Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Comput Biol ; 19(12): e1011708, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38109436

ABSTRACT

The sinoatrial node (SAN), the primary pacemaker of the heart, is responsible for the initiation and robust regulation of sinus rhythm. 3D mapping studies of the ex-vivo human heart suggested that the robust regulation of sinus rhythm relies on specialized fibrotically-insulated pacemaker compartments (head, center and tail) with heterogeneous expressions of key ion channels and receptors. They also revealed up to five sinoatrial conduction pathways (SACPs), which electrically connect the SAN with neighboring right atrium (RA). To elucidate the role of these structural-molecular factors in the functional robustness of human SAN, we developed comprehensive biophysical computer models of the SAN based on 3D structural, functional and molecular mapping of ex-vivo human hearts. Our key finding is that the electrical insulation of the SAN except SACPs, the heterogeneous expression of If, INa currents and adenosine A1 receptors (A1R) across SAN pacemaker-conduction compartments are required to experimentally reproduce observed SAN activation patterns and important phenomena such as shifts of the leading pacemaker and preferential SACP. In particular, we found that the insulating border between the SAN and RA, is required for robust SAN function and protection from SAN arrest during adenosine challenge. The heterogeneity in the expression of A1R within the human SAN compartments underlies the direction of pacemaker shift and preferential SACPs in the presence of adenosine. Alterations of INa current and fibrotic remodelling in SACPs can significantly modulate SAN conduction and shift the preferential SACP/exit from SAN. Finally, we show that disease-induced fibrotic remodeling, INa suppression or increased adenosine make the human SAN vulnerable to pacing-induced exit blocks and reentrant arrhythmia. In summary, our computer model recapitulates the structural and functional features of the human SAN and can be a valuable tool for investigating mechanisms of SAN automaticity and conduction as well as SAN arrhythmia mechanisms under different pathophysiological conditions.


Subject(s)
Heart Conduction System , Sinoatrial Node , Humans , Sinoatrial Node/physiology , Arrhythmias, Cardiac , Adenosine , Computer Simulation
2.
J Mol Cell Cardiol ; 151: 56-71, 2021 02.
Article in English | MEDLINE | ID: mdl-33130148

ABSTRACT

Atrial fibrillation (AF) occurrence and maintenance is associated with progressive remodeling of electrophysiological (repolarization and conduction) and 3D structural (fibrosis, fiber orientations, and wall thickness) features of the human atria. Significant diversity in AF etiology leads to heterogeneous arrhythmogenic electrophysiological and structural substrates within the 3D structure of the human atria. Since current clinical methods have yet to fully resolve the patient-specific arrhythmogenic substrates, mechanism-based AF treatments remain underdeveloped. Here, we review current knowledge from in-vivo, ex-vivo, and in-vitro human heart studies, and discuss how these studies may provide new insights on the synergy of atrial electrophysiological and 3D structural features in AF maintenance. In-vitro studies on surgically acquired human atrial samples provide a great opportunity to study a wide spectrum of AF pathology, including functional changes in single-cell action potentials, ion channels, and gene/protein expression. However, limited size of the samples prevents evaluation of heterogeneous AF substrates and reentrant mechanisms. In contrast, coronary-perfused ex-vivo human hearts can be studied with state-of-the-art functional and structural technologies, such as high-resolution near-infrared optical mapping and contrast-enhanced MRI. These imaging modalities can resolve atrial arrhythmogenic substrates and their role in reentrant mechanisms maintaining AF and validate clinical approaches. Nonetheless, longitudinal studies are not feasible in explanted human hearts. As no approach is perfect, we suggest that combining the strengths of direct human atrial studies with high fidelity approaches available in the laboratory and in realistic patient-specific computer models would elucidate deeper knowledge of AF mechanisms. We propose that a comprehensive translational pipeline from ex-vivo human heart studies to longitudinal clinically relevant AF animal studies and finally to clinical trials is necessary to identify patient-specific arrhythmogenic substrates and develop novel AF treatments.


Subject(s)
Atrial Fibrillation/physiopathology , Electrophysiological Phenomena , Heart Atria/pathology , Heart Atria/physiopathology , Imaging, Three-Dimensional , Myocardium/pathology , Artificial Intelligence , Humans
3.
Am J Physiol Heart Circ Physiol ; 321(2): H412-H421, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34213393

ABSTRACT

Detailed global maps of atrial electrical activity are needed to understand mechanisms of atrial rhythm disturbance in small animal models of heart disease. To date, optical mapping systems have not provided enough spatial resolution across sufficiently extensive regions of intact atrial preparations to achieve this goal. The aim of this study was to develop an integrated platform for quantifying regional electrical properties and analyzing reentrant arrhythmia in a biatrial preparation. Intact atria from 6/7-mo-old female spontaneously hypertensive rats (SHRs; n = 6) were isolated and secured in a constant flow superfusion chamber at 37°C. Optical mapping was performed with the membrane-voltage dye di-4-ANEPPS using LED excitation and a scientific complementary metal-oxide semiconductor (sCMOS) camera. Programmed stimulus trains were applied from right atrial (RA) and left atrial (LA) sites to assess rate-dependent electrical behavior and to induce atrial arrhythmia. Signal-to-noise ratio was improved by sequential processing steps that included spatial smoothing, temporal filtering, and, in stable rhythms, ensemble-averaging. Activation time, repolarization time, and action potential duration (APD) maps were constructed at high spatial resolution for a wide range of coupling intervals. These data were highly consistent within and between experiments. They confirmed preferential atrial conduction pathways and demonstrated distinct medial-to-lateral APD gradients. We also showed that reentrant arrhythmias induced in this preparation were explained by the spatial variation of these electrical properties. Our new methodology provides a robust means of 1) quantifying regional electrical properties in the intact rat atria at higher spatiotemporal resolution than previously reported, and 2) characterizing reentrant arrhythmia and analyzing mechanisms that give rise to it.NEW & NOTEWORTHY Despite wide-ranging optical mapping studies, detailed information on regional atrial electrical properties in small animal models of heart disease and how these contribute to reentrant arrhythmia remains limited. We have developed a novel experimental platform that enables both to be achieved in a geometrically intact isolated rat bi-atrial preparation.


Subject(s)
Arrhythmias, Cardiac/diagnostic imaging , Heart Atria/diagnostic imaging , Voltage-Sensitive Dye Imaging/methods , Animals , Arrhythmias, Cardiac/physiopathology , Heart Atria/physiopathology , Rats , Rats, Inbred SHR
4.
PLoS Comput Biol ; 16(2): e1007678, 2020 02.
Article in English | MEDLINE | ID: mdl-32097431

ABSTRACT

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and is a major cause of stroke and morbidity. Recent genome-wide association studies have shown that paired-like homeodomain transcription factor 2 (Pitx2) to be strongly associated with AF. However, the mechanisms underlying Pitx2 modulated arrhythmogenesis and variable effectiveness of antiarrhythmic drugs (AADs) in patients in the presence or absence of impaired Pitx2 expression remain unclear. We have developed multi-scale computer models, ranging from a single cell to tissue level, to mimic control and Pitx2-knockout atria by incorporating recent experimental data on Pitx2-induced electrical and structural remodeling in humans, as well as the effects of AADs. The key findings of this study are twofold. We have demonstrated that shortened action potential duration, slow conduction and triggered activity occur due to electrical and structural remodelling under Pitx2 deficiency conditions. Notably, the elevated function of calcium transport ATPase increases sarcoplasmic reticulum Ca2+ concentration, thereby enhancing susceptibility to triggered activity. Furthermore, heterogeneity is further elevated due to Pitx2 deficiency: 1) Electrical heterogeneity between left and right atria increases; and 2) Increased fibrosis and decreased cell-cell coupling due to structural remodelling slow electrical propagation and provide obstacles to attract re-entry, facilitating the initiation of re-entrant circuits. Secondly, our study suggests that flecainide has antiarrhythmic effects on AF due to impaired Pitx2 by preventing spontaneous calcium release and increasing wavelength. Furthermore, our study suggests that Na+ channel effects alone are insufficient to explain the efficacy of flecainide. Our study may provide the mechanisms underlying Pitx2-induced AF and possible explanation behind the AAD effects of flecainide in patients with Pitx2 deficiency.


Subject(s)
Atrial Fibrillation/metabolism , Computer Simulation , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Action Potentials , Animals , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/genetics , Atrial Remodeling , Calcium/metabolism , Electrophysiology , Endoplasmic Reticulum/metabolism , Fibrosis , Flecainide/pharmacology , Gene Expression Regulation , Genome-Wide Association Study , Heart Atria/physiopathology , Homeodomain Proteins/genetics , Humans , Kinetics , Mice , Mice, Knockout , Phenotype , Ryanodine Receptor Calcium Release Channel/pharmacology , Sarcoplasmic Reticulum/metabolism , Sodium/metabolism , Transcription Factors/genetics , Homeobox Protein PITX2
5.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33514068

ABSTRACT

Electrical remodelling as a result of homeodomain transcription factor 2 (Pitx2)-dependent gene regulation was linked to atrial fibrillation (AF) and AF patients with single nucleotide polymorphisms at chromosome 4q25 responded favorably to class I antiarrhythmic drugs (AADs). The possible reasons behind this remain elusive. The purpose of this study was to assess the efficacy of the AADs disopyramide, quinidine, and propafenone on human atrial arrhythmias mediated by Pitx2-induced remodelling, from a single cell to the tissue level, using drug binding models with multi-channel pharmacology. Experimentally calibrated populations of human atrial action po-tential (AP) models in both sinus rhythm (SR) and Pitx2-induced AF conditions were constructed by using two distinct models to represent morphological subtypes of AP. Multi-channel pharmaco-logical effects of disopyramide, quinidine, and propafenone on ionic currents were considered. Simulated results showed that Pitx2-induced remodelling increased maximum upstroke velocity (dVdtmax), and decreased AP duration (APD), conduction velocity (CV), and wavelength (WL). At the concentrations tested in this study, these AADs decreased dVdtmax and CV and prolonged APD in the setting of Pitx2-induced AF. Our findings of alterations in WL indicated that disopyramide may be more effective against Pitx2-induced AF than propafenone and quinidine by prolonging WL.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Atrial Fibrillation/drug therapy , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Computer Simulation , Disopyramide/chemistry , Disopyramide/pharmacology , Heart Atria/drug effects , Heart Atria/pathology , Humans , Mice , Propafenone/chemistry , Propafenone/therapeutic use , Quinidine/chemistry , Quinidine/pharmacology , Homeobox Protein PITX2
6.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34299303

ABSTRACT

Atrial fibrillation (AF) is a common arrhythmia. Better prevention and treatment of AF are needed to reduce AF-associated morbidity and mortality. Several major mechanisms cause AF in patients, including genetic predispositions to AF development. Genome-wide association studies have identified a number of genetic variants in association with AF populations, with the strongest hits clustering on chromosome 4q25, close to the gene for the homeobox transcription PITX2. Because of the inherent complexity of the human heart, experimental and basic research is insufficient for understanding the functional impacts of PITX2 variants on AF. Linking PITX2 properties to ion channels, cells, tissues, atriums and the whole heart, computational models provide a supplementary tool for achieving a quantitative understanding of the functional role of PITX2 in remodelling atrial structure and function to predispose to AF. It is hoped that computational approaches incorporating all we know about PITX2-related structural and electrical remodelling would provide better understanding into its proarrhythmic effects leading to development of improved anti-AF therapies. In the present review, we discuss advances in atrial modelling and focus on the mechanistic links between PITX2 and AF. Challenges in applying models for improving patient health are described, as well as a summary of future perspectives.


Subject(s)
Atrial Fibrillation/etiology , Atrial Fibrillation/genetics , Homeodomain Proteins/genetics , Models, Cardiovascular , Transcription Factors/genetics , Animals , Atrial Fibrillation/physiopathology , Atrial Remodeling/genetics , Atrial Remodeling/physiology , Body Patterning/genetics , Computer Simulation , Genes, Homeobox , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Heart/embryology , Homeodomain Proteins/physiology , Humans , Ion Channels/genetics , Ion Channels/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Transcription Factors/physiology , Homeobox Protein PITX2
7.
Philos Trans A Math Phys Eng Sci ; 378(2173): 20190557, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32448059

ABSTRACT

Delayed afterdepolarizations (DADs) and spontaneous depolarizations (SDs) are typically triggered by spontaneous diastolic Ca2+ release from the sarcoplasmic reticulum (SR) which is caused by an elevated SR Ca2+-ATPase (SERCA) uptake and dysfunctional ryanodine receptors. However, recent studies on the T-box transcription factor gene (TBX5) demonstrated that abnormal depolarizations could occur despite a reduced SERCA uptake. Similar findings have also been reported in experimental or clinical studies of diabetes and heart failure. To investigate the sensitivity of SERCA in the genesis of DADs/SDs as well as its dependence on other Ca2+ handling channels, we performed systematic analyses using the Maleckar et al. model. Results showed that the modulation of SERCA alone cannot trigger abnormal depolarizations, but can instead affect the interdependency of other Ca2+ handling channels in triggering DADs/SDs. Furthermore, we discovered the existence of a threshold value for the intracellular concentration of Ca2+ ([Ca2+]i) for abnormal depolarizations, which is modulated by the maximum SERCA uptake and the concentration of Ca2+ in the uptake and release compartments in the SR ([Ca2+]up and [Ca2+]rel). For the first time, our modelling study reconciles different mechanisms of abnormal depolarizations in the setting of 'lone' AF, reduced TBX5, diabetes and heart failure, and may lead to more targeted treatment for these patients. This article is part of the theme issue 'Uncertainty quantification in cardiac and cardiovascular modelling and simulation'.


Subject(s)
Action Potentials , Calcium/metabolism , Heart Atria/cytology , Models, Cardiovascular , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Cohort Studies , Humans , Protein Transport
8.
Cardiovasc Diabetol ; 18(1): 165, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31779619

ABSTRACT

BACKGROUND: Diabetes mellitus is an important risk factor for atrial fibrillation (AF) development. Sodium-glucose co-transporter-2 (SGLT-2) inhibitors are used for the treatment of type 2 diabetes mellitus (T2DM). Their cardioprotective effects have been reported but whether they prevent AF in T2DM patients are less well-explored. We tested the hypothesis that the SGLT-2 inhibitor, empagliflozin, can prevent atrial remodeling in a diabetic rat model. METHODS: High-fat diet and low-dose streptozotocin (STZ) treatment were used to induce T2DM. A total of 96 rats were randomized into the following four groups: (i) control (ii) T2DM, (iii) low-dose empagliflozin (10 mg/kg/day)/T2DM; and (iv) high-dose empagliflozin (30 mg/kg/day)/T2DM by the intragastric route for 8 weeks. RESULTS: Compared with the control group, left atrial diameter, interstitial fibrosis and the incidence of AF inducibility were significantly increased in the DM group. Moreover, atrial mitochondrial respiratory function, mitochondrial membrane potential, and mitochondrial biogenesis were impaired. Empagliflozin treatment significantly prevented the development of these abnormalities in DM rats, likely via the peroxisome proliferator-activated receptor-c coactivator 1α (PGC-1α)/nuclear respiratory factor-1 (NRF-1)/mitochondrial transcription factor A (Tfam) signaling pathway. CONCLUSIONS: Empagliflozin can ameliorate atrial structural and electrical remodeling as well as improve mitochondrial function and mitochondrial biogenesis in T2DM, hence may be potentially used in the prevention of T2DM-related atrial fibrillation.


Subject(s)
Atrial Fibrillation/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Heart Rate/drug effects , Mitochondria, Heart/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Disease Models, Animal , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Organelle Biogenesis , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction , Streptozocin
9.
Circulation ; 134(6): 486-98, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27462069

ABSTRACT

BACKGROUND: Adenosine provokes atrial fibrillation (AF) with a higher activation frequency in right atria (RA) versus left atria (LA) in patients, but the underlying molecular and functional substrates are unclear. We tested the hypothesis that adenosine-induced AF is driven by localized reentry in RA areas with highest expression of adenosine A1 receptor and its downstream GIRK (G protein-coupled inwardly rectifying potassium channels) channels (IK,Ado). METHODS: We applied biatrial optical mapping and immunoblot mapping of various atrial regions to reveal the mechanism of adenosine-induced AF in explanted failing and nonfailing human hearts (n=37). RESULTS: Optical mapping of coronary-perfused atria (n=24) revealed that adenosine perfusion (10-100 µmol/L) produced more significant shortening of action potential durations in RA (from 290±45 to 239±41 ms, 17.3±10.4%; P<0.01) than LA (from 307±24 to 286±23 ms, 6.7±6.6%; P<0.01). In 10 hearts, adenosine induced AF (317±116 s) that, when sustained (≥2 minutes), was primarily maintained by 1 to 2 localized reentrant drivers in lateral RA. Tertiapin (10-100 nmol/L), a selective GIRK channel blocker, counteracted adenosine-induced action potential duration shortening and prevented AF induction. Immunoblotting showed that the superior/middle lateral RA had significantly higher adenosine A1 receptor (2.7±1.7-fold; P<0.01) and GIRK4 (1.7±0.8-fold; P<0.05) protein expression than lateral/posterior LA. CONCLUSIONS: This study revealed a 3-fold RA-to-LA adenosine A1 receptor protein expression gradient in the human heart, leading to significantly greater RA versus LA repolarization sensitivity in response to adenosine. Sustained adenosine-induced AF is maintained by reentrant drivers localized in lateral RA regions with the highest adenosine A1 receptor/GIRK4 expression. Selective atrial GIRK channel blockade may effectively treat AF during conditions with increased endogenous adenosine.


Subject(s)
Adenosine/toxicity , Atrial Fibrillation/chemically induced , Atrial Fibrillation/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/biosynthesis , Heart Atria/metabolism , Receptor, Adenosine A1/biosynthesis , Adult , Aged , Female , Gene Expression Regulation , Heart/diagnostic imaging , Heart/drug effects , Heart Atria/diagnostic imaging , Heart Atria/drug effects , Heart Conduction System/diagnostic imaging , Heart Conduction System/drug effects , Heart Conduction System/metabolism , Humans , Male , Middle Aged , Organ Culture Techniques , Positron Emission Tomography Computed Tomography
10.
Echocardiography ; 33(10): 1546-1556, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783876

ABSTRACT

BACKGROUND: The anatomical substrate for the mid-mural ventricular hyperechogenic zone remains uncertain, but it may represent no more than ultrasound reflected from cardiomyocytes orientated orthogonally to the ultrasonic beam. We sought to ascertain the relationship between the echogenic zone and the orientation of the cardiomyocytes. METHODS: We used 3D echocardiography, diffusion tensor imaging, and microcomputed tomography to analyze the location and orientation of cardiomyocytes within the echogenic zone. RESULTS: We demonstrated that visualization of the echogenic zone is dependent on the position of the transducer and is most clearly seen from the apical window. Diffusion tensor imaging and microcomputed tomography show that the echogenic zone seen from the apical window corresponds to the position of the circumferentially orientated cardiomyocytes. An oblique band seen in the parasternal view relates to cardiomyocytes orientated orthogonally to the ultrasonic beam. CONCLUSIONS: The mid-mural ventricular hyperechogenic zone represents reflected ultrasound from cardiomyocytes aligned orthogonal to the ultrasonic beam. The echogenic zone does not represent a space, a connective tissue sheet, a boundary between ascending and descending limbs of a hypothetical helical ventricular myocardial band, nor an abrupt change in cardiomyocyte orientation.


Subject(s)
Echocardiography/methods , Heart Ventricles/cytology , Heart Ventricles/diagnostic imaging , Magnetic Resonance Imaging/methods , Myocytes, Cardiac/cytology , Tomography, X-Ray Computed/methods , Aged , Cardiac Imaging Techniques/methods , Female , Humans
11.
Eur Heart J ; 36(35): 2390-401, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26059724

ABSTRACT

AIMS: The complex architecture of the human atria may create physical substrates for sustained re-entry to drive atrial fibrillation (AF). The existence of sustained, anatomically defined AF drivers in humans has been challenged partly due to the lack of simultaneous endocardial-epicardial (Endo-Epi) mapping coupled with high-resolution 3D structural imaging. METHODS AND RESULTS: Coronary-perfused human right atria from explanted diseased hearts (n = 8, 43-72 years old) were optically mapped simultaneously by three high-resolution CMOS cameras (two aligned Endo-Epi views (330 µm2 resolution) and one panoramic view). 3D gadolinium-enhanced magnetic resonance imaging (GE-MRI, 80 µm3 resolution) revealed the atrial wall structure varied in thickness (1.0 ± 0.7-6.8 ± 2.4 mm), transmural fiber angle differences, and interstitial fibrosis causing transmural activation delay from 23 ± 11 to 43 ± 22 ms at increased pacing rates. Sustained AF (>90 min) was induced by burst pacing during pinacidil (30-100 µM) perfusion. Dual-sided sub-Endo-sub-Epi optical mapping revealed that AF was driven by spatially and temporally stable intramural re-entry with 107 ± 50 ms cycle length and transmural activation delay of 67 ± 31 ms. Intramural re-entrant drivers were captured primarily by sub-Endo mapping, while sub-Epi mapping visualized re-entry or 'breakthrough' patterns. Re-entrant drivers were anchored on 3D micro-anatomic tracks (15.4 ± 2.2 × 6.0 ± 2.3 mm2, 2.9 ± 0.9 mm depth) formed by atrial musculature characterized by increased transmural fiber angle differences and interstitial fibrosis. Targeted radiofrequency ablation of the tracks verified these re-entries as drivers of AF. CONCLUSIONS: Integrated 3D structural-functional mapping of diseased human right atria ex vivo revealed that the complex atrial microstructure caused significant differences between Endo vs. Epi activation during pacing and sustained AF driven by intramural re-entry anchored to fibrosis-insulated atrial bundles.


Subject(s)
Atrial Fibrillation/pathology , Heart Atria/pathology , Adult , Aged , Atrial Fibrillation/etiology , Atrial Fibrillation/physiopathology , Cardiac Imaging Techniques , Contrast Media , Epicardial Mapping/methods , Gadolinium , Heart Atria/physiopathology , Humans , Magnetic Resonance Angiography/methods , Middle Aged
12.
Clin Anat ; 29(3): 316-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26478993

ABSTRACT

How the cardiomyocytes are aggregated within the heart walls remains contentious. We still do not fully understand how the end-to-end longitudinal myocytic chains are arranged, nor the true extent and shape of the lamellar units they aggregate to form. In this article, we show that an understanding of the complex arrangement of cardiac musculature requires knowledge of three-dimensional myocyte orientation (helical and intrusion angle), and appreciation of myocyte packing within the connective tissue matrix. We show how visualization and segmentation of high-resolution three-dimensional image data can accurately identify the morphology and orientation of the myocytic chains, and the lamellar units. Some maintain that the ventricles can be unwrapped in the form of a "helical ventricular myocardial band," that is, as a compartmentalized band with selective regional innervation and deformation, and a defined origin and insertion like most skeletal muscles. In contrast to the simpler interpretation of the helical ventricular myocardial band, we provide insight as to how the complex myocytic chains, the heterogeneous lamellar units, and connective tissue matrix form an interconnected meshwork, which facilitates the complex internal deformations of the ventricular wall. We highlight the dangers of disregarding the intruding cardiomyocytes. Preparation of the band destroys intruding myocytic chains, and thus disregards the functional implications of the antagonistic auxotonic forces they produce. We conclude that the ventricular myocardium is not analogous to skeletal muscle, but is a complex three-dimensional meshwork, with a heterogeneous branching lamellar architecture.


Subject(s)
Myocardium/cytology , Myocytes, Cardiac/cytology , Animals , Diffusion Tensor Imaging , Heart/anatomy & histology , Heart/diagnostic imaging , Heart Conduction System/anatomy & histology , Muscle, Skeletal/cytology
13.
Circ Res ; 112(5): 834-48, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23449546

ABSTRACT

Impulse propagation in the heart depends on the excitability of individual cardiomyocytes, impulse transmission between adjacent myocytes, and the 3-dimensional arrangement of those cells. Here, we review the role of each of these factors in normal and aberrant cardiac electric activation, with particular emphasis on the effects of 3-dimensional myocyte architecture at the tissue scale. The analysis draws on findings from in vivo and in vitro experiments, as well as biophysically based computer models that have been used to integrate and interpret these experimental data. It indicates that discontinuous arrangement of myocytes and extracellular connective tissue at the tissue scale can give rise to current source-to-sink mismatch, spatiotemporal distribution of refractoriness, and rate-sensitive electric instability, which contribute to the initiation and maintenance of reentrant cardiac arrhythmia. This exacerbates the risk of rhythm disturbance associated with heart disease. We conclude that structure-based, multiscale computer models that incorporate accurate information about local cellular electric activity provide a powerful platform for investigating the basis of reentrant cardiac arrhythmia. However, it is important that these models capture key features of structure and related electric function at the tissue scale.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Heart Conduction System/physiopathology , Heart/physiopathology , Arrhythmias, Cardiac/pathology , Cell Communication/physiology , Computer Simulation , Heart Conduction System/pathology , Humans , Models, Cardiovascular , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology
14.
Int J Mol Sci ; 16(5): 10834-54, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25984605

ABSTRACT

Atrial fibrillation (AF) is the most common heart rhythm disturbance, and its treatment is an increasing economic burden on the health care system. Despite recent intense clinical, experimental and basic research activity, the treatment of AF with current antiarrhythmic drugs and catheter/surgical therapies remains limited. Radiofrequency catheter ablation (RFCA) is widely used to treat patients with AF. Current clinical ablation strategies are largely based on atrial anatomy and/or substrate detected using different approaches, and they vary from one clinical center to another. The nature of clinical ablation leads to ambiguity regarding the optimal patient personalization of the therapy partly due to the fact that each empirical configuration of ablation lines made in a patient is irreversible during one ablation procedure. To investigate optimized ablation lesion line sets, in silico experimentation is an ideal solution. 3D computer models give us a unique advantage to plan and assess the effectiveness of different ablation strategies before and during RFCA. Reliability of in silico assessment is ensured by inclusion of accurate 3D atrial geometry, realistic fiber orientation, accurate fibrosis distribution and cellular kinetics; however, most of this detailed information in the current computer models is extrapolated from animal models and not from the human heart. The predictive power of computer models will increase as they are validated with human experimental and clinical data. To make the most from a computer model, one needs to develop 3D computer models based on the same functionally and structurally mapped intact human atria with high spatial resolution. The purpose of this review paper is to summarize recent developments in clinically-derived computer models and the clinical insights they provide for catheter ablation.


Subject(s)
Atrial Fibrillation/surgery , Catheter Ablation , Computer Simulation , Animals , Fibrosis , Humans , Models, Cardiovascular , Pulmonary Veins/pathology
15.
Article in English | MEDLINE | ID: mdl-38083390

ABSTRACT

Atrial fibrillation (AF) is the most common, sustained cardiac arrhythmia. Early intervention and treatment could have a much higher chance of reversing AF. An electrocardiogram (ECG) is widely used to check the heart's rhythm and electrical activity in clinics. The current manual processing of ECGs and clinical classification of AF types (paroxysmal, persistent and permanent AF) is ill-founded and does not truly reflect the seriousness of the disease. In this paper, we proposed a new machine learning method for beat-wise classification of ECGs to estimate AF burden, which was defined by the percentage of AF beats found in the total recording time. Both morphological and temporal features for categorizing AF were extracted via two combined classifiers: a 1D U-Net that evaluates fiducial points and segmentation to locate each heartbeat; and the other Recurrent Neural Network (RNN) to enhance the temporal classification of an individual heartbeat. The output of the classifiers had four target classes: Normal Sinus Rhythm (SN), AF, Noises (NO), and Others (OT). The approach was trained and validated on the Icentia11k dataset, with 1001 and 250 patients' ECGs, respectively. The testing accuracy for the four classes was found to be 0.86, 0.81, 0.79, and 0.75, respectively. Our study demonstrated the feasibility and superior performance of combing U-net and RNN to conduct a beat-wise classification of ECGs for AF burden. However, further investigation is warranted to validate this deep learning approach.Clinical relevance- This paper proposes a novel machine learning network for ECG beatwise classification, specifically for aiding AF burden determination.


Subject(s)
Atrial Fibrillation , Deep Learning , Humans , Atrial Fibrillation/diagnosis , Neural Networks, Computer , Heart Rate , Electrocardiography/methods
16.
Interface Focus ; 13(6): 20230041, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38106913

ABSTRACT

Fibrosis has been mechanistically linked to arrhythmogenesis in multiple cardiovascular conditions, including atrial fibrillation (AF). Previous studies have demonstrated that fibrosis can create functional barriers to conduction which may promote excitation wavebreak and the generation of re-entry, while also acting to pin re-entrant excitation in stable rotors during AF. However, few studies have investigated the role of fibrosis in the generation of AF triggers in detail. We apply our in-house computational framework to study the impact of fibrosis on the generation of AF triggers and trigger-substrate interactions in two- and three-dimensional atrial tissue models. Our models include a reduced and efficient description of stochastic, spontaneous cellular triggers as well as a simple model of heterogeneous inter-cellular coupling. Our results demonstrate that fibrosis promotes the emergence of focal excitations, primarily through reducing the electrotonic load on individual fibre strands. This enables excitation to robustly initiate within these single strands before spreading to neighbouring strands and inducing a full tissue focal excitation. Enhanced conduction block can allow trigger-substrate interactions that result in the emergence of complex, re-entrant excitation patterns. This study provides new insight into the mechanisms by which fibrosis promotes the triggers and substrate necessary to induce and sustain arrhythmia.

17.
Interface Focus ; 13(6): 20230039, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38106916

ABSTRACT

This study aimed to use multi-scale atrial models to investigate pulmonary arterial hypertension (PAH)-induced atrial fibrillation mechanisms. The results of our computer simulations revealed that, at the single-cell level, PAH-induced remodelling led to a prolonged action potential (AP) (ΔAPD: 49.6 ms in the right atria (RA) versus 41.6 ms in the left atria (LA)) and an increased calcium transient (CaT) (ΔCaT: 7.5 × 10-2 µM in the RA versus 0.9 × 10-3 µM in the LA). Moreover, heterogeneous remodelling increased susceptibility to afterdepolarizations, particularly in the RA. At the tissue level, we observed a significant reduction in conduction velocity (CV) (ΔCV: -0.5 m s-1 in the RA versus -0.05 m s-1 in the LA), leading to a shortened wavelength in the RA, but not in the LA. Additionally, afterdepolarizations in the RA contributed to enhanced repolarization dispersion and facilitated unidirectional conduction block. Furthermore, the increased fibrosis in the RA amplified the likelihood of excitation wave breakdown and the occurrence of sustained re-entries. Our results indicated that the RA is characterized by increased susceptibility to afterdepolarizations, slow conduction, reduced wavelength and upregulated fibrosis. These findings shed light on the underlying factors that may promote atrial fibrillation in patients with PAH.

18.
Interface Focus ; 13(6): 20230044, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38106912

ABSTRACT

Persistent atrial fibrillation (AF) is not effectively treated due to a lack of adequate tools for identifying patient-specific AF substrates. Recent studies revealed that in 30-50% of patients, persistent AF is maintained by localized drivers not only in the left atrium (LA) but also in the right atrium (RA). The chamber-specific atrial wall thickness (AWT) features underlying AF remain elusive, though the important role of AWT in AF is widely acknowledged. We aimed to provide direct evidence of the existence of distinguished RA and LA AWT features underlying AF drivers by analysing functionally and structurally mapped human hearts ex vivo. Coronary-perfused intact human atria (n = 7, 47 ± 14 y.o.; two female) were mapped using panoramic near-infrared optical mapping during pacing-induced AF. Then the hearts were imaged at approximately 170 µm3 resolution by 9.4 T gadolinium-enhanced MRI. The heart was segmented, and 3D AWT throughout atrial chambers was estimated and analysed. Optical mapping identified six localized RA re-entrant drivers in four hearts and four LA drivers in three hearts. All RA AF drivers were anchored to the pectinate muscle junctions with the crista terminalis or atrial walls. The four LA AF drivers were in the posterior LA. RA (n = 4) with AF drivers were thicker with greater AWT variation than RA (n = 3) without drivers (5.4 ± 2.6 mm versus 5.0 ± 2.4 mm, T-test p < 0.05; F-test p < 0.05). Furthermore, AWT in RA driver regions was thicker and varied more than in RA non-driver regions (5.1 ± 2.5 mm versus 4.4 ± 2.2 mm, T-test p < 0.05; F-test p < 0.05). On the other hand, LA (n = 3) with drivers was thinner than the LA (n = 4) without drivers. In particular, LA driver regions were thinner than the rest of LA regions (3.4 ± 1.0 mm versus 4.2 ± 1.0 mm, T-test p < 0.05). This study demonstrates chamber-specific AWT features of AF drivers. In RA, driver regions are thicker and have more variable AWT than non-driver regions. By contrast, LA drivers are thinner than non-drivers. Robust evaluation of patient-specific AWT features should be considered for chamber-specific targeted ablation.

19.
Front Physiol ; 13: 957604, 2022.
Article in English | MEDLINE | ID: mdl-36111152

ABSTRACT

Atrial fibrillation (AF) with multiple complications, high morbidity and mortality, and low cure rates, has become a global public health problem. Although significant progress has been made in the treatment methods represented by anti-AF drugs and radiofrequency ablation, the therapeutic effect is not as good as expected. The reason is mainly because of our lack of understanding of AF mechanisms. This field has benefited from mechanistic and (or) statistical methodologies. Recent renewed interest in digital twin techniques by synergizing between mechanistic and statistical models has opened new frontiers in AF analysis. In the review, we briefly present findings that gave rise to the AF pathophysiology and current therapeutic modalities. We then summarize the achievements of digital twin technologies in three aspects: understanding AF mechanisms, screening anti-AF drugs and optimizing ablation strategies. Finally, we discuss the challenges that hinder the clinical application of the digital twin heart. With the rapid progress in data reuse and sharing, we expect their application to realize the transition from AF description to response prediction.

20.
Comput Biol Med ; 146: 105551, 2022 07.
Article in English | MEDLINE | ID: mdl-35533458

ABSTRACT

Electrocardiograms (ECG) provide an effective, non-invasive approach for clinical diagnosis and monitoring treatment in patients with cardiac diseases including the most common cardiac arrhythmia, atrial fibrillation (AF). Portable ECG recording devices including Apple Watch and Kardia devices have been developed for AF detection. However, the efficacy of these smart devices has not been fully validated. We aimed to develop an open-source deep learning framework for automatic AF detection using the largest publicly available single-lead ECG dataset through a mobile Kardia device enhanced with style transfer-driven data augmentation. We developed and validated a 37-layer convolutional recurrent network (CRN) using 5,834 single-lead ECGs with a mean length of 30 seconds from the 2017 PhysioNet Challenge to automatically detect sinus rhythm and AF. To address the challenge of a lack of a large number of AF samples, we proposed a novel style transfer generator that fuses patient-specific clinical ECGs and mathematically modelled ECG features to synthesize realistic ECGs by five-fold. The differences between synthesized and clinical ECGs were analyzed by studying their average ECG morphologies and frequency distributions. Our results indicated the style transfer-driven data augmentation was not classifier-dependent. Validation on 2,917 clinical ECGs showed an F1 score of 96.4%, with the generated ECGs contributing to a 3% improvement in AF detection for the Kardia event recorder. By developing and evaluating our approach on an open-source ECG dataset, we have demonstrated that our framework is both robust and verifiable, and potentially can be used in portable devices for effective AF classification.


Subject(s)
Atrial Fibrillation , Wearable Electronic Devices , Atrial Fibrillation/diagnosis , Electrocardiography/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL