Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Transl Pediatr ; 13(1): 146-163, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38323181

RESUMO

Background and Objective: Computational models of the cardiovascular system allow for a detailed and quantitative investigation of both physiological and pathological conditions, thanks to their ability to combine clinical-possibly patient-specific-data with physical knowledge of the processes underlying the heart function. These models have been increasingly employed in clinical practice to understand pathological mechanisms and their progression, design medical devices, support clinicians in improving therapies. Hinging upon a long-year experience in cardiovascular modeling, we have recently constructed a computational multi-physics and multi-scale integrated model of the heart for the investigation of its physiological function, the analysis of pathological conditions, and to support clinicians in both diagnosis and treatment planning. This narrative review aims to systematically discuss the role that such model had in addressing specific clinical questions, and how further impact of computational models on clinical practice are envisaged. Methods: We developed computational models of the physical processes encompassed by the heart function (electrophysiology, electrical activation, force generation, mechanics, blood flow dynamics, valve dynamics, myocardial perfusion) and of their inherently strong coupling. To solve the equations of such models, we devised advanced numerical methods, implemented in a flexible and highly efficient software library. We also developed computational procedures for clinical data post-processing-like the reconstruction of the heart geometry and motion from diagnostic images-and for their integration into computational models. Key Content and Findings: Our integrated computational model of the heart function provides non-invasive measures of indicators characterizing the heart function and dysfunctions, and sheds light on its underlying processes and their coupling. Moreover, thanks to the close collaboration with several clinical partners, we addressed specific clinical questions on pathological conditions, such as arrhythmias, ventricular dyssynchrony, hypertrophic cardiomyopathy, degeneration of prosthetic valves, and the way coronavirus disease 2019 (COVID-19) infection may affect the cardiac function. In multiple cases, we were also able to provide quantitative indications for treatment. Conclusions: Computational models provide a quantitative and detailed tool to support clinicians in patient care, which can enhance the assessment of cardiac diseases, the prediction of the development of pathological conditions, and the planning of treatments and follow-up tests.

2.
Front Cardiovasc Med ; 10: 1233991, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37817867

RESUMO

Ventricular arrhythmias, particularly ventricular tachycardia, are ubiquitously linked to 300,000 deaths annually. However, the current interventional procedure-the cardiac ablation-predict only short-term responses to treatment as the heart constantly remodels itself post-arrhythmia. To assist in the design of computational methods which focuses on long-term arrhythmia prediction, this review postulates three interdependent prospectives. The main objective is to propose computational methods for predicting long-term heart response to interventions in ventricular tachycardia Following a general discussion on the importance of devising simulations predicting long-term heart response to interventions, each of the following is discussed: (i) application of "metabolic sink theory" to elucidate the "re-entry" mechanism of ventricular tachycardia; (ii) application of "growth laws" to explain "mechanical load" translation in ventricular tachycardia; (iii) derivation of partial differential equations (PDE) to establish a pipeline to predict long-term clinical outcomes in ventricular tachycardia.

3.
BMC Bioinformatics ; 24(1): 389, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37828428

RESUMO

BACKGROUND: Simulating the cardiac function requires the numerical solution of multi-physics and multi-scale mathematical models. This underscores the need for streamlined, accurate, and high-performance computational tools. Despite the dedicated endeavors of various research teams, comprehensive and user-friendly software programs for cardiac simulations, capable of accurately replicating both normal and pathological conditions, are still in the process of achieving full maturity within the scientific community. RESULTS: This work introduces [Formula: see text]-ep, a publicly available software for numerical simulations of the electrophysiology activity of the cardiac muscle, under both normal and pathological conditions. [Formula: see text]-ep employs the monodomain equation to model the heart's electrical activity. It incorporates both phenomenological and second-generation ionic models. These models are discretized using the Finite Element method on tetrahedral or hexahedral meshes. Additionally, [Formula: see text]-ep integrates the generation of myocardial fibers based on Laplace-Dirichlet Rule-Based Methods, previously released in Africa et al., 2023, within [Formula: see text]-fiber. As an alternative, users can also choose to import myofibers from a file. This paper provides a concise overview of the mathematical models and numerical methods underlying [Formula: see text]-ep, along with comprehensive implementation details and instructions for users. [Formula: see text]-ep features exceptional parallel speedup, scaling efficiently when using up to thousands of cores, and its implementation has been verified against an established benchmark problem for computational electrophysiology. We showcase the key features of [Formula: see text]-ep through various idealized and realistic simulations conducted in both normal and pathological scenarios. Furthermore, the software offers a user-friendly and flexible interface, simplifying the setup of simulations using self-documenting parameter files. CONCLUSIONS: [Formula: see text]-ep provides easy access to cardiac electrophysiology simulations for a wide user community. It offers a computational tool that integrates models and accurate methods for simulating cardiac electrophysiology within a high-performance framework, while maintaining a user-friendly interface. [Formula: see text]-ep represents a valuable tool for conducting in silico patient-specific simulations.


Assuntos
Técnicas Eletrofisiológicas Cardíacas , Software , Humanos , Simulação por Computador , Miocárdio , África
4.
Prog Biomed Eng (Bristol) ; 5(3): 032004, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37360227

RESUMO

Computational models of the heart are now being used to assess the effectiveness and feasibility of interventions through in-silico clinical trials (ISCTs). As the adoption and acceptance of ISCTs increases, best practices for reporting the methodology and analysing the results will emerge. Focusing in the area of cardiology, we aim to evaluate the types of ISCTs, their analysis methods and their reporting standards. To this end, we conducted a systematic review of cardiac ISCTs over the period of 1 January 2012-1 January 2022, following the preferred reporting items for systematic reviews and meta-analysis (PRISMA). We considered cardiac ISCTs of human patient cohorts, and excluded studies of single individuals and those in which models were used to guide a procedure without comparing against a control group. We identified 36 publications that described cardiac ISCTs, with most of the studies coming from the US and the UK. In 75% of the studies, a validation step was performed, although the specific type of validation varied between the studies. ANSYS FLUENT was the most commonly used software in 19% of ISCTs. The specific software used was not reported in 14% of the studies. Unlike clinical trials, we found a lack of consistent reporting of patient demographics, with 28% of the studies not reporting them. Uncertainty quantification was limited, with sensitivity analysis performed in only 19% of the studies. In 97% of the ISCTs, no link was provided to provide easy access to the data or models used in the study. There was no consistent naming of study types with a wide range of studies that could potentially be considered ISCTs. There is a clear need for community agreement on minimal reporting standards on patient demographics, accepted standards for ISCT cohort quality control, uncertainty quantification, and increased model and data sharing.

5.
BMC Bioinformatics ; 24(1): 143, 2023 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-37046208

RESUMO

BACKGROUND: Modeling the whole cardiac function involves the solution of several complex multi-physics and multi-scale models that are highly computationally demanding, which call for simpler yet accurate, high-performance computational tools. Despite the efforts made by several research groups, no software for whole-heart fully-coupled cardiac simulations in the scientific community has reached full maturity yet. RESULTS: In this work we present [Formula: see text]-fiber, an innovative tool for the generation of myocardial fibers based on Laplace-Dirichlet Rule-Based Methods, which are the essential building blocks for modeling the electrophysiological, mechanical and electromechanical cardiac function, from single-chamber to whole-heart simulations. [Formula: see text]-fiber is the first publicly released module for cardiac simulations based on [Formula: see text], an open-source, high-performance Finite Element solver for multi-physics, multi-scale and multi-domain problems developed in the framework of the iHEART project, which aims at making in silico experiments easily reproducible and accessible to a wide community of users, including those with a background in medicine or bio-engineering. CONCLUSIONS: The tool presented in this document is intended to provide the scientific community with a computational tool that incorporates general state of the art models and solvers for simulating the cardiac function within a high-performance framework that exposes a user- and developer-friendly interface. This report comes with an extensive technical and mathematical documentation to welcome new users to the core structure of [Formula: see text]-fiber and to provide them with a possible approach to include the generated cardiac fibers into more sophisticated computational pipelines. In the near future, more modules will be successively published either as pre-compiled binaries for x86-64 Linux systems or as open source software.


Assuntos
Medicina , Software , Miócitos Cardíacos , Simulação por Computador
6.
Int J Numer Method Biomed Eng ; 39(2): e3670, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36510350

RESUMO

We present a simple, yet efficient adaptive time stepping scheme for cardiac electrophysiology (EP) simulations based on standard operator splitting techniques. The general idea is to exploit the relation between the splitting error and the reaction's magnitude-found in a previous one-dimensional analytical study by Spiteri and Ziaratgahi-to construct the new time step controller for three-dimensional problems. Accordingly, we propose to control the time step length of the operator splitting scheme as a function of the reaction magnitude, in addition to the common approach of adapting the reaction time step. This conforms with observations in numerical experiments supporting the need for a significantly smaller time step length during depolarization than during repolarization. The proposed scheme is compared with classical proportional-integral-differential controllers using state-of-the-art error estimators, which are also presented in details as they have not been previously applied in the context of cardiac EP with operator splitting techniques. Benchmarks show that choosing the time step as a sigmoidal function of the reaction magnitude is highly efficient and full cardiac cycles can be computed with precision even in a realistic biventricular setup. The proposed scheme outperforms common adaptive time stepping techniques, while depending on fewer tuning parameters.


Assuntos
Eletrofisiologia Cardíaca , Técnicas Eletrofisiológicas Cardíacas , Coração/fisiologia
7.
Ann Biomed Eng ; 51(2): 343-351, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35900706

RESUMO

Cardiac resynchronization therapy (CRT) is an effective treatment for a subgroup of heart failure (HF) patients, but more than 30% of those selected do not improve after CRT implantation. Imperfect pre-procedural criteria for patient selection and optimization are the main causes of the high non-response rate. In this study, we evaluated a novel measure for assessing CRT response. We used a computational modeling framework to calculate the regional stress of the left ventricular wall of seven CRT patients and seven healthy controls. The standard deviation of regional wall stress at the time of mitral valve closure (SD_MVC) was used to quantify dyssynchrony and compared between patients and controls and among the patients. The results show that SD_MVC is significantly lower in controls than patients and correlates with long-term response in patients, based on end-diastolic volume reduction. In contrast to our initial hypothesis, patients with lower SD_MVC respond better to therapy. The patient with the highest SD_MVC was the only non-responder in the patient cohort. The distribution of fiber stress at the beginning of the isovolumetric phase seems to correlate with the degree of response and the use of this measurement could potentially improve selection criteria for CRT implantation. Further studies with a larger cohort of patients are needed to validate these results.


Assuntos
Terapia de Ressincronização Cardíaca , Insuficiência Cardíaca , Humanos , Terapia de Ressincronização Cardíaca/efeitos adversos , Terapia de Ressincronização Cardíaca/métodos , Insuficiência Cardíaca/terapia , Ventrículos do Coração , Resultado do Tratamento
9.
Front Physiol ; 13: 847118, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35197869
10.
Front Cardiovasc Med ; 9: 1074562, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36733827

RESUMO

Atrial fibrillation (AF) underlies almost one third of all ischaemic strokes, with the left atrial appendage (LAA) identified as the primary thromboembolic source. Current stroke risk stratification approaches, such as the CHA2DS2-VASc score, rely mostly on clinical comorbidities, rather than thrombogenic mechanisms such as blood stasis, hypercoagulability and endothelial dysfunction-known as Virchow's triad. While detection of AF-related thrombi is possible using established cardiac imaging techniques, such as transoesophageal echocardiography, there is a growing need to reliably assess AF-patient thrombogenicity prior to thrombus formation. Over the past decade, cardiac imaging and image-based biophysical modelling have emerged as powerful tools for reproducing the mechanisms of thrombogenesis. Clinical imaging modalities such as cardiac computed tomography, magnetic resonance and echocardiographic techniques can measure blood flow velocities and identify LA fibrosis (an indicator of endothelial dysfunction), but imaging remains limited in its ability to assess blood coagulation dynamics. In-silico cardiac modelling tools-such as computational fluid dynamics for blood flow, reaction-diffusion-convection equations to mimic the coagulation cascade, and surrogate flow metrics associated with endothelial damage-have grown in prevalence and advanced mechanistic understanding of thrombogenesis. However, neither technique alone can fully elucidate thrombogenicity in AF. In future, combining cardiac imaging with in-silico modelling and integrating machine learning approaches for rapid results directly from imaging data will require development under a rigorous framework of verification and clinical validation, but may pave the way towards enhanced personalised stroke risk stratification in the growing population of AF patients. This Review will focus on the significant progress in these fields.

11.
Front Physiol ; 12: 745349, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34819872

RESUMO

Background: Remodeling due to myocardial infarction (MI) significantly increases patient arrhythmic risk. Simulations using patient-specific models have shown promise in predicting personalized risk for arrhythmia. However, these are computationally- and time- intensive, hindering translation to clinical practice. Classical machine learning (ML) algorithms (such as K-nearest neighbors, Gaussian support vector machines, and decision trees) as well as neural network techniques, shown to increase prediction accuracy, can be used to predict occurrence of arrhythmia as predicted by simulations based solely on infarct and ventricular geometry. We present an initial combined image-based patient-specific in silico and machine learning methodology to assess risk for dangerous arrhythmia in post-infarct patients. Furthermore, we aim to demonstrate that simulation-supported data augmentation improves prediction models, combining patient data, computational simulation, and advanced statistical modeling, improving overall accuracy for arrhythmia risk assessment. Methods: MRI-based computational models were constructed from 30 patients 5 days post-MI (the "baseline" population). In order to assess the utility biophysical model-supported data augmentation for improving arrhythmia prediction, we augmented the virtual baseline patient population. Each patient ventricular and ischemic geometry in the baseline population was used to create a subfamily of geometric models, resulting in an expanded set of patient models (the "augmented" population). Arrhythmia induction was attempted via programmed stimulation at 17 sites for each virtual patient corresponding to AHA LV segments and simulation outcome, "arrhythmia," or "no-arrhythmia," were used as ground truth for subsequent statistical prediction (machine learning, ML) models. For each patient geometric model, we measured and used choice data features: the myocardial volume and ischemic volume, as well as the segment-specific myocardial volume and ischemia percentage, as input to ML algorithms. For classical ML techniques (ML), we trained k-nearest neighbors, support vector machine, logistic regression, xgboost, and decision tree models to predict the simulation outcome from these geometric features alone. To explore neural network ML techniques, we trained both a three - and a four-hidden layer multilayer perceptron feed forward neural networks (NN), again predicting simulation outcomes from these geometric features alone. ML and NN models were trained on 70% of randomly selected segments and the remaining 30% was used for validation for both baseline and augmented populations. Results: Stimulation in the baseline population (30 patient models) resulted in reentry in 21.8% of sites tested; in the augmented population (129 total patient models) reentry occurred in 13.0% of sites tested. ML and NN models ranged in mean accuracy from 0.83 to 0.86 for the baseline population, improving to 0.88 to 0.89 in all cases. Conclusion: Machine learning techniques, combined with patient-specific, image-based computational simulations, can provide key clinical insights with high accuracy rapidly and efficiently. In the case of sparse or missing patient data, simulation-supported data augmentation can be employed to further improve predictive results for patient benefit. This work paves the way for using data-driven simulations for prediction of dangerous arrhythmia in MI patients.

12.
Front Med (Lausanne) ; 8: 700824, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34395480

RESUMO

Background: Moderate therapeutic hypothermia (TH) is a well-recognized cardio-protective strategy. The instillation of fluid into the peritoneum provides an opportunity to deliver moderate hypothermia as primary prevention against cardiovascular events. We aimed to to investigate both cardiac perfusion consequences (overall blood flow and detailed assessment of perfusion heterogeneity) and subsequently simulate the associated arrhythmic risk for patients undergoing peritoneal dialysis (PD) induced TH. Methods: Patients underwent high resolution myocardial perfusion scanning using high resolution 256 slice CT scanning, at rest and with adenosine stress. The first visit using the patient's usual PD regimen, on the second visit the same regime was utilized but with cooled peritoneal dialysate at 32°C. Myocardial blood flow (MBF) was quantified from generated perfusion maps, reconstructed in 3D. MBF heterogeneity was assessed by fractal dimension (FD) measurement on the 3D left ventricular reconstruction. Arrhythmogenicity was quantified from a sophisticated computational simulation using a multi-scale human 3D ventricle wedge electrophysiological computational model. Results: We studied 7 PD patients, mean age of 60 ± 7 and mean vintage dialysis of 23.6 ± 17.6 months. There were no significant different in overall segmental MBF between normothermic condition (NT) and TH. MBF heterogeneity was significantly decreased (-14%, p = 0.03) at rest and after stress (-14%, p = 0.03) when cooling was applied. Computational simulation showed that TH allowed a normalization of action potential, QT duration and T wave. Conclusion: TH-PD results in moderate hypothermia leading to a reduction in perfusion heterogeneity and simulated risk of non-terminating malignant ventricular arrhythmias.

13.
Comput Biol Med ; 135: 104549, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34171640

RESUMO

Accurate values for the six cardiac bidomain conductivities are crucial for meaningful computational studies of conduction in cardiac tissue, and are yet to be determined by experimental means. Although previous studies have proposed an approach using a multi-electrode array to measure potentials, from which the conductivities can be determined, it has been found that the conductivities cannot be retrieved consistently when the noise in the potentials varies. This paper presents a protocol, which not only has been shown to retrieve the conductivities to a reasonable accuracy, but does so under the presence of a more appropriate additive Gaussian noise model, while using fewer computational resources. Through repetitions of the protocol, a comparison of two pre-fabricated 128 electrode arrays, one array with a square arrangement of electrodes and the other with a rectangular arrangement, was made against a 75-electrode array proposed in previous studies. Results indicated that the two pre-fabricated arrays were generally more capable of obtaining the cardiac conductivities to a higher degree of accuracy than the 75-electrode array. The 128-electrode rectangular array was orientated such that the length of the array first ran along the direction of the fibres, then was reorientated such that the length of the array ran perpendicular to the direction of the fibres. The 128-electrode rectangular array, when orientated in this manner, was more capable of retrieving the conductivities than the remainder of the arrays tested, and thus we suggest this arrangement be used during experimental trials.


Assuntos
Coração , Modelos Cardiovasculares , Simulação por Computador , Condutividade Elétrica , Eletrodos
14.
Artigo em Inglês | MEDLINE | ID: mdl-32863678

RESUMO

Microscopic structural features of cardiac tissue play a fundamental role in determining complex spatio-temporal excitation dynamics at the macroscopic level. Recent efforts have been devoted to the development of mathematical models accounting for non-local spatio-temporal coupling able to capture these complex dynamics without the need of resolving tissue heterogeneities down to the micro-scale. In this work, we analyse in detail several important aspects affecting the overall predictive power of these modelling tools and provide some guidelines for an effective use of space-fractional models of cardiac electrophysiology in practical applications. Through an extensive computational study in simplified computational domains, we highlight the robustness of models belonging to different categories, i.e., physiological and phenomenological descriptions, against the introduction of non-locality, and lay down the foundations for future research and model validation against experimental data. A modern genetic algorithm framework is used to investigate proper parameterisations of the considered models, and the crucial role played by the boundary assumptions in the considered settings is discussed. Several numerical results are provided to support our claims.

15.
Stud Health Technol Inform ; 271: 215-223, 2020 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-32578566

RESUMO

BACKGROUND: Antitachycardial pacing (ATP) is a painless method for terminating ventricular tachycardias (VT) which would otherwise be treated using a painful high energy shock. However, it is well known that not each VT can be successfully terminated by ATP. Furthermore, ATP can be parametrized in several ways using scan, ramp or scan ramp approaches and can be applied in the right ventricle or in both ventricles (biventricular). In this work, we investigate the therapeutically most convenient ATP protocol based on a computer simulation using a patient individual model. METHODS: A patient individual model generated from a 3D/4D data set and a hybrid automaton was used for modeling and simulation of different VT scenarios. On the different VTs (from cycle length 288 ms up to 408 ms) different ATP approaches derived from the ADVANCE-CRT trial were applied in order to determine the effectiveness of these approaches. RESULTS: In this computer simulation study we were able to verify and validate the results from the ADVANCE-CRT trial. Biventricular ATP does not prove to be more effective than RV ATP but has a slight advantage in terminating fast VTs. CONCLUSIONS: The availability of a patient individual model and knowledge about the ischemic area and the underlying mechanism of the VTs will allow the use of these models to optimize ATP management.


Assuntos
Desfibriladores Implantáveis , Taquicardia Ventricular , Algoritmos , Estimulação Cardíaca Artificial , Simulação por Computador , Eletrocardiografia , Ventrículos do Coração , Humanos
17.
Front Physiol ; 9: 1513, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30425648

RESUMO

The field of computational cardiology has steadily progressed toward reliable and accurate simulations of the heart, showing great potential in clinical applications such as the optimization of cardiac interventions and the study of pro-arrhythmic effects of drugs in humans, among others. However, the computational effort demanded by in-silico studies of the heart remains challenging, highlighting the need of novel numerical methods that can improve the efficiency of simulations while targeting an acceptable accuracy. In this work, we propose a semi-implicit non-conforming finite-element scheme (SINCFES) suitable for cardiac electrophysiology simulations. The accuracy and efficiency of the proposed scheme are assessed by means of numerical simulations of the electrical excitation and propagation in regular and biventricular geometries. We show that the SINCFES allows for coarse-mesh simulations that reduce the computation time when compared to fine-mesh models while delivering wavefront shapes and conduction velocities that are more accurate than those predicted by traditional finite-element formulations based on the same coarse mesh, thus improving the accuracy-efficiency trade-off of cardiac simulations.

18.
J Mol Cell Cardiol ; 109: 48-56, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28676412

RESUMO

AIMS: Cardiovascular disease, one of the most common causes of death in western populations, is characterized by changes in RNA splicing and expression. Circular RNAs (circRNA) originate from back-splicing events, which link a downstream 5' splice site to an upstream 3' splice site. Several back-splicing junctions (BSJ) have been described in heart biopsies from human, rat and mouse hearts (Werfel et al., 2016; Jakobi et al., 2016 ). Here, we use human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) to identify circRNA and host gene dynamics in cardiac development and disease. In parallel, we explore candidate interactions of selected homologs in mouse and rat via RIP-seq experiments. METHODS AND RESULTS: Deep RNA sequencing of cardiomyocyte development and ß-adrenergic stimulation uncovered 4518 circRNAs. The set of circular RNA host genes is enriched for chromatin modifiers and GTPase activity regulators. RNA-seq and qRT-PCR data showed that circular RNA expression is highly dynamic in the hiPSC-CM model with 320 circRNAs showing significant expression changes. Intriguingly, 82 circRNAs are independently regulated to their host genes. We validated the same circRNA dynamics for circRNAs from ATXN10, CHD7, DNAJC6 and SLC8A1 in biopsy material from human dilated cardiomyopathy (DCM) and control patients. Finally, we could show that rodent homologs of circMYOD, circSLC8A1, circATXN7 and circPHF21A interact with either the ribosome or Argonaute2 protein complexes. CONCLUSION: CircRNAs are dynamically expressed in a hiPSC-CM model of cardiac development and stress response. Some circRNAs show similar, host-gene independent expression dynamics in patient samples and may interact with the ribosome and RISC complex. In summary, the hiPSC-CM model uncovered a new signature of potentially disease relevant circRNAs which may serve as novel therapeutic targets.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Cardiovasculares , Proteínas Musculares/biossíntese , Miócitos Cardíacos/metabolismo , RNA/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Proteínas Musculares/genética , Miócitos Cardíacos/patologia , RNA/genética , RNA Circular , Ratos
19.
Biomed Eng Online ; 16(1): 70, 2017 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-28595607

RESUMO

BACKGROUND: Electrophysiological behavior is of great importance for analyzing the cardiac functional mechanism under cardiac physiological and pathological condition. Due to the complexity of cardiac structure and biophysiological function, visualization of a cardiac electrophysiological model compositively is still a challenge. The lack of either modality of the whole organ structure or cardiac electrophysiological behaviors makes analysis of the intricate mechanisms of cardiac dynamic function a difficult task. This study aims at exploring 3D conduction of stimulus and electrical excitation reactivity on the level of organ with the authentic fine cardiac anatomy structure. METHODS: In this paper, a cardiac electrical excitation propagation model is established based on the human cardiac cross-sectional data to explore detailed cardiac electrical activities. A novel biophysical merging visualization method is then presented for biophysical integration of cardiac anatomy and electrophysiological properties in the form of the merging optical model, which provides the corresponding position, spatial relationship and the whole process in 3D space with the context of anatomical structure for representing the biophysical detailed electrophysiological activity. RESULTS: The visualization result present the action potential propagation of the left ventricle within the excitation cycle with the authentic fine cardiac organ anatomy. In the visualized images, all vital organs are identified and distinguished without ambiguity. The three dimensional spatial position, relation and the process of cardiac excitation conduction and re-entry propagation in the anatomical structure during the phase of depolarization and repolarization is also shown in the result images, which exhibits the performance of a more detailed biophysical understanding of the electrophysiological kinetics of human heart in vivo. CONCLUSIONS: Results suggest that the proposed merging optical model can merge cardiac electrophysiological activity with the anatomy structure. By specifying the respective opacity for the cardiac anatomy structure and the electrophysiological model in the merging attenuation function, the visualized images can provide an in-depth insight into the biophysical detailed cardiac functioning phenomena and the corresponding electrophysiological behavior mechanism, which is helpful for further speculating cardiac physiological and pathological responses and is fundamental to the cardiac research and clinical diagnoses.


Assuntos
Fenômenos Eletrofisiológicos , Coração/fisiologia , Modelos Cardiovasculares , Estudos Transversais , Coração/diagnóstico por imagem , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/fisiologia , Humanos , Imageamento por Ressonância Magnética , Tomografia Computadorizada por Raios X
20.
J Mol Cell Cardiol ; 100: 25-34, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27663173

RESUMO

In-silico models of human cardiac electrophysiology are now being considered for prediction of cardiotoxicity as part of the preclinical assessment phase of all new drugs. We ask the question whether any of the available models are actually fit for this purpose. We tested three models of the human ventricular action potential, the O'hara-Rudy (ORD11), the Grandi-Bers (GB10) and the Ten Tusscher (TT06) models. We extracted clinical QT data for LQTS1 and LQTS2 patients with nonsense mutations that would be predicted to cause 50% loss of function in IKs and IKr respectively. We also obtained clinical QT data for LQTS3 patients. We then used a global optimization approach to improve the existing in silico models so that they reproduced all three clinical data sets more closely. We also examined the effects of adrenergic stimulation in the different LQTS subsets. All models, in their original form, produce markedly different and unrealistic predictions of QT prolongation for LQTS1, 2 and 3. After global optimization of the maximum conductances for membrane channels, all models have similar current densities during the action potential, despite differences in kinetic properties of the channels in the different models, and more closely reproduce the prolongation of repolarization seen in all LQTS subtypes. In-silico models of cardiac electrophysiology have the potential to be tremendously useful in complementing traditional preclinical drug testing studies. However, our results demonstrate they should be carefully validated and optimized to clinical data before they can be used for this purpose.


Assuntos
Sistema de Condução Cardíaco , Ventrículos do Coração/fisiopatologia , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/fisiopatologia , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Fenótipo , Estudos de Casos e Controles , Simulação por Computador , Bases de Dados Factuais , Eletrocardiografia , Fenômenos Eletrofisiológicos , Humanos , Síndrome do QT Longo/etiologia , Miócitos Cardíacos/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA