Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 602(5): 835-853, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38372694

RESUMEN

Atrial fibrillation (AF) is the most common cardiac arrhythmia and is sustained by spontaneous focal excitations and re-entry. Spontaneous electrical firing in the pulmonary vein (PV) sleeves is implicated in AF generation. The aim of this simulation study was to identify the mechanisms determining the localisation of AF triggers in the PVs and their contribution to the genesis of AF. A novel biophysical model of the canine atria was used that integrates stochastic, spontaneous subcellular Ca2+ release events (SCRE) with regional electrophysiological heterogeneity in ionic properties and a detailed three-dimensional model of atrial anatomy, microarchitecture and patchy fibrosis. Simulations highlighted the importance of the smaller inward rectifier potassium current (IK1 ) in PV cells compared to the surrounding atria, which enabled SCRE more readily to result in delayed-afterdepolarisations that induced triggered activity. There was a leftward shift in the dependence of the probability of triggered activity on sarcoplasmic reticulum Ca2+ load. This feature was accentuated in 3D tissue compared to single cells (Δ half-maximal [Ca2+ ]SR  = 58 µM vs. 22 µM). In 3D atria incorporating electrical heterogeneity, excitations preferentially emerged from the PV region. These triggered focal excitations resulted in transient re-entry in the left atrium. Addition of fibrotic patches promoted localised emergence of focal excitations and wavebreaks that had a more substantial impact on generating AF-like patterns than the PVs. Thus, a reduced IK1 , less negative resting membrane potential, and fibrosis-induced changes of the electrotonic load all contribute to the emergence of complex excitation patterns from spontaneous focal triggers. KEY POINTS: Focal excitations in the atria are most commonly associated with the pulmonary veins, but the mechanisms for this localisation are yet to be elucidated. We applied a multi-scale computational modelling approach to elucidate the mechanisms underlying such localisations. Myocytes in the pulmonary vein region of the atria have a less negative resting membrane potential and reduced time-independent potassium current; we demonstrate that both of these factors promote triggered activity in single cells and tissues. The less negative resting membrane potential also contributes to heterogeneous inactivation of the fast sodium current, which can enable re-entrant-like excitation patterns to emerge without traditional conduction block.


Asunto(s)
Fibrilación Atrial , Venas Pulmonares , Animales , Perros , Fibrilación Atrial/etiología , Calcio , Atrios Cardíacos , Calcio de la Dieta , Potenciales de Acción , Fibrosis , Potasio
2.
J Cardiovasc Electrophysiol ; 33(7): 1450-1459, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35606341

RESUMEN

INTRODUCTION: Esophageal injury is rare but potentially a devastating complication of atrial fibrillation (AF) ablation. The goal here was to provide insight into the short-term natural history of esophageal thermal injury (ETI) after radiofrequency catheter ablation (RFCA) for AFby esophagogastroduodenoscopy (EGD). METHODS: We screened patients who underwent RFCA for AF and EGD based on esophageal late gadolinium enhancement (LGE) in postablation magnetic resonance imaging. Patients with ETI diagnosed with EGD were included. We defined severity of ETI according to Kansas City classification: type 1: erythema; type 2: ulcers (2a: superficial; 2b deep); type 3 perforation (3a: perforation; 3b: perforation with atrioesophageal fistula [AEF]). Repeated EGD was performed within 1-14 days after the last EGD if recommended and possible until any certain healing signs (visible reduction in size without deepening of ETI or complete resolution) were observed. RESULTS: ETI was observed in 62 of 378 patients who underwent EGD after RFCA. Out of these 62 patients with ETI, 21% (13) were type 1, 50% (31) were type 2a and 29% (18) were type 2b at the initial EGD. All esophageal lesions, but one type 2b lesion that developed into an AEF, showed signs of healing in repeated EGD studies within 14 days after the procedure. The one type 2b lesion developing into an AEF showed an increase in size and ulcer deepening in repeat EGD 8 days after the procedure. CONCLUSION: We found that all ETI which did not progress to AEF presented healing signs within 14 days after the procedure and that worsening ETI might be an early signal for developing esophageal perforation.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Fístula Esofágica , Fístula , Fibrilación Atrial/complicaciones , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/cirugía , Ablación por Catéter/efectos adversos , Ablación por Catéter/métodos , Medios de Contraste , Fístula Esofágica/diagnóstico por imagen , Fístula Esofágica/etiología , Fístula/etiología , Gadolinio , Humanos , Complicaciones Posoperatorias/etiología
3.
J Cardiovasc Electrophysiol ; 33(7): 1460-1471, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35644036

RESUMEN

BACKGROUND: Esophageal thermal injury (ETI) is a known and potentially serious complication of catheter ablation for atrial fibrillation. We intended to evaluate the distance between the esophagus and the left atrium posterior wall (LAPW) and its association with esophageal thermal injury. METHODS: A retrospective analysis of 73 patients who underwent esophagogastroduodenoscopy (EGD) after LA radiofrequency catheter ablation for symptomatic atrial fibrillation and pre-ablation magnetic resonance imaging (MRI) was used to identify the minimum distance between the inner lumen of the esophagus and the ablated atrial endocardium (pre-ablation atrial esophageal distance; pre-AED) and occurrence of ETI. Parameters of ablation index (AI, Visitag Surpoint) were collected in 30 patients from the CARTO3 system and compared with assess if ablation strategies and AI further impacted risk of ETI. RESULTS: Pre-AED was significantly larger in patients without ETI than those with ETI (5.23 ± 0.96 mm vs. 4.31 ± 0.75 mm, p < .001). Pre-AED showed high accuracy for predicting ETI with the best cutoff value of 4.37 mm. AI was statistically comparable between Visitag lesion markers with and without associated esophageal late gadolinium enhancement (LGE) detected by postablation MRI in the low-power long-duration ablation group (LPLD, 25-40 W for 10-30 s, 393.16 [308.62-408.86] vs. 406.58 [364.38-451.22], p = .16) and high-power short-duration group (HPSD, 50 W for 5-10 s, 336.14 [299.66-380.11] vs. 330.54 [286.21-384.71], p = .53), respectively. CONCLUSION: Measuring the distance between the LA and the esophagus in pre-ablation LGE-MRI could be helpful in predicting ETI after LAPW ablation.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/epidemiología , Fibrilación Atrial/cirugía , Ablación por Catéter/efectos adversos , Ablación por Catéter/métodos , Medios de Contraste , Esófago/lesiones , Gadolinio , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/cirugía , Humanos , Estudios Retrospectivos
4.
J Electrocardiol ; 69S: 51-54, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34649726

RESUMEN

INTRODUCTION: Accurate reconstruction of cardiac activation wavefronts is crucial for clinical diagnosis, management, and treatment of cardiac arrhythmias. Furthermore, reconstruction of activation profiles within the intramural myocardium has long been impossible because electrical mapping was only performed on the endocardial surface. Recent advancements in electrocardiographic imaging (ECGI) have made endocardial and epicardial activation mapping possible. We propose a novel approach to use both endocardial and epicardial mapping in a combined approach to reconstruct intramural activation times. OBJECTIVE: To implement and validate a combined epicardial/endocardial intramural activation time reconstruction technique. METHODS: We used 11 simulations of ventricular activation paced from sites throughout myocardial wall and extracted endocardial and epicardial activation maps at approximate clinical resolution. From these maps, we interpolated the activation times through the myocardium using thin-plate-spline radial basis functions. We evaluated activation time reconstruction accuracy using root-mean-squared error (RMSE) of activation times and the percent of nodes within 1 ms of the ground truth. RESULTS: Reconstructed intramural activation times showed an RMSE and percentage of nodes within 1 ms of the ground truth simulations of 3 ms and 70%, respectively. In the worst case, the RMSE and percentage of nodes were 4 ms and 60%, respectively. CONCLUSION: We showed that a simple, yet effective combination of clinical endocardial and epicardial activation maps can accurately reconstruct intramural wavefronts. Furthermore, we showed that this approach provided robust reconstructions across multiple intramural stimulation sites.


Asunto(s)
Electrocardiografía , Humanos , Mapeo del Potencial de Superficie Corporal , Estimulación Cardíaca Artificial , Estudios de Factibilidad
5.
J Electrocardiol ; 68: 56-64, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34339897

RESUMEN

OBJECTIVE: Test the hypothesis that exercise and pharmacological cardiac stressors create different electrical ischemic signatures. INTRODUCTION: Current clinical stress tests for detecting ischemia lack sensitivity and specificity. One unexplored source of the poor detection is whether pharmacological stimulation and regulated exercise produce identical cardiac stress. METHODS: We used a porcine model of acute myocardial ischemia in which animals were instrumented with transmural plunge-needle electrodes, an epicardial sock array, and torso arrays to simultaneously measure cardiac electrical signals within the heart wall, the epicardial surface, and the torso surface, respectively. Ischemic stress via simulated exercise and pharmacological stimulation were created with rapid electrical pacing and dobutamine infusion, respectively, and mimicked clinical stress tests of five 3-minute stages. Perfusion to the myocardium was regulated by a hydraulic occluder around the left anterior descending coronary artery. Ischemia was measured as deflections to the ST-segment on ECGs and electrograms. RESULTS: Across eight experiments with 30 (14 simulated exercise and 16 dobutamine) ischemic interventions, the spatial correlations between exercise and pharmacological stress diverged at stage three or four during interventions (p<0.05). We found more detectable ST-segment changes on the epicardial surface during simulated exercise than with dobutamine (p<0.05). The intramyocardial ischemia formed during simulated exercise had larger ST40 potential gradient magnitudes (p<0.05). CONCLUSION: We found significant differences on the epicardium between cardiac stress types using our experimental model, which became more pronounced at the end stages of each test. A possible mechanism for these differences was the larger ST40 potential gradient magnitudes within the myocardium during exercise. The presence of microvascular dysfunction during exercise and its absence during dobutamine stress may explain these differences.


Asunto(s)
Electrocardiografía , Isquemia Miocárdica , Animales , Dobutamina/farmacología , Prueba de Esfuerzo , Isquemia , Isquemia Miocárdica/diagnóstico , Pericardio , Porcinos
6.
J Electrocardiol ; 69S: 38-44, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34384615

RESUMEN

BACKGROUND: Acute myocardial ischemia has several characteristic ECG findings, including clinically detectable ST-segment deviations. However, the sensitivity and specificity of diagnosis based on ST-segment changes are low. Furthermore, ST-segment deviations have been shown to be transient and spontaneously recover without any indication the ischemic event has subsided. OBJECTIVE: Assess the transient recovery of ST-segment deviations on remote recording electrodes during a partial occlusion cardiac stress test and compare them to intramyocardial ST-segment deviations. METHODS: We used a previously validated porcine experimental model of acute myocardial ischemia with controllable ischemic load and simultaneous electrical measurements within the heart wall, on the epicardial surface, and on the torso surface. Simulated cardiac stress tests were induced by occluding a coronary artery while simultaneously pacing rapidly or infusing dobutamine to stimulate cardiac function. Postexperimental imaging created anatomical models for data visualization and quantification. Markers of ischemia were identified as deviations in the potentials measured at 40% of the ST-segment. Intramural cardiac conduction speed was also determined using the inverse gradient method. We assessed changes in intramyocardial ischemic volume proportion, conduction speed, clinical presence of ischemia on remote recording arrays, and regional changes to intramyocardial ischemia. We defined the peak deviation response time as the time interval after onset of ischemia at which maximum ST-segment deviation was achieved, and ST-recovery time was the interval when ST deviation returned to below thresholded of ST elevation. RESULTS: In both epicardial and torso recordings, the peak ST-segment deviation response time was 4.9±1.1 min and the ST-recovery time was approximately 7.9±2.5 min, both well before the termination of the ischemic stress. At peak response time, conduction speed was reduced by 50% and returned to near baseline at ST-recovery. The overall ischemic volume proportion initially increased, on average, to 37% at peak response time; however, it recovered to only 30% at the ST-recovery time. By contrast, the subepicardial region of the myocardial wall showed 40% ischemic volume at peak response time and recovered much more strongly to 25% as epicardial ST-segment deviations returned to baseline. CONCLUSION: Our data show that remote ischemic signal recovery correlates with a recovery of the subepicardial myocardium, whereas subendocardial ischemic development persists.


Asunto(s)
Electrocardiografía , Isquemia Miocárdica , Animales , Corazón , Isquemia , Isquemia Miocárdica/diagnóstico , Porcinos , Torso
7.
J Electrocardiol ; 66: 86-94, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33836460

RESUMEN

INTRODUCTION: Acute myocardial ischemia occurs when coronary perfusion to the heart is inadequate, which can perturb the highly organized electrical activation of the heart and can result in adverse cardiac events including sudden cardiac death. Ischemia is known to influence the ST and repolarization phases of the ECG, but it also has a marked effect on propagation (QRS); however, studies investigating propagation during ischemia have been limited. METHODS: We estimated conduction velocity (CV) and ischemic stress prior to and throughout 20 episodes of experimentally induced ischemia in order to quantify the progression and correlation of volumetric conduction changes during ischemia. To estimate volumetric CV, we 1) reconstructed the activation wavefront; 2) calculated the elementwise gradient to approximate propagation direction; and 3) estimated conduction speed (CS) with an inverse-gradient technique. RESULTS: We found that acute ischemia induces significant conduction slowing, reducing the global median speed by 20 cm/s. We observed a biphasic response in CS (acceleration then deceleration) early in some ischemic episodes. Furthermore, we noted a high temporal correlation between ST-segment changes and CS slowing; however, when comparing these changes over space, we found only moderate correlation (corr. = 0.60). DISCUSSION: This study is the first to report volumetric CS changes (acceleration and slowing) during episodes of acute ischemia in the whole heart. We showed that while CS changes progress in a similar time course to ischemic stress (measured by ST-segment shifts), the spatial overlap is complex and variable, showing extreme conduction slowing both in and around regions experiencing severe ischemia.


Asunto(s)
Sistema de Conducción Cardíaco , Isquemia Miocárdica , Arritmias Cardíacas , Electrocardiografía , Corazón , Humanos
8.
J Cardiovasc Electrophysiol ; 31(2): 450-456, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31916637

RESUMEN

BACKGROUND: Recent guidelines recommend a 3-month blanking period after atrial fibrillation (AF) ablations, which are based on clinical observation. Our goal was to quantify the timeline of the radiofrequency ablation lesion maturation using serial late gadolinium enhancement-magnetic resonance imaging (LGE-MRI) and to develop a blanking period estimate based on visible lesion maturation. METHODS: Inclusion criteria targeted patients who underwent AF ablation and at least four MRI scans: at baseline before ablation, within 24 hours after (acute), between 24 hours and 90 days after (subacute), and more than 90 days after ablation (chronic). Central nonenhanced (NE) and surrounding hyperenhanced (HE) area volumes were measured and normalized to chronic lesion volume. RESULTS: This study assessed 75 patients with 309 MRIs. The acute lesion was heterogeneous with a HE region surrounding a central NE region in LGE-MRI; the acute volume of the total (HE + NE) lesion was 2.62 ± 0.46 times larger than that of the chronic lesion. Acute T2-weighted imaging also showed a relatively large area of edema. Both NE and HE areas gradually receded over time and NE was not observed after 30 days. Larger initial NE volume was associated with a significantly greater chronic scar volume and this total lesion volume receded to equal the chronic lesion size at approximately 72.5 days (95% prediction interval: 57.4-92.2). CONCLUSION: On the basis of serial MRI, atrial ablation lesions are often fully mature before the typical 90-day blanking period, which could support more timely clinical decision making for arrhythmia recurrence.


Asunto(s)
Fibrilación Atrial/cirugía , Ablación por Catéter , Cicatriz/diagnóstico por imagen , Atrios Cardíacos/cirugía , Frecuencia Cardíaca , Imagen por Resonancia Magnética , Potenciales de Acción , Anciano , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/fisiopatología , Remodelación Atrial , Ablación por Catéter/efectos adversos , Cicatriz/etiología , Cicatriz/fisiopatología , Medios de Contraste/administración & dosificación , Femenino , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/fisiopatología , Humanos , Masculino , Meglumina/análogos & derivados , Persona de Mediana Edad , Compuestos Organometálicos , Valor Predictivo de las Pruebas , Recurrencia , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
9.
J Cardiovasc Electrophysiol ; 31(11): 2824-2832, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32931635

RESUMEN

INTRODUCTION: Late gadolinium enhancement (LGE) cardiac magnetic resonance imaging (MRI) can be used to detect postablation atrial scar (PAAS) but its reproducibility and reliability in clinical scans across different magnetic flux densities and scar detection methods are unknown. METHODS: Patients (n = 45) having undergone two consecutive MRIs (3 months apart) on 3T and 1.5T scanners were studied. We compared PAAS detection reproducibility using four methods of thresholding: simple thresholding, Otsu thresholding, 3.3 standard deviations (SD) above blood pool (BP) mean intensity, and image intensity ratio (IIR). We performed a texture study by dividing the left atrial wall intensity histogram into deciles and evaluated the correlation of the same decile of the two scans as well as to a randomized distribution of intensities, quantified using Dice Similarity Coefficient (DSC). RESULTS: The choice of scanner did not significantly affect the reproducibility. The scar detection performed by Otsu thresholding (DSC of 71.26 ± 8.34) resulted in a better correlation of the two scans compared with the methods of 3.3 SD above BP mean intensity (DSC of 57.78 ± 21.2, p < .001) and IIR above 1.61 (DSC of 45.76 ± 29.55, p <.001). Texture analysis showed that correlation only for voxels with intensities in deciles above the 70th percentile of wall intensity histogram was better than random distribution (p < .001). CONCLUSIONS: Our results demonstrate that clinical LGE-MRI can be reliably used for visualizing PAAS across different magnetic flux densities if the threshold is greater than 70th percentile of the wall intensity distribution. Also, atrial wall-based thresholding is better than BP-based thresholding for reproducible PAAS detection.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/patología , Fibrilación Atrial/cirugía , Ablación por Catéter/efectos adversos , Cicatriz/diagnóstico por imagen , Cicatriz/etiología , Cicatriz/patología , Medios de Contraste , Gadolinio , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/patología , Atrios Cardíacos/cirugía , Humanos , Imagen por Resonancia Magnética , Reproducibilidad de los Resultados
10.
J Electrocardiol ; 51(6S): S67-S71, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30029778

RESUMEN

BACKGROUND: Differentiating between atrial fibrillation (AF) and atrial tachycardia (AT) or atrial flutter (AFL) on surface ECG can be challenging. The same problem arises in animal models of AF, in which atrial arrhythmias are induced by pacing or pharmacological intervention with the goal of making mechanistic determinations. Some of these induced arrhythmias can be AFL or AT, even though it might appear as AF on the body-surface ECG based on irregular R-R intervals. We hypothesize that a dominant frequency (DF) analysis of the ECG can differentiate between the two distinct arrhythmias, even when it is not evident by the presence of flutter waves or beat-to-beat regularity when looking at brief recordings. METHODS: Canine model (n = 15, 10 controls and 5 Persistent AF animals with >6 months of AF) was used to test the hypothesis. Atrial arrhythmia was induced by rapid atrial pacing. Five blinded observers evaluated the 3­lead surface ECGs recorded during atrial arrhythmia and classified the rhythm as AFL/AT or AF. The 64-electrode Constellation (Boston Scientific) catheter was used to acquire left (entire group) and right (7 of 10 controls) atrial intracardiac electrograms. For the surface ECG and the intracardiac electrograms, Welch method with a hamming window and 50% overlap was used to calculate DF of two-minute segments. Mean of standard deviations of the DF values were calculated for both ECGs and intracardiac EGMs. Ground truth came from activations maps and DF analysis derived from the intracardiac electrograms recorded in the two chambers. RESULTS: Rapid pacing induced atrial arrhythmias in all the control animals. The ECG in 8 of the 10 control cases was read as AF by at least 80% percent of observers even though the EGMs from the Constellation showed organized activation and consistent DF (STD of DF < 0.001) in all the electrodes confirming the arrhythmia as AFL in 10/10 cases. In the persistent AF group, the DF from the three lead ECGs were significantly different (Mean of STDs = 2.65 ±â€¯0.99) whereas the DF in the control animals with AFL was consistent across all ECG channels (Mean of STDs < 0.001), and the DF in the control animals ECGs was in agreement with the DF of the intracardiac electrograms. CONCLUSION: Surface ECG recordings can mimic AF even when the underlying atrial arrhythmia is AFL in control canine models. DF variation of the signals from multiple surface ECG leads can help differentiate between the AF and AFL.


Asunto(s)
Fibrilación Atrial/diagnóstico , Aleteo Atrial/diagnóstico , Electrocardiografía/métodos , Animales , Fibrilación Atrial/fisiopatología , Aleteo Atrial/fisiopatología , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Perros
11.
J Electrocardiol ; 51(6S): S116-S120, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30122455

RESUMEN

BACKGROUND: Myocardial ischemia has a complex and time-varying electrocardiographic signature that is used to diagnose and stratify severity. Despite the ubiquitous clinical use of the ECG to detect ischemia, the sensitivity and specificity of ECG based detection of myocardial ischemia are still inadequate. PURPOSE: The purpose of this study was to compare, using animal models, the performance of several traditional ECG-based metrics for detecting acute ischemia against two novel metrics, the Laplacian Eigenmap (LE) parameters and a three-dimensional estimate of Conduction Velocity (CV). METHODS: LE is a machine learning technique that reduces the dimensions of simultaneously recorded time signals using a non-linear embedding followed by an singular value decomposition to represent each multichannel recording as a single trajectory on a manifold. Perturbations in the trajectories suggest the presence of myocardial ischemia. CV was computed using a tetrahedral mesh created from the electrode locations of transmural plunge needles. To validate the results, we used electrograms collected over 95 episodes of acutely induced myocardial ischemia in 15 canine and 2 porcine subjects. The LE and CV metrics were compared against traditional metrics derived from the ST segment, the T wave, the QRS of the same electrograms. The response time and robustness of each metric was quantified using parameters we defined as time to threshold (TTT) and contrast ratio (CR). RESULTS: The temporal performance of the metrics evaluated throughout the ischemic episodes showed a consistent relationship; the LE metrics changed earlier than those from the T wave, which were followed by those from the ST segment, and finally from the QRS. The CV results showed median drops in conduction velocity throughout the perfusion bed of more than 23% in canines and over 12% during half of the induced ischemia episodes in swine. The other half of the episodes in swine produced a 76% drop. CONCLUSIONS: Our results suggest that the LE metric is more sensitive to acute ischemia than traditional single parameters used in previous studies, likely because it incorporates the entire QRST across multiple electrodes in a way that captures their most salient features in a low-dimensional space. The estimates of conduction velocity suggest substantial, in some cases dramatic slowing of the spread of activation, a finding that is not surprising but has not been documented in such three-dimensional detail before. The experiments and these new metrics provide a means to both explore details of the acute ischemic response not available from humans and suggest a path to translate this knowledge into improvements in clinical scoring of ischemia.


Asunto(s)
Electrocardiografía/métodos , Aprendizaje Automático , Isquemia Miocárdica/diagnóstico , Animales , Modelos Animales de Enfermedad , Perros , Sensibilidad y Especificidad , Porcinos , Factores de Tiempo
12.
J Cardiovasc Electrophysiol ; 27(10): 1220-1229, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27447370

RESUMEN

INTRODUCTION: Large animal models of progressive atrial fibrosis would provide an attractive platform to study relationship between structural and electrical remodeling in atrial fibrillation (AF). Here we established a new transgenic goat model of AF with cardiac specific overexpression of TGF-ß1 and investigated the changes in the cardiac structure and function leading to AF. METHODS AND RESULTS: Transgenic goats with cardiac specific overexpression of constitutively active TGF-ß1 were generated by somatic cell nuclear transfer. We examined myocardial tissue, ECGs, echocardiographic data, and AF susceptibility in transgenic and wild-type control goats. Transgenic goats exhibited significant increase in fibrosis and myocyte diameters in the atria compared to controls, but not in the ventricles. P-wave duration was significantly greater in transgenic animals starting at 12 months of age, but no significant chamber enlargement was detected, suggesting conduction slowing in the atria. Furthermore, this transgenic goat model exhibited a significant increase in AF vulnerability. Six of 8 transgenic goats (75%) were susceptible to AF induction and exhibited sustained AF (>2 minutes), whereas none of 6 controls displayed sustained AF (P < 0.01). Length of induced AF episodes was also significantly greater in the transgenic group compared to controls (687 ± 212.02 seconds vs. 2.50 ± 0.88 seconds, P < 0.0001), but no persistent or permanent AF was observed. CONCLUSION: A novel transgenic goat model with a substrate for AF was generated. In this model, cardiac overexpression of TGF-ß1 led to an increase in fibrosis and myocyte size in the atria, and to progressive P-wave prolongation. We suggest that these factors underlie increased AF susceptibility.


Asunto(s)
Fibrilación Atrial/metabolismo , Remodelación Atrial , Cabras/genética , Atrios Cardíacos/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Potenciales de Acción , Animales , Animales Modificados Genéticamente , Fibrilación Atrial/genética , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Biopsia , Ecocardiografía , Electrocardiografía , Fibrosis , Predisposición Genética a la Enfermedad , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Frecuencia Cardíaca , Humanos , Microscopía Confocal , Fenotipo , Factor de Crecimiento Transformador beta1/genética
13.
J Cardiovasc Electrophysiol ; 26(12): 1352-60, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26249367

RESUMEN

INTRODUCTION: Patients with paroxysmal atrial fibrillation (AF) often transition between sinus rhythm and AF. For AF to initiate there must be both a trigger and a substrate that facilitates reentrant activity. This trigger is often caused by a premature atrial contraction or focal activations within the atrium. We hypothesize that specific architectures of fibrosis alter local conduction to enable AF. METHODS AND RESULTS: Control goats (n = 13) and goats in chronic AF (for an average of 6 months, n = 6) had a high-density electrode plaque placed on the LA appendage. Conduction patterns following a premature atrial contraction, caused by an electrical stimulation, were quantified to determine regions of conduction slowing. These regions were compared to architecture, either diffuse fibrosis or regions of obstructive fibrosis, and overall fibrosis levels as determined by histology from the mapped region. The chronic AF goats had more obstructive fibrosis than the controls (17.5 ± 8.0 fibers/mm(2) vs. 8.6 ± 3.0 fibers/mm(2)). Conduction velocity of the AF goats was significantly slowed compared to the control goats in the transverse direction (0.40 ± 0.04 m/s vs. 0.53 ± 0.15 m/s) but not in the longitudinal direction (0.70 ± 0.27 m/s vs. 0.76 ± 0.18 m/s). CONCLUSIONS: AF-induced atrial remodeling leads to increased obstructive fibrosis and conduction velocity slowing transverse to fiber orientation following premature stimuli. The decrease in conduction velocity causes a decrease in the cardiac wavelength, and increases the likelihood of reentry and AF onset.


Asunto(s)
Fibrilación Atrial/complicaciones , Fibrilación Atrial/fisiopatología , Atrios Cardíacos/patología , Animales , Fibrilación Atrial/patología , Complejos Atriales Prematuros/complicaciones , Complejos Atriales Prematuros/etiología , Complejos Atriales Prematuros/fisiopatología , Remodelación Atrial , Enfermedad Crónica , Estimulación Eléctrica , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Fenómenos Electrofisiológicos , Fibrosis , Cabras , Sistema de Conducción Cardíaco , Marcapaso Artificial
14.
J Cardiovasc Electrophysiol ; 25(7): 774-80, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24762029

RESUMEN

BACKGROUND: Voltage mapping is an important tool for characterizing proarrhythmic electrophysiological substrate, yet it is subject to geometric factors that influence bipolar amplitudes and thus compromise performance. The aim of this study was to characterize the impact of catheter orientation on the ability of bipolar amplitudes to accurately discriminate between healthy and diseased tissues. METHODS AND RESULTS: We constructed a 3-dimensional, in silico, bidomain model of cardiac tissue containing transmural lesions of varying diameter. A planar excitation wave was stimulated and electrograms were sampled with a realistic catheter model at multiple positions and orientations. We carried out validation studies in animal experiments of acute ablation lesions mapped with a clinical mapping system. Bipolar electrograms sampled at higher inclination angles of the catheter with respect to the tissue demonstrated improvements in both sensitivity and specificity of lesion detection. Removing low-voltage electrograms with concurrent activation of both electrodes, suggesting false attenuation of the bipolar electrogram due to alignment with the excitation wavefront, had little effect on the accuracy of voltage mapping. CONCLUSIONS: Our results demonstrate possible mechanisms for the impact of catheter orientation on voltage mapping accuracy. Moreover, results from our simulations suggest that mapping accuracy may be improved by selectively controlling the inclination of the catheter to record at higher angles with respect to the tissue.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Cateterismo Cardíaco , Simulación por Computador , Sistema de Conducción Cardíaco/fisiopatología , Modelos Cardiovasculares , Potenciales de Acción , Animales , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/cirugía , Cateterismo Cardíaco/instrumentación , Catéteres Cardíacos , Ablación por Catéter , Técnicas Electrofisiológicas Cardíacas/instrumentación , Sistema de Conducción Cardíaco/cirugía , Frecuencia Cardíaca , Humanos , Cinética , Modelos Animales , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Procesamiento de Señales Asistido por Computador , Porcinos
15.
J Cardiovasc Electrophysiol ; 25(5): 457-463, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24383404

RESUMEN

BACKGROUND: Three-dimensional electroanatomic mapping (EAM) is routinely used to mark ablated areas during radiofrequency ablation. We hypothesized that, in atrial fibrillation (AF) ablation, EAM overestimates scar formation in the left atrium (LA) when compared to the scar seen on late-gadolinium enhancement magnetic resonance imaging (LGE-MRI). METHODS AND RESULTS: Of the 235 patients who underwent initial ablation for AF at our institution between August 2011 and December 2012, we retrospectively identified 70 patients who had preprocedural magnetic resonance angiography merged with LA anatomy in EAM software and had a 3-month postablation LGE-MRI for assessment of scar. Ablated area was marked intraprocedurally using EAM software and quantified retrospectively. Scarred area was quantified in 3-month postablation LGE-MRI. The mean ablated area in EAM was 30.5 ± 7.5% of the LA endocardial surface and the mean scarred area in LGE-MRI was 13.9 ± 5.9% (P < 0.001). This significant difference in the ablated area marked in the EAM and scar area in the LGE-MRI was present for each of the 3 independent operators. Complete pulmonary vein (PV) encirclement representing electrical isolation was observed in 87.8% of the PVs in EAM as compared to only 37.4% in LGE-MRI (P < 0.001). CONCLUSIONS: In AF ablation, EAM significantly overestimates the resultant scar as assessed with a follow-up LGE-MRI.


Asunto(s)
Fibrilación Atrial/terapia , Ablación por Catéter , Cicatriz/diagnóstico , Técnicas Electrofisiológicas Cardíacas , Atrios Cardíacos/cirugía , Angiografía por Resonancia Magnética , Venas Pulmonares/cirugía , Cirugía Asistida por Computador , Anciano , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/fisiopatología , Cicatriz/patología , Cicatriz/fisiopatología , Femenino , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Persona de Mediana Edad , Variaciones Dependientes del Observador , Valor Predictivo de las Pruebas , Venas Pulmonares/patología , Venas Pulmonares/fisiopatología , Reproducibilidad de los Resultados , Estudios Retrospectivos , Programas Informáticos , Resultado del Tratamiento , Utah
16.
Artículo en Inglés | MEDLINE | ID: mdl-38587576

RESUMEN

BACKGROUND: The immediate impact of catheter ablation on left atrial mechanical function and the timeline for its recovery in patients undergoing ablation for atrial fibrillation (AF) remain uncertain. The mechanical function response to catheter ablation in patients with different AF types is poorly understood. METHODS: A total of 113 AF patients were included in this retrospective study. Each patient had three magnetic resonance imaging (MRI) studies in sinus rhythm: one pre-ablation, one immediate post-ablation (within 2 days after ablation), and one post-ablation follow-up MRI (≤ 3 months). We used feature tracking in the MRI cine images to determine peak longitudinal atrial strain (PLAS). We evaluated the change in strain from pre-ablation, immediately after ablation to post-ablation follow-up in a short-term study (< 50 days) and a 3-month study (3 months after ablation). RESULTS: The PLAS exhibited a notable reduction immediately after ablation, compared to both pre-ablation levels and those observed in follow-up studies conducted at short-term (11.1 ± 9.0 days) and 3-month (69.6 ± 39.6 days) intervals. However, there was no difference between follow-up and pre-ablation PLAS. The PLAS returned to 95% pre-ablation level within 10 days. Paroxysmal AF patients had significantly higher pre-ablation PLAS than persistent AF patients in pre-ablation MRIs. Both type AF patients had significantly lower immediate post-ablation PLAS compared with pre-ablation and post-ablation PLAS. CONCLUSION: The present study suggested a significant drop in PLAS immediately after ablation. Left atrial mechanical function recovered within 10 days after ablation. The drop in PLAS did not show a substantial difference between paroxysmal and persistent AF patients.

17.
Biomech Model Mechanobiol ; 23(3): 927-940, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38361087

RESUMEN

Central to the clinical adoption of patient-specific modeling strategies is demonstrating that simulation results are reliable and safe. Indeed, simulation frameworks must be robust to uncertainty in model input(s), and levels of confidence should accompany results. In this study, we applied a coupled uncertainty quantification-finite element (FE) framework to understand the impact of uncertainty in vascular material properties on variability in predicted stresses. Univariate probability distributions were fit to material parameters derived from layer-specific mechanical behavior testing of human coronary tissue. Parameters were assumed to be probabilistically independent, allowing for efficient parameter ensemble sampling. In an idealized coronary artery geometry, a forward FE model for each parameter ensemble was created to predict tissue stresses under physiologic loading. An emulator was constructed within the UncertainSCI software using polynomial chaos techniques, and statistics and sensitivities were directly computed. Results demonstrated that material parameter uncertainty propagates to variability in predicted stresses across the vessel wall, with the largest dispersions in stress within the adventitial layer. Variability in stress was most sensitive to uncertainties in the anisotropic component of the strain energy function. Moreover, unary and binary interactions within the adventitial layer were the main contributors to stress variance, and the leading factor in stress variability was uncertainty in the stress-like material parameter that describes the contribution of the embedded fibers to the overall artery stiffness. Results from a patient-specific coronary model confirmed many of these findings. Collectively, these data highlight the impact of material property variation on uncertainty in predicted artery stresses and present a pipeline to explore and characterize forward model uncertainty in computational biomechanics.


Asunto(s)
Vasos Coronarios , Análisis de Elementos Finitos , Estrés Mecánico , Humanos , Vasos Coronarios/fisiología , Incertidumbre , Fenómenos Biomecánicos , Modelos Cardiovasculares , Simulación por Computador , Anisotropía
18.
ArXiv ; 2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38344225

RESUMEN

Central to the clinical adoption of patient-specific modeling strategies is demonstrating that simulation results are reliable and safe. Indeed, simulation frameworks must be robust to uncertainty in model input(s), and levels of confidence should accompany results. In this study, we applied a coupled uncertainty quantification-finite element (FE) framework to understand the impact of uncertainty in vascular material properties on variability in predicted stresses. Univariate probability distributions were fit to material parameters derived from layer-specific mechanical behavior testing of human coronary tissue. Parameters were assumed to be probabilistically independent, allowing for efficient parameter ensemble sampling. In an idealized coronary artery geometry, a forward FE model for each parameter ensemble was created to predict tissue stresses under physiologic loading. An emulator was constructed within the UncertainSCI software using polynomial chaos techniques, and statistics and sensitivities were directly computed. Results demonstrated that material parameter uncertainty propagates to variability in predicted stresses across the vessel wall, with the largest dispersions in stress within the adventitial layer. Variability in stress was most sensitive to uncertainties in the anisotropic component of the strain energy function. Moreover, unary and binary interactions within the adventitial layer were the main contributors to stress variance, and the leading factor in stress variability was uncertainty in the stress-like material parameter that describes the contribution of the embedded fibers to the overall artery stiffness. Results from a patient-specific coronary model confirmed many of these findings. Collectively, these data highlight the impact of material property variation on uncertainty in predicted artery stresses and present a pipeline to explore and characterize forward model uncertainty in computational biomechanics.

19.
Biophys J ; 104(12): 2764-73, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23790385

RESUMEN

Atrial fibrillation (AF), the most common arrhythmia in humans, is initiated when triggered activity from the pulmonary veins propagates into atrial tissue and degrades into reentrant activity. Although experimental and clinical findings show a correlation between atrial fibrosis and AF, the causal relationship between the two remains elusive. This study used an array of 3D computational models with different representations of fibrosis based on a patient-specific atrial geometry with accurate fibrotic distribution to determine the mechanisms by which fibrosis underlies the degradation of a pulmonary vein ectopic beat into AF. Fibrotic lesions in models were represented with combinations of: gap junction remodeling; collagen deposition; and myofibroblast proliferation with electrotonic or paracrine effects on neighboring myocytes. The study found that the occurrence of gap junction remodeling and the subsequent conduction slowing in the fibrotic lesions was a necessary but not sufficient condition for AF development, whereas myofibroblast proliferation and the subsequent electrophysiological effect on neighboring myocytes within the fibrotic lesions was the sufficient condition necessary for reentry formation. Collagen did not alter the arrhythmogenic outcome resulting from the other fibrosis components. Reentrant circuits formed throughout the noncontiguous fibrotic lesions, without anchoring to a specific fibrotic lesion.


Asunto(s)
Fibrilación Atrial/fisiopatología , Remodelación Atrial , Modelos Cardiovasculares , Potenciales de Acción , Anciano , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Proliferación Celular , Colágeno/metabolismo , Femenino , Fibrosis/patología , Fibrosis/fisiopatología , Uniones Comunicantes/patología , Atrios Cardíacos/patología , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Miofibroblastos/metabolismo , Miofibroblastos/fisiología , Comunicación Paracrina
20.
Pacing Clin Electrophysiol ; 36(4): 467-76, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23356963

RESUMEN

BACKGROUND: Magnetic resonance imaging (MRI) can visualize locations of both the ablation scar on the left atrium (LA) after atrial fibrillation (AF) ablation and epicardial fat pads (FPs) containing ganglionated plexi (GP). METHODS: We investigated 60 patients who underwent pulmonary vein antrum (PVA) isolation along with LA posterior wall and septal debulking for AF. FPs around the LA surface in well-known GP areas (which were considered as the substitution of GP areas around the LA) were segmented from the dark-blood MRI. Then the FP and the ablation scar image visualized by late gadolinium enhancement (LGE)-MRI on the LA were merged together. Overlapping areas of FP and the ablation scar image were considered as the ablated FP areas containing GP. Patients underwent 24-hour Holter monitoring after ablation for the analysis of heart rate variability. RESULTS: Ablated FP area was significantly wider in patients without AF recurrence than those in patients with recurrence (5.6 ± 3.1 cm(2) vs 4.2 ± 2.7 cm(2) , P = 0.03). The mean values of both percentage of differences greater than 50 ms in the RR intervals (pRR > 50) and standard deviation of RR intervals over the entire analyzed period (SDNN), which were obtained from 24-hour Holter monitoring 1-day post-AF ablation, were significantly lower in patients without recurrence than those in patients with recurrence (5.8 ± 6.0% vs 14.0 ± 10.1%; P = 0.0005, 78.7 ± 32.4 ms vs 109.2 ± 43.5 ms; P = 0.005). There was a significant negative correlation between SDNN and the percentage of ablated FP area (Y = -1.3168X + 118.96, R(2) = 0.1576, P = 0.003). CONCLUSION: Extensively ablating LA covering GP areas along with PVA isolation enhanced the denervation of autonomic nerve system and seemed to improve procedural outcome in patients with AF.


Asunto(s)
Tejido Adiposo/cirugía , Fibrilación Atrial/cirugía , Ablación por Catéter/métodos , Imagen por Resonancia Magnética/métodos , Anciano , Distribución de Chi-Cuadrado , Medios de Contraste , Electrocardiografía Ambulatoria , Femenino , Humanos , Masculino , Meglumina/análogos & derivados , Persona de Mediana Edad , Compuestos Organometálicos , Pericardio/cirugía , Venas Pulmonares/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA