Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Indian Pacing Electrophysiol J ; 22(1): 54-57, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34775047

RESUMO

Anti-tachycardia pacing (ATP) is frequently used to terminate ventricular tachycardia (VT), however it is not always successful and may accelerate VT requiring defibrillation. REVRAMP is a novel concept of ATP that involves delivering pacing at a faster rate than VT, but instead of abruptly terminating pacing after eight beats, pacing is gradually slowed until VT continues or normal rhythm is restored. In a pilot study we show that REVRAMP can restore normal rhythm, and that if REVRAMP is unsuccessful, VT is not accelerated.

2.
J Cardiovasc Magn Reson ; 22(1): 13, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32036784

RESUMO

BACKGROUND: Using cardiovascular magnetic resonance imaging (CMR), it is possible to detect diffuse fibrosis of the left ventricle (LV) in patients with atrial fibrillation (AF), which may be independently associated with recurrence of AF after ablation. By conducting CMR, clinical, electrophysiology and biomarker assessment we planned to investigate LV myocardial fibrosis in patients undergoing AF ablation. METHODS: LV fibrosis was assessed by T1 mapping in 31 patients undergoing percutaneous ablation for AF. Galectin-3, coronary sinus type I collagen C terminal telopeptide (ICTP), and type III procollagen N terminal peptide were measured with ELISA. Comparison was made between groups above and below the median for LV extracellular volume fraction (ECV), followed by regression analysis. RESULTS: On linear regression analysis LV ECV had significant associations with invasive left atrial pressure (Beta 0.49, P = 0.008) and coronary sinus ICTP (Beta 0.75, P < 0.001), which remained significant on multivariable regression. CONCLUSION: LV fibrosis in patients with AF is associated with left atrial pressure and invasively measured levels of ICTP turnover biomarker.


Assuntos
Fibrilação Atrial/diagnóstico por imagem , Ventrículos do Coração/diagnóstico por imagem , Imagem Cinética por Ressonância Magnética , Função Ventricular Esquerda , Remodelação Ventricular , Adulto , Idoso , Fibrilação Atrial/sangue , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/cirurgia , Função do Átrio Esquerdo , Pressão Atrial , Biomarcadores/sangue , Proteínas Sanguíneas , Ablação por Cateter , Colágeno Tipo I/sangue , Técnicas Eletrofisiológicas Cardíacas , Feminino , Fibrose , Galectina 3/sangue , Galectinas , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/sangue , Peptídeos/sangue , Valor Preditivo dos Testes , Pró-Colágeno/sangue
3.
Europace ; 20(FI1): f13-f19, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29016773

RESUMO

Aims: The identification of arrhythmogenic right ventricular dysplasia (ARVD) from 12-channel standard electrocardiogram (ECG) is challenging. High density ECG data may identify lead locations and criteria with a higher sensitivity. Methods and results: Eighty-channel ECG recording from patients diagnosed with ARVD and controls were quantified by magnitude and integral measures of QRS and T waves and by a measure (the average silhouette width) of differences in the shapes of the normalized ECG cycles. The channels with the best separability between ARVD patients and controls were near the right ventricular wall, at the third intercostal space. These channels showed pronounced differences in P waves compared to controls as well as the expected differences in QRS and T waves. Conclusion: Multichannel recordings, as in body surface mapping, add little to the reliability of diagnosing ARVD from ECGs. However, repositioning ECG electrodes to a high anterior position can improve the identification of ECG variations in ARVD. Additionally, increased P wave amplitude appears to be associated with ARVD.


Assuntos
Potenciais de Ação , Displasia Arritmogênica Ventricular Direita/diagnóstico , Eletrocardiografia , Frequência Cardíaca , Ventrículos do Coração/fisiopatologia , Adulto , Idoso , Displasia Arritmogênica Ventricular Direita/fisiopatologia , Estudos de Casos e Controles , Eletrocardiografia/instrumentação , Feminino , Ventrículos do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico
4.
Europace ; 19(12): 1944-1950, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28339804

RESUMO

AIMS: Measurement of circulating biomarkers of fibrosis may have a role in selecting patients and treatment strategy for catheter ablation. Pro-collagen type III N-terminal pro-peptide (PIIINP), C-telopeptide of type I collagen (ICTP), fibroblast growth factor 23 (FGF-23), and galectin 3 (gal-3) have all been suggested as possible biomarkers for this indication, but studies assessing whether peripheral levels reflect intra-cardiac levels are scarce. METHODS AND RESULTS: We studied 93 patients undergoing ablation for paroxysmal atrial fibrillation (AF) (n = 63) or non-paroxysmal AF (n = 30). Femoral venous, left and right atrial, and coronary sinus blood were analysed using ELISA to determine biomarker levels. Levels were compared with control patients (n = 36) and baseline characteristics, including left atrial voltage mapping data. C-telopeptide of type I collagen levels were higher in AF than in non-AF patients (P = 0.007). Peripheral ICTP levels were higher than all intra-cardiac levels (P < 0.001). Peripheral gal-3 levels were higher than left atrial levels (P = 0.001). Peripheral levels of FGF-23 and PIIINP were not significantly different from intra-cardiac levels. CS levels of ICTP were higher than right and left atrial levels (P < 0.001). gal-3 was higher in women vs. men (P ≤ 0.001) and with higher body mass index (P ≤ 0.001). ICTP levels increased with reducing ejection fraction (P ≤ 0.012). CONCLUSIONS: Atrial fibrillation patients have higher levels of circulating ICTP than matched non-AF controls. In AF ablation patients, intra-cardiac sampling of FGF-23 or PIIINP gives no further information over peripheral sampling. For gal-3 and ICTP, intra-cardiac sampling may be necessary to assess their association with intra-cardiac processes. None of the biomarkers is related to fibrosis assessed by left atrial voltage.


Assuntos
Fibrilação Atrial/sangue , Fibrilação Atrial/cirurgia , Remodelamento Atrial , Ablação por Cateter , Colágeno Tipo I/sangue , Fatores de Crescimento de Fibroblastos/sangue , Galectina 3/sangue , Átrios do Coração/metabolismo , Fragmentos de Peptídeos/sangue , Peptídeos/sangue , Pró-Colágeno/sangue , Idoso , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/fisiopatologia , Biomarcadores/sangue , Proteínas Sanguíneas , Estudos de Casos e Controles , Tomada de Decisão Clínica , Técnicas Eletrofisiológicas Cardíacas , Ensaio de Imunoadsorção Enzimática , Feminino , Fator de Crescimento de Fibroblastos 23 , Fibrose , Galectinas , Átrios do Coração/patologia , Átrios do Coração/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Seleção de Pacientes , Valor Preditivo dos Testes , Resultado do Tratamento , Função Ventricular Esquerda
5.
Europace ; 16(5): 758-65, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24798966

RESUMO

AIMS: We aim to engineer a computational model of propagation during normal sinus rhythm in the foetal human heart, by modifying models for adult cardiac tissue to match foetal electrocardiogram (fECG) characteristics. The model will be partially validated by fECG data, and applied to explore possible mechanisms of arrhythmogenesis in the foetal heart. METHODS AND RESULTS: Foetal electrocardiograms have been recorded during pregnancy, with P- and T-waves, and the QRS complex, identified by averaging and signal processing. Intervals of the fECG are extracted and used to modify currently available human adult cardiomyocyte models. RR intervals inform models of the pacemaking cells by constraining their rate, the QT interval and its rate dependence constrain models of ventricular cells, and the width of the P-wave, the QR and PR intervals constrain propagation times, conduction velocities, and intercellular coupling. These cell models are coupled into a one-dimensional (1D) model of propagation during normal sinus rhythm in the human foetal heart. We constructed a modular, heterogeneous 1D model for propagation in the foetal heart, and predicted the effects of reduction in L-type Ca(++) current. These include bradycardia and atrioventricular conduction blocks. These may account quantitatively for congenital heart block produced by positive IgG antibodies. CONCLUSION: The fECG can be interpreted mechanistically and quantitatively by using a simple computational model for propagation. After further validation, by clinical recordings of the fECG and the electrophysiological experiments on foetal cardiac cells and tissues, the model may be used to predict the effects of maternally administered pharmaceuticals on the fECG.


Assuntos
Potenciais de Ação/fisiologia , Função Atrial/fisiologia , Simulação por Computador , Coração Fetal/fisiologia , Bloqueio Cardíaco/congênito , Ramos Subendocárdicos/fisiologia , Nó Sinoatrial/fisiologia , Função Ventricular/fisiologia , Eletrocardiografia , Feminino , Átrios do Coração/citologia , Átrios do Coração/fisiopatologia , Bloqueio Cardíaco/fisiopatologia , Ventrículos do Coração/citologia , Ventrículos do Coração/fisiopatologia , Humanos , Modelos Cardiovasculares , Gravidez , Ramos Subendocárdicos/citologia , Ramos Subendocárdicos/fisiopatologia , Nó Sinoatrial/citologia , Nó Sinoatrial/fisiopatologia
6.
Europace ; 16(10): 1524-33, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25085203

RESUMO

AIMS: Atrial anti-arrhythmic effects of ß-adrenoceptor antagonists (ß-blockers) may involve both a suppression of pro-arrhythmic effects of catecholamines, and an adaptational electrophysiological response to chronic ß-blocker use; so-called 'pharmacological remodelling'. In human atrium, such remodelling decreases the transient outward (Ito) and inward rectifier (IK1) K(+) currents, and increases the cellular action potential duration (APD) and effective refractory period (ERP). However, the consequences of these changes on mechanisms of genesis and maintenance of atrial fibrillation (AF) are unknown. Using mathematical modelling, we tested the hypothesis that the long-term adaptational decrease in human atrial Ito and IK1 caused by chronic ß-blocker therapy, i.e. independent of acute electrophysiological effects of ß-blockers, in an otherwise un-remodelled atrium, could suppress AF. METHODS AND RESULTS: Contemporarily, biophysically detailed human atrial cell and tissue models were used to investigate effects of the ß-blocker-based pharmacological remodelling. Chronic ß-blockade remodelling prolonged atrial cell APD and ERP. The incidence of small amplitude APD alternans in the CRN model was reduced. At the 1D tissue level, ß-blocker remodelling decreased the maximum pacing rate at which APs could be conducted. At the three-dimensional organ level, ß-blocker remodelling reduced the life span of re-entry scroll waves. CONCLUSION: This study improves our understanding of the electrophysiological mechanisms of AF suppression by chronic ß-blocker therapy. Atrial fibrillation suppression may involve a reduced propensity for maintenance of re-entrant excitation waves, as a consequence of increased APD and ERP.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Fibrilação Atrial/tratamento farmacológico , Remodelamento Atrial , Simulação por Computador , Potenciais de Ação/efeitos dos fármacos , Fibrilação Atrial/metabolismo , Fibrilação Atrial/fisiopatologia , Humanos , Canais Iônicos/metabolismo
7.
Commun Biol ; 6(1): 651, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37336943

RESUMO

The voltage-gated K+ channel plays a key role in atrial excitability, conducting the ultra-rapid rectifier K+ current (IKur) and contributing to the repolarization of the atrial action potential. In this study, we examine its regulation by hydrogen sulfide (H2S) in HL-1 cardiomyocytes and in HEK293 cells expressing human Kv1.5. Pacing induced remodeling resulted in shorting action potential duration, enhanced both Kv1.5 channel and H2S producing enzymes protein expression in HL-1 cardiomyocytes. H2S supplementation reduced these remodeling changes and restored action potential duration through inhibition of Kv1.5 channel. H2S also inhibited recombinant hKv1.5, lead to nitric oxide (NO) mediated S-nitrosylation and activated endothelial nitric oxide synthase (eNOS) by increased phosphorylation of Ser1177, prevention of NO formation precluded these effects. Regulation of Ikur by H2S has important cardiovascular implications and represents a novel and potential therapeutic target.


Assuntos
Fibrilação Atrial , Sulfeto de Hidrogênio , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Humanos , Sulfeto de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/metabolismo , Fibrilação Atrial/metabolismo , Células HEK293 , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Miócitos Cardíacos/metabolismo
8.
Am J Physiol Heart Circ Physiol ; 302(1): H287-98, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22021329

RESUMO

It has been shown by histology that cardiac myocytes are organized into laminae and this structure is important in function, both influencing the spread of electrical activation and enabling myocardial thickening in systole by laminar sliding. We have carried out high-spatial resolution three-dimensional MRI of the ventricular myolaminae of the entire volume of the isolated rat heart after contrast perfusion [dimeglumine gadopentate (Gd-DTPA)]. Four ex vivo rat hearts were perfused with Gd-DTPA and fixative and high-spatial resolution MRI was performed on a 9.4T MRI system. After MRI, cryosectioning followed by histology was performed. Images from MRI and histology were aligned, described, and quantitatively compared. In the three-dimensional MR images we directly show the presence of laminae and demonstrate that these are highly branching and are absent from much of the subepicardium. We visualized these MRI volumes to demonstrate laminar architecture and quantitatively demonstrated that the structural features observed are similar to those imaged in histology. We showed qualitatively and quantitatively that laminar architecture is similar in the four hearts. MRI can be used to image the laminar architecture of ex vivo hearts in three dimensions, and the images produced are qualitatively and quantitatively comparable with histology. We have demonstrated in the rat that: 1) laminar architecture is consistent between hearts; 2) myolaminae are absent from much of the subepicardium; and 3) although localized orthotropy is present throughout the myocardium, tracked myolaminae are branching structures and do not have a discrete identity.


Assuntos
Meios de Contraste , Gadolínio DTPA , Coração/anatomia & histologia , Imageamento por Ressonância Magnética , Animais , Fixadores , Formaldeído , Interpretação de Imagem Assistida por Computador , Imageamento Tridimensional , Masculino , Perfusão , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Fixação de Tecidos
9.
Eur Biophys J ; 40(5): 627-39, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21234558

RESUMO

Mutations to hERG which result in changes to the rapid delayed rectifier current I(Kr) can cause long and short QT syndromes and are associated with an increased risk of cardiac arrhythmias. Experimental recordings of I(Kr) reveal the effects of mutations at the channel level, but how these changes translate to the cell and tissue levels remains unclear. We used computational models of human ventricular myocytes and tissues to predict and quantify the effects that de novo hERG mutations would have on cell and tissue electrophysiology. Mutations that decreased I(Kr) maximum conductance resulted in an increased cell and tissue action potential duration (APD) and a long QT interval on the electrocardiogram (ECG), whereas those that caused a positive shift in the inactivation curve resulted in a decreased APD and a short QT. Tissue vulnerability to re-entrant arrhythmias was correlated with transmural dispersion of repolarisation, and any change to this vulnerability could be inferred from the ECG QT interval or T wave peak-to-end time. Faster I(Kr) activation kinetics caused cell APD alternans to appear over a wider range of pacing rates and with a larger magnitude, and spatial heterogeneity in these cellular alternans resulted in discordant alternans at the tissue level. Thus, from channel kinetic data, we can predict the tissue-level electrophysiological effects of any hERG mutations and identify how the mutation would manifest clinically, as either a long or short QT syndrome with or without an increased risk of alternans and re-entrant arrhythmias.


Assuntos
Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Simulação por Computador , Canais de Potássio Éter-A-Go-Go/genética , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Mutação , Potenciais de Ação , Animais , Arritmias Cardíacas/fisiopatologia , Canal de Potássio ERG1 , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Predisposição Genética para Doença , Ventrículos do Coração/patologia , Humanos , Modelos Moleculares , Células Musculares/citologia , Células Musculares/metabolismo , Células Musculares/patologia , Nucleotídeos Cíclicos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Reprodutibilidade dos Testes
10.
Front Pharmacol ; 12: 651050, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995065

RESUMO

Chronic exposure to low levels of Carbon Monoxide is associated with an increased risk of cardiac arrhythmia. Microelectrode recordings from rat and guinea pig single isolated ventricular myocytes exposed to CO releasing molecule CORM-2 and excited at 0.2/s show repolarisation changes that develop over hundreds of seconds: action potential prolongation by delayed repolarisation, EADs, multiple EADs and oscillations around the plateau, leading to irreversible repolarisation failure. The measured direct effects of CO on currents in these cells, and ion channels expressed in mammalian systems showed an increase in prolonged late Na+, and a decrease in the maximal T- and L-type Ca++. peak and late Na+, ultra-rapid delayed, delayed rectifier, and the inward rectifier K+ currents. Incorporation of these CO induced changes in maximal currents in ventricular cell models; (Gattoni et al., J. Physiol., 2016, 594, 4193-4224) (rat) and (Luo and Rudy, Circ. Res., 1994, 74, 1071-1096) (guinea-pig) and human endo-, mid-myo- and epi-cardial (O'Hara et al., PLoS Comput. Biol., 2011, 7, e1002061) models, by changes in maximal ionic conductance reproduces these repolarisation abnormalities. Simulations of cell populations with Gaussian distributions of maximal conductance parameters predict a CO induced increase in APD and its variability. Incorporation of these predicted CO induced conductance changes in human ventricular cell electrophysiology into ventricular tissue and wall models give changes in indices for the probability of the initiation of re-entrant arrhythmia.

11.
Altern Lab Anim ; 38 Suppl 1: 87-99, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21275487

RESUMO

The electrical activity of cardiac and uterine tissues has been reconstructed by detailed computer models in the form of virtual tissues. Virtual tissues are biophysically and anatomically detailed, and represent quantitatively predictive models of the physiological and pathophysiological behaviours of tissue within an isolated organ. The cell excitation properties are quantitatively reproduced by equations that describe the kinetics of a few dozen proteins. These equations are derived from experimental measurements of membrane potentials, ionic currents, fluxes, and concentrations. Some of the measurements were taken from human cells and human ion channel proteins expressed in non-human cells, but they were mostly taken from cells of other animal species. Data on tissue geometry and architecture are obtained from the diffusion tensor magnetic resonance imaging of ex vivo or post mortem tissue, and are used to compute the spread of current in the tissue. Cardiac virtual tissues are well established and reproduce normal and pathological patterns of cardiac excitation within the atria or ventricles of the human heart. They have been applied to increase the understanding of normal cardiac electrophysiology, to evaluate the candidate mechanisms for re-entrant arrhythmias that lead to sudden cardiac death, and to predict the tissue level effects of mutant or pharmacologically-modified ion channels. The human full-term virtual uterus is still in development. This virtual tissue reproduces the in vitro behaviour of uterine tissue biopsies, and provides possible mechanisms for premature labour.


Assuntos
Morte Súbita Cardíaca/etiologia , Coração/fisiologia , Nascimento Prematuro/etiologia , Útero/fisiologia , Simulação por Computador , Feminino , Humanos , Miócitos Cardíacos/fisiologia , Engenharia Tecidual
12.
Prog Biophys Mol Biol ; 96(1-3): 112-31, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17905416

RESUMO

Idiopathic short QT syndrome (SQTS) is a recently identified, genetically heterogeneous condition characterised by abbreviated QT intervals and an increased susceptibility to arrhythmia and sudden death. This simulation study identifies mechanisms by which cellular electrophysiological changes in the SQT2 (slow delayed rectifier, IKs, -linked) SQTS variant increases arrhythmia risk. The channel kinetics of the V307L mutation of the KCNQ1 subunit of the IKs channel were incorporated into human ventricular action potential (AP) models and into 1D and 2D transmural tissue simulations. Incorporating the V307L mutation into simulations reproduced defining features of the SQTS: abbreviation of the QT interval, and increases in T wave amplitude and Tpeak-Tend duration. In the single-cell model, the V307L mutation abbreviated ventricular cell AP duration at 90% repolarisation (APD90) and increased the maximal transmural voltage heterogeneity (deltaV) during APs; this resulted in augmented transmural heterogeneity of APD90 and of the effective refractory period (ERP). In the intact tissue model, the vulnerable window for unidirectional conduction block was also increased. In 2D tissue the V307L mutation facilitated and maintained reentrant excitation. Thus, in SQT2 increases in transmural heterogeneity of APD, deltaV, ERP and an increased vulnerable window for unidirectional conduction block generate an electrical substrate favourable to reentrant arrhythmia.


Assuntos
Substituição de Aminoácidos/genética , Arritmias Cardíacas/fisiopatologia , Simulação por Computador , Sistema de Condução Cardíaco/fisiopatologia , Canal de Potássio KCNQ1/genética , Modelos Cardiovasculares , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Eletrocardiografia , Humanos , Canal de Potássio KCNQ1/fisiologia , Síndrome
13.
Prog Biophys Mol Biol ; 96(1-3): 187-208, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17915298

RESUMO

We have constructed computational models of canine ventricular cells and tissues, ultimately combining detailed tissue architecture and heterogeneous transmural electrophysiology. The heterogeneity is introduced by modifying the Hund-Rudy canine cell model in order to reproduce experimentally reported electrophysiological properties of endocardial, midmyocardial (M) and epicardial cells. These models are validated against experimental data for individual ionic current and action potential characteristics, and their rate dependencies. 1D and 3D heterogeneous virtual tissues are constructed, with detailed tissue architecture (anisotropy and orthotropy, due to fibre orientation and sheet structure) of the left ventricular wall wedge extracted from a diffusion tensor imaging data set. The models are used to study the effects of tissue heterogeneity and class III drugs on transmural propagation and tissue vulnerability to re-entry. We have determined relationships between the transmural dispersion of action potential duration (APD) and the vulnerable window in the 1D virtual ventricular wall, and demonstrated how changes in the transmural heterogeneity, and hence tissue vulnerability, can lead to generation of re-entry in the 3D ventricular wedge. Two class III drugs with opposite qualitative effects on transmural APD heterogeneity are considered: d-sotalol that increases transmural APD dispersion, and amiodarone that decreases it. Simulations with the 1D virtual ventricular wall show that under d-sotalol conditions the vulnerable window is substantially wider compared to amiodarone conditions, primarily in the epicardial region where unidirectional conduction block persists until the adjacent M cells are fully repolarised. Further simulations with the 3D ventricular wedge have shown that ectopic stimulation of the epicardial region results in generation of sustained re-entry under d-sotalol conditions, but not under amiodarone conditions or in control. Again, APD increase in M cells was identified as the major contributor to tissue vulnerability--re-entry was initiated primarily due to ectopic excitation propagating around the unidirectional conduction block in the M cell region. This suggests an electrophysiological mechanism for the anti- and proarrhythmic effects of the class III drugs: the relative safety of amiodarone in comparison to d-sotalol can be explained by relatively low transmural APD dispersion, and hence, a narrow vulnerable window and low probability of re-entry in the tissue.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Antiarrítmicos/farmacologia , Arritmias Cardíacas/etiologia , Simulação por Computador , Sistema de Condução Cardíaco/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Modelos Cardiovasculares , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/prevenção & controle , Cães
14.
Prog Biophys Mol Biol ; 98(2-3): 186-97, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19041665

RESUMO

Atrial fibrillation (AF) has been linked to increased inward rectifier potassium current, I(K1), either due to AF-induced electrical remodelling, or from functional changes due to the Kir2.1 V93I mutation. The aim of this simulation study was to identify at cell and tissue levels' mechanisms by which increased I(K1) facilitates and perpetuates AF. The Courtemanche et al. human atrial cell action potential (AP) model was modified to incorporate reported changes in I(K1) induced by the Kir2.1 V93I mutation in both heterozygous (Het) and homozygous (Hom) mutant forms. The modified models for wild type (WT), Het and Hom conditions were incorporated into homogeneous 1D, 2D and 3D tissue models. Restitution curves of AP duration (APD), effective refractory period (ERP) and conduction velocity (CV) were computed and both the temporal and the spatial vulnerability of atrial tissue to re-entry were measured. The lifespan and tip meandering pattern of re-entry were also characterised. For comparison, parallel simulations were performed by incorporating into the Courtmanche et al. model a linear increase in maximal I(K1) conductance. It was found that the gain-in-function of V93I 'mutant'I(K1) led to abbreviated atrial APs and flattened APD, ERP and CV restitution curves. It also hyperpolarised atrial resting membrane potential and slowed down intra-atrial conduction. V93I 'mutant'I(K1) reduced the tissue's temporal vulnerability but increased spatial vulnerability to initiate and sustain re-entry, resulting in an increased overall susceptibility of atrial tissue to arrhythmogenesis. In the 2D model, spiral waves self-terminated for WT (lifespan < 3.3 s) tissue, but persisted in Het and Hom tissues for the whole simulation period (lifespan > 10 s). The tip of the spiral wave meandered more in WT tissue than in Het and Hom tissues. Increased I(K1) due to augmented maximal conductance produced similar results to those of Het and Hom Kir2.1 V93I mutant conditions. In the 3D model the dynamic behaviour of scroll waves was stabilized by increased I(K1). In conclusion, increased I(K1) current, either by the Kir2.1 V93I mutation or by augmented maximal conductance, increases atrial susceptibility to arrhythmia by increasing the lifespan of re-entrant spiral waves and the stability of scroll waves in 3D tissue, thereby facilitating initiation and maintenance of re-entrant circuits.


Assuntos
Fibrilação Atrial/genética , Fibrilação Atrial/fisiopatologia , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Potenciais de Ação , Algoritmos , Animais , Fibrilação Atrial/etiologia , Células COS , Chlorocebus aethiops , Átrios do Coração/patologia , Átrios do Coração/fisiopatologia , Heterozigoto , Homozigoto , Humanos , Imageamento Tridimensional , Modelos Cardiovasculares , Miócitos Cardíacos/fisiologia
15.
Exp Physiol ; 94(5): 496-508, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19139064

RESUMO

Quantitative understanding of the Ca(2+) handling in cardiac ventricular myocytes requires accurate knowledge of cardiac ultrastructure and protein distribution. We have therefore developed high-resolution imaging and analysis approaches to measure the three-dimensional distribution of immunolabelled proteins with confocal microscopy. Labelling of single rat cardiac myocytes with an antibody to the Z-line marker alpha-actinin revealed a complex architecture of sarcomere misalignment across single cells. Double immunolabelling was used to relate the Z-line structure to the distribution of ryanodine receptors (RyRs, the intracellular Ca(2+) release channels) and the transverse tubular system. Both RyR and transverse tubular system distributions exhibited frequent dislocations from the simple planar geometry generally assumed in existing mathematical models. To investigate potential effects of these irregularities on Ca(2+) dynamics, we determined the three-dimensional distribution of RyR clusters within an extended section of a single rat ventricular myocyte to construct a model of stochastic Ca(2+) dynamics with a measured Ca(2+) release unit (CRU) distribution. Calculations with this model were compared with a second model in which all CRUs were placed on flat planes. The model with a realistic CRU distribution supported Ca(2+) waves that spread axially along the cell at velocities of approximately 50 mum s(-1). By contrast, in the model with planar CRU distribution the axial wave spread was slowed roughly twofold and wave propagation often nearly faltered. These results demonstrate that spatial features of the CRU distribution on multiple length scales may significantly affect intracellular Ca(2+) dynamics and must be captured in detailed mechanistic models to achieve quantitative as well as qualitative insight.


Assuntos
Sinalização do Cálcio , Modelos Cardiovasculares , Miócitos Cardíacos/metabolismo , Animais , Técnica Indireta de Fluorescência para Anticorpo , Imageamento Tridimensional , Técnicas In Vitro , Microscopia Confocal , Miócitos Cardíacos/ultraestrutura , Miofibrilas/metabolismo , Miofibrilas/ultraestrutura , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Processos Estocásticos
16.
J Atr Fibrillation ; 12(4): 2217, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32435345

RESUMO

Cardioversion and defibrillation by a single high energy shock applied by myocardial or body surface electrodes is painful, causes long term tissue damage, and is associated with worsening long term outcomes, but is almost always required for treatment of ventricular fibrillation . As a initial step towards developing methods that can terminate ventricular arrhythmias painlessly, we aim to determine if pacing stimuli at a rate of 5/s applied via an implantable cardiac defibrillator (ICD) can modify human ventricular fibrillation. In 8 patients undergoing defibrillation testing of a new/exchanged intracardiac defibrillator, five seconds of pacing at five stimuli per second was applied during the 10-20 seconds of induced ventricular fibrillation before the defibrillation shock was automatically applied, and the cardiac electrograms recorded and analyzed. The high frequency pacing did not entrain the ventricular fibrillation, but altered the dominant frequency in all 8 patients, and modulated the phase computed via the Hilbert Transform, in four of the patients. In this pilot study we demonstrate that high frequency pacing applied via ICD electrodes during VF can alter the dominant frequency and modulate the probability density of the phase of the electrogram of the ventricular fibrillation.

17.
Sci Rep ; 8(1): 15362, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30337648

RESUMO

The human heart develops through complex mechanisms producing morphological and functional changes during gestation. We have recently demonstrated using diffusion tensor MRI that over the relatively short space of 40 days, between 100-140 days gestational age, the ventricular myocardium transforms from a disorganised tissue to the ordered structure characteristic of mature cardiac tissue. However, the genetic basis underpinning this maturation is unclear. Herein, we have used RNA-Seq to establish the developmentally-regulated transcriptome of gene expression in the developing human heart across three gestational ages in the first and second trimester. By comparing 9 weeks gestational age (WGA) with 12 WGA, we find 288 genes show significant differential expression. 305 genes were significantly altered comparing 12 and 16 WGA, and 806 genes differentially expressed between 9 and 16 WGA. Network analysis was used to identify genetic interactions, node properties and gene ontology categories. In summary, we present a comprehensive transcriptomic analysis of human heart development during early gestation, and identify differentially expressed genes during heart development between 9 and 16 weeks, overlapping the first and early second trimester.


Assuntos
Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Coração/embriologia , Coração/fisiologia , Segundo Trimestre da Gravidez , Transcriptoma , Feminino , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Gravidez
18.
Sci Rep ; 8(1): 9159, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29904075

RESUMO

Aberrant uterine myometrial activities in humans are major health issues. However, the cellular and tissue mechanism(s) that maintain the uterine myometrium at rest during gestation, and that initiate and maintain long-lasting uterine contractions during delivery are incompletely understood. In this study we construct a computational model for describing the electrical activity (simple and complex action potentials), intracellular calcium dynamics and mechanical contractions of isolated uterine myocytes from the pregnant rat. The model reproduces variant types of action potentials - from spikes with a smooth plateau, to spikes with an oscillatory plateau, to bursts of spikes - that are seen during late gestation under different physiological conditions. The effects of the hormones oestradiol (via reductions in calcium and potassium selective channel conductance), oxytocin (via an increase in intracellular calcium release) and the tocolytic nifedipine (via a block of L-type calcium channels currents) on action potentials and contractions are also reproduced, which quantitatively match to experimental data. All of these results validated the cell model development. In conclusion, the developed model provides a computational platform for further investigations of the ionic mechanism underlying the genesis and control of electrical and mechanical activities in the rat uterine myocytes.


Assuntos
Sinalização do Cálcio/fisiologia , Modelos Biológicos , Células Musculares/metabolismo , Gravidez/fisiologia , Contração Uterina/fisiologia , Útero/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Feminino , Células Musculares/citologia , Ocitocina/metabolismo , Canais de Potássio/metabolismo , Ratos , Útero/citologia
19.
PLoS One ; 13(1): e0189936, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29293545

RESUMO

AIMS: To test the ability of four circulating biomarkers of fibrosis, and of low left atrial voltage, to predict recurrence of atrial fibrillation after catheter ablation. BACKGROUND: Circulating biomarkers potentially may be used to improve patient selection for atrial fibrillation ablation. Low voltage areas in the left atrium predict arrhythmia recurrence when mapped in sinus rhythm. This study tested type III procollagen N terminal peptide (PIIINP), galectin-3 (gal-3), fibroblast growth factor 23 (FGF-23), and type I collagen C terminal telopeptide (ICTP), and whether low voltage areas in the left atrium predicted atrial fibrillation recurrence, irrespective of the rhythm during mapping. METHODS: 92 atrial fibrillation ablation patients were studied. Biomarker levels in peripheral and intra-cardiac blood were measured with enzyme-linked immunosorbent assay. Low voltage (<0.5mV) was expressed as a proportion of the mapped left atrial surface area. Follow-up was one year. The primary endpoint was recurrence of arrhythmia. The secondary endpoint was a composite of recurrence despite two procedures, or after one procedure if no second procedure was undertaken. RESULTS: The biomarkers were not predictive of either endpoint. After multivariate Cox regression analysis, high proportion of low voltage area in the left atrium was found to predict the primary endpoint in sinus rhythm mapping (hazard ratio 4.323, 95% confidence interval 1.337-13.982, p = 0.014) and atrial fibrillation mapping (hazard ratio 5.195, 95% confidence interval 1.032-26.141, p = 0.046). This effect was also apparent for the secondary endpoint. CONCLUSION: The studied biomarkers do not predict arrhythmia recurrence after catheter ablation. Left atrial voltage is an independent predictor of recurrence, whether the left atrium is mapped in atrial fibrillation or sinus rhythm.


Assuntos
Fibrilação Atrial/cirurgia , Biomarcadores/sangue , Ablação por Cateter/métodos , Átrios do Coração/fisiopatologia , Adulto , Idoso , Fibrilação Atrial/fisiopatologia , Proteínas Sanguíneas , Colágeno Tipo I/sangue , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fibrose , Galectina 3/sangue , Galectinas , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/sangue , Peptídeos/sangue , Pró-Colágeno/sangue , Estudos Prospectivos
20.
Eur J Cardiothorac Surg ; 32(2): 231-49, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17462906

RESUMO

The architecture of the heart remains controversial despite extensive effort and recent advances in imaging techniques. Several opposing and non-mutually compatible models have been proposed to explain cardiac structure, and these models, although limited, have advanced the study and understanding of heart structure, function and development. We describe key areas of similarity and difference, highlight areas of contention and point to the important limitations of these models. Recent research in animal models on the nature, geometry and interaction of cardiac sheet structure allows unification of some seemingly conflicting features of the structural models. Intriguingly, evidence points to significant inter-individual structural variability (within constrained limits) in the canine, leading to the concept of a continuum (or distribution) of cardiac structures. This variability in heart structure partly explains the ongoing debate on myocardial architecture. These developments are used to construct an integrated description of cardiac structure unifying features of fibre, sheet and band architecture that provides a basis for (i) explaining cardiac electromechanics, (ii) computational simulations of cardiac physiology and (iii) designing interventions.


Assuntos
Coração/anatomia & histologia , Modelos Cardiovasculares , Animais , Cães , Ventrículos do Coração/anatomia & histologia , Humanos , Imageamento por Ressonância Magnética/métodos , Microscopia Confocal/métodos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miofibrilas , Terminologia como Assunto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa