental rapid pacing. In anisotropic monolayers paced at 2 Hz, conduction velocity (CV) was faster longitudinally than transversely, with an anisotropy ratio [AR = CV(L)/CV(T), where CV(L) and CV(T) are CV in the longitudinal and transverse directions, respectively], averaging 2.1 ± 0.8. Interventions decreasing Na current availability, such as S1S2 pacing and TTX, slowed CV(L) and CV(T) proportionately, without changing the AR. Conduction block preferentially occurred longitudinal to fiber direction, commonly initiating reentry. Interventions that decreased gap junction conductance, such as heptanol, decreased CV(T) more than CV(L), increasing the AR and causing preferential transverse conduction block and reentry. Rapid pacing resembled the latter, increasing the AR and promoting transverse conduction block and reentry, which was prevented by the Ca(i) chelator 1,2-bis oaminophenoxy ethane-N,N,N',N'-tetraacetic acid (BAPTA). In contrast to isotropic and uniformly anisotropic monolayers, in which reentrant rotors drifted and self-terminated, bidirectional anisotropy (i.e., an abrupt change in fiber direction exceeding 45°) caused reentry to anchor near the zone of fiber direction change in 77% of monolayers. In anisotropic monolayers, unidirectional conduction block initiating reentry can occur longitudinal or transverse to fiber direction, depending on whether the experimental intervention reduces Na current availability or decreases gap junction conductance, agreeing with theoretical predictions.


Assuntos
Bloqueio Atrioventricular/fisiopatologia , Miócitos Cardíacos/fisiologia , Função Ventricular/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Anisotropia , Bloqueio Atrioventricular/metabolismo , Cálcio/metabolismo , Células Cultivadas , Distribuição de Qui-Quadrado , Junções Comunicantes/fisiologia , Imuno-Histoquímica , Miócitos Cardíacos/citologia , Ratos , Ratos Sprague-Dawley
10.
Biophys J ; 99(5): 1408-15, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20816052

RESUMO

How early (EADs) and delayed afterdepolarizations (DADs) overcome electrotonic source-sink mismatches in tissue to trigger premature ventricular complexes remains incompletely understood. To study this question, we used a rabbit ventricular action potential model to simulate tissues in which a central area of contiguous myocytes susceptible to EADs or DADs was surrounded by unsusceptible tissue. In 1D tissue with normal longitudinal conduction velocity (0.55 m/s), the numbers of contiguous susceptible myocytes required for an EAD and a barely suprathreshold DAD to trigger a propagating action potential were 70 and 80, respectively. In 2D tissue, these numbers increased to 6940 and 7854, and in 3D tissue to 696,910 and 817,280. These numbers were significantly decreased by reduced gap junction conductance, simulated fibrosis, reduced repolarization reserve and heart failure electrical remodeling. In conclusion, the source-sink mismatch in well-coupled cardiac tissue powerfully protects the heart from arrhythmias due to sporadic afterdepolarizations. Structural and electrophysiological remodeling decrease these numbers significantly but still require synchronization mechanisms for EADs and DADs to overcome the robust protective effects of source-sink mismatch.


Assuntos
Potenciais de Ação , Modelos Biológicos , Células Musculares/citologia , Células Musculares/patologia , Animais , Anisotropia , Condutividade Elétrica , Fibrose , Junções Comunicantes/metabolismo , Insuficiência Cardíaca/patologia , Coelhos , Complexos Ventriculares Prematuros/patologia
11.
Am J Physiol Heart Circ Physiol ; 297(2): H775-84, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19482965

RESUMO

Recent experimental studies have shown that fibroblasts can electrotonically couple to myocytes via gap junctions. In this study, we investigated how this coupling affects action potential and intracellular calcium (Ca(i)) cycling dynamics in simulated fibroblast-myocyte pairs and in two-dimensional tissue with random fibroblast insertions. We show that a fibroblast coupled with a myocyte generates a gap junction current flowing to the myocyte with two main components: an early pulse of transient outward current, similar to the fast transient outward current, and a later background current during the repolarizing phase. Depending on the relative prominence of the two components, fibroblast-myoycte coupling can 1) prolong or shorten action potential duration (APD), 2) promote or suppress APD alternans due to steep APD restitution (voltage driven) and also result in a novel mechanism of APD alternans at slow heart rates, 3) promote Ca(i)-driven alternans and electromechanically discordant alternans, and 4) promote spatially discordant alternans by two mechanisms: by altering conduction velocity restitution and by heterogeneous fibroblast distribution causing electromechanically concordant and discordant alternans in different regions of the tissue. Thus, through their coupling with myocytes, fibroblasts alter repolarization and Ca(i) cycling alternans at both the cellular and tissue scales, which may play important roles in arrhythmogenesis in diseased cardiac tissue with fibrosis.


Assuntos
Fibroblastos/citologia , Fibroblastos/fisiologia , Modelos Cardiovasculares , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Potenciais de Ação/fisiologia , Animais , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Cálcio/fisiologia , Comunicação Celular/fisiologia , Células Cultivadas , Simulação por Computador , Fibrose , Sistema de Condução Cardíaco/fisiologia , Potenciais da Membrana/fisiologia , Coelhos
12.
Am J Physiol Heart Circ Physiol ; 297(5): H1594-605, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19767530

RESUMO

Oxidative stress with hydrogen peroxide (H(2)O(2)) readily promotes early afterdepolarizations (EADs) and triggered activity (TA) in isolated rat and rabbit ventricular myocytes. Here we examined the effects of H(2)O(2) on arrhythmias in intact Langendorff rat and rabbit hearts using dual-membrane voltage and intracellular calcium optical mapping and glass microelectrode recordings. Young adult rat (3-5 mo, N = 25) and rabbit (3-5 mo, N = 6) hearts exhibited no arrhythmias when perfused with H(2)O(2) (0.1-2 mM) for up to 3 h. However, in 33 out of 35 (94%) aged (24-26 mo) rat hearts, 0.1 mM H(2)O(2) caused EAD-mediated TA, leading to ventricular tachycardia (VT) and fibrillation (VF). Aged rabbits (life span, 8-12 yr) were not available, but 4 of 10 middle-aged rabbits (3-5 yr) developed EADs, TA, VT, and VF. These arrhythmias were suppressed by the reducing agent N-acetylcysteine (2 mM) and CaMKII inhibitor KN-93 (1 microM) but not by its inactive form (KN-92, 1 microM). There were no significant differences between action potential duration (APD) or APD restitution slope before or after H(2)O(2) in aged or young adult rat hearts. In histological sections, however, trichrome staining revealed that aged rat hearts exhibited extensive fibrosis, ranging from 10-90%; middle-aged rabbit hearts had less fibrosis (5-35%), whereas young adult rat and rabbit hearts had <4% fibrosis. In aged rat hearts, EADs and TA arose most frequently (70%) from the left ventricular base where fibrosis was intermediate ( approximately 30%). Computer simulations in two-dimensional tissue incorporating variable degrees of fibrosis showed that intermediate (but not mild or severe) fibrosis promoted EADs and TA. We conclude that in aged ventricles exposed to oxidative stress, fibrosis facilitates the ability of cellular EADs to emerge and generate TA, VT, and VF at the tissue level.


Assuntos
Envelhecimento , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Taquicardia Ventricular/metabolismo , Fibrilação Ventricular/metabolismo , Acetilcisteína/farmacologia , Potenciais de Ação , Fatores Etários , Animais , Antioxidantes/farmacologia , Benzilaminas/farmacologia , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Comunicação Celular , Simulação por Computador , Modelos Animais de Doenças , Eletrocardiografia , Ativação Enzimática , Fibrose , Ventrículos do Coração/metabolismo , Peróxido de Hidrogênio , Masculino , Modelos Cardiovasculares , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Oxidantes , Estresse Oxidativo/efeitos dos fármacos , Perfusão , Inibidores de Proteínas Quinases/farmacologia , Coelhos , Ratos , Ratos Endogâmicos F344 , Sulfonamidas/farmacologia , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/patologia , Taquicardia Ventricular/fisiopatologia , Taquicardia Ventricular/prevenção & controle , Fatores de Tempo , Fibrilação Ventricular/induzido quimicamente , Fibrilação Ventricular/patologia , Fibrilação Ventricular/fisiopatologia , Fibrilação Ventricular/prevenção & controle
13.
New J Phys ; 10(5): 55001-55024, 2008 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21423856

RESUMO

Low-dimensional iterated map models have been widely used to study action potential dynamics in isolated cardiac cells. Coupled iterated map models have also been widely used to investigate action potential propagation dynamics in one-dimensional (1D) coupled cardiac cells, however, these models are usually empirical and not carefully validated. In this study, we first developed two coupled iterated map models which are the standard forms of diffusively coupled maps and overcome the limitations of the previous models. We then determined the coupling strength and space constant by quantitatively comparing the 1D action potential duration profile from the coupled cardiac cell model described by differential equations with that of the coupled iterated map models. To further validate the coupled iterated map models, we compared the stability conditions of the spatially uniform state of the coupled iterated maps and those of the 1D ionic model and showed that the coupled iterated map model could well recapitulate the stability conditions, i.e., the spatially uniform state is stable unless the state is chaotic. Finally, we combined conduction into the developed coupled iterated map model to study the effects of coupling strength on wave stabilities and showed that the diffusive coupling between cardiac cells tends to suppress instabilities during reentry in a 1D ring and the onset of discordant alternans in a periodically paced 1D cable.

14.
J Am Heart Assoc ; 7(13)2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29929992

RESUMO

BACKGROUND: Ryanodine receptors (RyR) mediate sarcoplasmic reticulum calcium (Ca2+) release and influence myocyte Ca2+ homeostasis and arrhythmias. In cardiac myocytes, RyRs are found in clusters of various sizes and shapes, and RyR cluster size may critically influence normal and arrhythmogenic Ca2+ spark and wave formation. However, the actual RyR cluster sizes at specific Ca2+ spark sites have never been measured in the physiological setting. METHODS AND RESULTS: Here we measured RyR cluster size and Ca2+ sparks simultaneously to assess how RyR cluster size influences Ca2+ sparks and sarcoplasmic reticulum Ca2+ leak. For small RyR cluster sizes (<50), Ca2+ spark frequency is very low but then increases dramatically at larger cluster sizes. In contrast, Ca2+ spark amplitude is nearly maximal even at relatively small RyR cluster size (≈10) and changes little at larger cluster size. These properties agreed with computational simulations of RyR gating within clusters. CONCLUSIONS: Our study explains how this combination of properties may limit arrhythmogenic Ca2+ sparks and wave propagation (at many junctions) while preserving the efficacy and spatial synchronization of Ca2+-induced Ca2+-release during normal excitation-contraction coupling. However, variations in RyR cluster size among individual junctions and RyR sensitivity could exacerbate heterogeneity of local sarcoplasmic reticulum Ca2+ release and arrhythmogenesis under pathological conditions.


Assuntos
Arritmias Cardíacas/metabolismo , Sinalização do Cálcio , Frequência Cardíaca , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Simulação por Computador , Acoplamento Excitação-Contração , Humanos , Ativação do Canal Iônico , Modelos Cardiovasculares , Ratos Sprague-Dawley , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo
15.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 23(4): 713-6, 2006 Aug.
Artigo em Zh | MEDLINE | ID: mdl-17002091

RESUMO

An ECG information database system was established with 1297 ECG information records and clinical informations. The data and the analysis software in this database can be shared with the international physiological signal databases such as MIT-BIH Arrhythmia. The establishment of this database will facilitate the clinical interpretation, diagnosis and prediction of cardiovascular diseases, especially the cardiac sudden death (SCD).


Assuntos
Sistemas de Gerenciamento de Base de Dados , Bases de Dados Factuais , Eletrocardiografia/estatística & dados numéricos , Inteligência Artificial , Cardiopatias/diagnóstico , Humanos
16.
J Appl Physiol (1985) ; 99(6): 2416-22, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16051715

RESUMO

Stress ulcer is clinically prevalent, but the underlying mechanisms are not well understood. This study aimed to investigate the role of sympathovagal imbalance in the development of water immersion restraint stress (WRS)-induced gastric mucosal lesion. Wistar rats were subjected to either restraint stress (RS) (n = 7) or WRS (n = 7) for 5 h. Linear parameters of heart rate variability and Poincaré plot were analyzed on the basis of the surface ECGs. Gastric mucosal lesion was evaluated by gross anatomy and histology. Mean R-R intervals significantly increased (P < 0.001) in a time-dependent manner in the WRS group but slightly decreased (P < 0.001) in the RS group. Root mean square of successive differences of R-R intervals and high-frequency norm (high-frequency power normalized by the total frequency power) were significantly higher in the WRS group than the RS group (P < 0.001). Low-frequency norm and low-to-high-frequency ratio increased significantly 1 h after stress and then declined to similar levels in both groups. The Poincaré plots of R-R intervals in the WRS group shifted right-upwardly and showed dispersed patterns compared with the RS group. Gastric mucosae showed serious hemorrhage, effusion, and structural collapse in the WRS group but remained normal in the RS group. Bilateral cervical vagotomy suppressed the increase of heart rate variability and prevented the gastric mucosal lesion induced by WRS. We conclude that parasympathetic overactivity is the predominant autonomic response to WRS and is most probably the leading mechanism of WRS-induced gastric mucosal lesion in rat.


Assuntos
Mucosa Gástrica/inervação , Mucosa Gástrica/fisiopatologia , Frequência Cardíaca , Imersão/efeitos adversos , Imersão/fisiopatologia , Sistema Nervoso Parassimpático/fisiopatologia , Estômago/inervação , Estômago/fisiopatologia , Animais , Modelos Animais de Doenças , Mucosa Gástrica/patologia , Coração/inervação , Coração/fisiopatologia , Masculino , Ratos , Ratos Wistar , Estresse Fisiológico/etiologia , Estresse Fisiológico/patologia , Estresse Fisiológico/fisiopatologia
17.
Circ Arrhythm Electrophysiol ; 8(6): 1472-80, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26407967

RESUMO

BACKGROUND: Most cardiac arrhythmias occur intermittently. As a cellular precursor of lethal cardiac arrhythmias, early afterdepolarizations (EADs) during action potentials(APs) have been extensively investigated, and mechanisms for the occurrence of EADs on a beat-to-beat basis have been proposed. However, no previous study explains slow fluctuations in EADs, which may underlie intermittency of EAD trains and consequent arrhythmias. We hypothesize that the feedback of intracellular calcium and sodium concentrations ([Na](i) and [Ca](i)) that influence membrane voltage (V) can explain EAD intermittency. METHODS AND RESULTS: AP recordings in rabbit ventricular myocytes revealed intermittent EADs, with slow fluctuations between runs of APs with EADs present or absent. We then used dynamical systems analysis and detailed mathematical models of rabbit ventricular myocytes that replicate the observed behavior and investigated the underlying mechanism. We found that a dominance of inward Na-Ca exchanger current (I(NCX)) over Ca-dependent inactivation of L-type Ca current (I(CaL)) forms a positive feedback between [Ca](i) and V, thus resulting in 2 stable AP states, with and without EADs (ie, bistability). Slow changes in [Na](i) determine the transition between these 2 states, forming a bistable on-off switch of EADs. Tissue simulations showed that this bistable switch of cellular EADs provided both a trigger and a functional substrate for intermittent arrhythmias in homogeneous tissues. CONCLUSIONS: Our study demonstrates that the interaction among V, [Ca](i), and [Na](i) causes slow on-off switching (or bistability) of AP duration in cardiac myocytes and EAD-mediated arrhythmias and suggests a novel possible mechanism for intermittency of cardiac arrhythmias.


Assuntos
Arritmias Cardíacas/etiologia , Sinalização do Cálcio , Cálcio/metabolismo , Sistema de Condução Cardíaco/metabolismo , Potenciais da Membrana , Miócitos Cardíacos/metabolismo , Sódio/metabolismo , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Estimulação Cardíaca Artificial , Simulação por Computador , Retroalimentação Fisiológica , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca , Técnicas In Vitro , Cinética , Masculino , Modelos Cardiovasculares , Coelhos
18.
Circ Arrhythm Electrophysiol ; 8(3): 694-702, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25772542

RESUMO

BACKGROUND: Transient outward K currents (Ito) have been reported both to suppress and to facilitate early afterdepolarizations (EADs) when repolarization reserve is reduced. Here, we used the dynamic clamp technique to analyze how Ito accounts for these paradoxical effects on EADs by influencing the dynamic evolution of repolarization reserve during the action potential. METHODS AND RESULTS: Isolated patch-clamped rabbit ventricular myocytes were exposed to either oxidative stress (H2O2) or hypokalemia to induce bradycardia-dependent EADs at a long pacing cycle length of 6 s, when native rabbit Ito is substantial. EADs disappeared when the pacing cycle length was shortened to 1 s, when Ito becomes negligible because of incomplete recovery from inactivation. During 6-s pacing cycle length, EADs were blocked by the Ito blocker 4-aminopyridine, but reappeared when a virtual current with appropriate Ito-like properties was reintroduced using the dynamic clamp (n=141 trials). During 1-s pacing cycle length in the absence of 4-aminopyridine, adding a virtual Ito-like current (n=1113 trials) caused EADs to reappear over a wide range of Ito conductance (0.005-0.15 nS/pF), particularly when inactivation kinetics were slow (τinact≥20 ms) and the pedestal (noninactivating component) was small (<25% of peak Ito). Faster inactivation or larger pedestals tended to suppress EADs. CONCLUSIONS: Repolarization reserve evolves dynamically during the cardiac action potential. Whereas sufficiently large Ito can suppress EADs, a wide range of intermediate Ito properties can promote EADs by influencing the temporal evolution of other currents affecting late repolarization reserve. These findings raise caution in targeting Ito as an antiarrhythmic strategy.


Assuntos
Potenciais de Ação , Miócitos Cardíacos/metabolismo , Canais de Potássio/metabolismo , Potássio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Bradicardia/metabolismo , Bradicardia/fisiopatologia , Estimulação Cardíaca Artificial , Peróxido de Hidrogênio/farmacologia , Hipopotassemia/metabolismo , Hipopotassemia/fisiopatologia , Técnicas In Vitro , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Coelhos , Fatores de Tempo
19.
J Gen Physiol ; 145(5): 395-404, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25918358

RESUMO

Early afterdepolarizations (EADs) associated with prolongation of the cardiac action potential (AP) can create heterogeneity of repolarization and premature extrasystoles, triggering focal and reentrant arrhythmias. Because the L-type Ca(2+) current (ICa,L) plays a key role in both AP prolongation and EAD formation, L-type Ca(2+) channels (LTCCs) represent a promising therapeutic target to normalize AP duration (APD) and suppress EADs and their arrhythmogenic consequences. We used the dynamic-clamp technique to systematically explore how the biophysical properties of LTCCs could be modified to normalize APD and suppress EADs without impairing excitation-contraction coupling. Isolated rabbit ventricular myocytes were first exposed to H2O2 or moderate hypokalemia to induce EADs, after which their endogenous ICa,L was replaced by a virtual ICa,L with tunable parameters, in dynamic-clamp mode. We probed the sensitivity of EADs to changes in the (a) amplitude of the noninactivating pedestal current; (b) slope of voltage-dependent activation; (c) slope of voltage-dependent inactivation; (d) time constant of voltage-dependent activation; and (e) time constant of voltage-dependent inactivation. We found that reducing the amplitude of the noninactivating pedestal component of ICa,L effectively suppressed both H2O2- and hypokalemia-induced EADs and restored APD. These results, together with our previous work, demonstrate the potential of this hybrid experimental-computational approach to guide drug discovery or gene therapy strategies by identifying and targeting selective properties of LTCC.


Assuntos
Potenciais de Ação , Canais de Cálcio Tipo L/metabolismo , Miócitos Cardíacos/fisiologia , Animais , Células Cultivadas , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Masculino , Potenciais da Membrana , Miócitos Cardíacos/metabolismo , Coelhos
20.
Cardiovasc Res ; 93(2): 242-51, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22049532

RESUMO

AIMS: Fibrosis is known to promote cardiac arrhythmias by disrupting myocardial structure. Given recent evidence that myofibroblasts form gap junctions with myocytes at least in co-cultures, we investigated whether myofibroblast-myocyte coupling can promote arrhythmia triggers, such as early afterdepolarizations (EADs), by directly influencing myocyte electrophysiology. METHODS AND RESULTS: Using the dynamic voltage clamp technique, patch-clamped adult rabbit ventricular myocytes were electrotonically coupled to one or multiple virtual fibroblasts or myofibroblasts programmed with eight combinations of capacitance, membrane resistance, resting membrane potential, and gap junction coupling resistance, spanning physiologically realistic ranges. Myocytes were exposed to oxidative (1 mmol/L H(2)O(2)) or ionic (2.7 mmol/L hypokalaemia) stress to induce bradycardia-dependent EADs. In the absence of myofibroblast-myocyte coupling, EADs developed during slow pacing (6 s), but were completely suppressed by faster pacing (1 s). However, in the presence of myofibroblast-myocyte coupling, EADs could no longer be suppressed by rapid pacing, especially when myofibroblast resting membrane potential was depolarized (-25 mV). Analysis of the myofibroblast-myocyte virtual gap junction currents revealed two components: an early transient-outward I(to)-like current and a late sustained current. Selective elimination of the I(to)-like component prevented EADs, whereas selective elimination of the late component did not. CONCLUSION: Coupling of myocytes to myofibroblasts promotes EAD formation as a result of a mismatch in early vs. late repolarization reserve caused by the I(to)-like component of the gap junction current. These cellular and ionic mechanisms may contribute to the pro-arrhythmic risk in fibrotic hearts.


Assuntos
Arritmias Cardíacas/etiologia , Miócitos Cardíacos/fisiologia , Miofibroblastos/fisiologia , Potenciais de Ação , Animais , Bradicardia/fisiopatologia , Junções Comunicantes/fisiologia , Masculino , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA