Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cells Tissues Organs ; 212(1): 96-110, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35008089

RESUMEN

Peristalsis is a nuanced mechanical stimulus comprised of multi-axial strain (radial and axial strain) and shear stress. Forces associated with peristalsis regulate diverse biological functions including digestion, reproductive function, and urine dynamics. Given the central role peristalsis plays in physiology and pathophysiology, we were motivated to design a bioreactor capable of holistically mimicking peristalsis. We engineered a novel rotating screw-drive based design combined with a peristaltic pump, in order to deliver multi-axial strain and concurrent shear stress to a biocompatible polydimethylsiloxane (PDMS) membrane "wall." Radial indentation and rotation of the screw drive against the wall demonstrated multi-axial strain evaluated via finite element modeling. Experimental measurements of strain using piezoelectric strain resistors were in close alignment with model-predicted values (15.9 ± 4.2% vs. 15.2% predicted). Modeling of shear stress on the "wall" indicated a uniform velocity profile and a moderate shear stress of 0.4 Pa. Human mesenchymal stem cells (hMSCs) seeded on the PDMS "wall" and stimulated with peristalsis demonstrated dramatic changes in actin filament alignment, proliferation, and nuclear morphology compared to static controls, perfusion, or strain, indicating that hMSCs sensed and responded to peristalsis uniquely. Lastly, significant differences were observed in gene expression patterns of calponin, caldesmon, smooth muscle actin, and transgelin, corroborating the propensity of hMSCs toward myogenic differentiation in response to peristalsis. Collectively, our data suggest that the peristalsis bioreactor is capable of generating concurrent multi-axial strain and shear stress on a "wall." hMSCs experience peristalsis differently than perfusion or strain, resulting in changes in proliferation, actin fiber organization, smooth muscle actin expression, and genetic markers of differentiation. The peristalsis bioreactor device has broad utility in the study of development and disease in several organ systems.


Asunto(s)
Biomimética , Peristaltismo , Humanos , Peristaltismo/fisiología , Actinas , Diferenciación Celular , Reactores Biológicos
2.
J Biomech Eng ; 144(7)2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34993547

RESUMEN

Three-dimensional (3D) extrusion bioprinting typically requires an ad hoc trial-and-error optimization of the ink composition toward enhanced resolution. The ink solutions are solidified after leaving cone-shaped or cylindrical nozzles. The presence of ink instability not only hampers the extrusion resolution but also affects the behavior of embedded cellular components. This is a key factor in selecting (bio)inks and bioprinting design parameters for well-established desktop and handheld bioprinters. In this work, we developed an analytical solution for the process of ink deposition and compared its predictions against numerical simulations of the deposition. We estimated the onset of ink instability as a function of ink rheological properties and nozzle geometry. Our analytical results suggest that enhancing the shear-thinning behavior of the ink shortens the toe region of the deposition. Such an extrusion process is often desired, as it leads to faster depositions. However, we demonstrated that such conditions increase the possibility of lateral buckling of the strand once touching the substrate defined as instability in this study. The present study serves as a benchmark for detailed simulations of the extrusion process for optimal bioprinting.


Asunto(s)
Bioimpresión , Tinta , Bioimpresión/métodos , Hidrogeles/química , Impresión Tridimensional , Reología , Ingeniería de Tejidos/métodos
3.
Annu Rev Biomed Eng ; 21: 417-442, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167105

RESUMEN

Understanding and predicting the mechanical behavior of myocardium under healthy and pathophysiological conditions are vital to developing novel cardiac therapies and promoting personalized interventions. Within the past 30 years, various constitutive models have been proposed for the passive mechanical behavior of myocardium. These models cover a broad range of mathematical forms, microstructural observations, and specific test conditions to which they are fitted. We present a critical review of these models, covering both phenomenological and structural approaches, and their relations to the underlying structure and function of myocardium. We further explore the experimental and numerical techniques used to identify the model parameters. Next, we provide a brief overview of continuum-level electromechanical models of myocardium, with a focus on the methods used to integrate the active and passive components of myocardial behavior. We conclude by pointing to future directions in the areas of optimal form as well as new approaches for constitutive modeling of myocardium.


Asunto(s)
Corazón/fisiología , Modelos Cardiovasculares , Animales , Fenómenos Biomecánicos , Ingeniería Biomédica , Colágeno/química , Colágeno/fisiología , Simulación por Computador , Fenómenos Electrofisiológicos , Corazón/anatomía & histología , Humanos , Contracción Miocárdica/fisiología , Miocardio/química , Miocardio/ultraestructura , Miocitos Cardíacos/química , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/ultraestructura , Miofibrillas/química , Miofibrillas/fisiología
4.
J Biomech Eng ; 141(9)2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31260516

RESUMEN

Pulmonary arterial hypertension (PAH) exerts substantial pressure overload on the right ventricle (RV), inducing RV remodeling and myocardial tissue adaptation often leading to right heart failure. The associated RV free wall (RVFW) adaptation involves myocardial hypertrophy, augmented intrinsic contractility, collagen fibrosis, and structural remodeling in an attempt to cope with pressure overload. If RVFW adaptation cannot maintain the RV stroke volume (SV), RV dilation will prevail as an exit mechanism, which usually decompensates RV function, leading to RV failure. Our knowledge of the factors determining the transition from the upper limit of RVFW adaptation to RV decompensation and the role of fiber remodeling events such as extracellular fibrosis and fiber reorientation in this transition remains very limited. Computational heart models that connect the growth and remodeling (G&R) events at the fiber and tissue levels with alterations in the organ-level function are essential to predict the temporal order and the compensatory level of the underlying mechanisms. In this work, building upon our recently developed rodent heart models (RHM) of PAH, we integrated mathematical models that describe volumetric growth of the RV and structural remodeling of the RVFW. The time-evolution of RV remodeling from control and post-PAH time points was simulated. The results suggest that the augmentation of the intrinsic contractility of myofibers, accompanied by an increase in passive stiffness of RVFW, is among the first remodeling events through which the RV strives to maintain the cardiac output. Interestingly, we found that the observed reorientation of the myofibers toward the longitudinal (apex-to-base) direction was a maladaptive mechanism that impaired the RVFW contractile pattern and advanced along with RV dilation at later stages of PAH. In fact, although individual fibers were more contractile post-PAH, the disruption in the optimal transmural fiber architecture compromised the effective contractile function of the RVFW, contributing to the depressed ejection fraction (EF) of the RV. Our findings clearly demonstrate the critical need for developing multiscale approaches that can model and delineate relationships between pathological alterations in cardiac function and underlying remodeling events across fiber, cellular, and molecular levels.

5.
ArXiv ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38855538

RESUMEN

Left ventricular diastolic dysfunction (LVDD) is a group of diseases that adversely affect the passive phase of the cardiac cycle and can lead to heart failure. While left ventricular end-diastolic pressure (LVEDP) is a valuable prognostic measure in LVDD patients, traditional invasive methods of measuring LVEDP present risks and limitations, highlighting the need for alternative approaches. This paper investigates the possibility of measuring LVEDP non-invasively using inverse in-silico modeling. We propose the adoption of patient-specific cardiac modeling and simulation to estimate LVEDP and myocardial stiffness from cardiac strains. We have developed a high-fidelity patient-specific computational model of the left ventricle. Through an inverse modeling approach, myocardial stiffness and LVEDP were accurately estimated from cardiac strains that can be acquired from in vivo imaging, indicating the feasibility of computational modeling to augment current approaches in the measurement of ventricular pressure. Integration of such computational platforms into clinical practice holds promise for early detection and comprehensive assessment of LVDD with reduced risk for patients.

6.
Acta Biomater ; 173: 109-122, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37925122

RESUMEN

Myocardial infarction (MI) is accompanied by the formation of a fibrotic scar in the left ventricle (LV) and initiates significant alterations in the architecture and constituents of the LV free wall (LVFW). Previous studies have shown that LV adaptation is highly individual, indicating that the identification of remodeling mechanisms post-MI demands a fully subject-specific approach that can integrate a host of structural alterations at the fiber-level to changes in bulk biomechanical adaptation at the tissue-level. We present an image-driven micromechanical approach to characterize remodeling, assimilating new biaxial mechanical data, histological studies, and digital image correlation data within an in-silico framework to elucidate the fiber-level remodeling mechanisms that drive tissue-level adaptation for each subject. We found that a progressively diffused collagen fiber structure combined with similarly disorganized myofiber architecture in the healthy region leads to the loss of LVFW anisotropy post-MI, offering an important tissue-level hallmark for LV maladaptation. In contrast, our results suggest that reductions in collagen undulation are an adaptive mechanism competing against LVFW thinning. Additionally, we show that the inclusion of subject-specific geometry when modeling myocardial tissue is essential for accurate prediction of tissue kinematics. Our approach serves as an essential step toward identifying fiber-level remodeling indices that govern the transition of MI to systolic heart failure. These indices complement the traditional, organ-level measures of LV anatomy and function that often fall short of early prognostication of heart failure in MI. In addition, our approach offers an integrated methodology to advance the design of personalized interventions, such as hydrogel injection, to reinforce and suppress native adaptive and maladaptive mechanisms, respectively, to prevent the transition of MI to heart failure. STATEMENT OF SIGNIFICANCE: Biomechanical and architectural adaptation of the LVFW remains a central, yet overlooked, remodeling process post-MI. Our study indicates the biomechanical adaptation of the LVFW post-MI is highly individual and driven by altered fiber network architecture and collective changes in collagen fiber content, undulation, and stiffness. Our findings demonstrate the possibility of using cardiac strains to infer such fiber-level remodeling events through in-silico modeling, paving the way for in-vivo characterization of multiscale biomechanical indices in humans. Such indices will complement the traditional, organ-level measures of LV anatomy and function that often fall short of early prognostication of heart failure in MI.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Humanos , Remodelación Ventricular , Miocardio/patología , Infarto del Miocardio/patología , Insuficiencia Cardíaca/patología , Colágeno
7.
Res Sq ; 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38883756

RESUMEN

Myocardial infarction (MI) continues to be a leading cause of death worldwide. The precise quantification of infarcted tissue is crucial to diagnosis, therapeutic management, and post-MI care. Late gadolinium enhancement-cardiac magnetic resonance (LGE-CMR) is regarded as the gold standard for precise infarct tissue localization in MI patients. A fundamental limitation of LGE-CMR is the invasive intravenous introduction of gadolinium-based contrast agents that present potential high-risk toxicity, particularly for individuals with underlying chronic kidney diseases. Herein, we develop a completely non-invasive methodology that identifies the location and extent of an infarct region in the left ventricle via a machine learning (ML) model using only cardiac strains as inputs. In this transformative approach, we demonstrate the remarkable performance of a multi-fidelity ML model that combines rodent-based in-silico-generated training data (low-fidelity) with very limited patient-specific human data (high-fidelity) in predicting LGE ground truth. Our results offer a new paradigm for developing feasible prognostic tools by augmenting synthetic simulation-based data with very small amounts of in-vivo human data. More broadly, the proposed approach can significantly assist with addressing biomedical challenges in healthcare where human data are limited.

8.
bioRxiv ; 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38895325

RESUMEN

Myocardial infarction (MI) continues to be a leading cause of death worldwide. The precise quantification of infarcted tissue is crucial to diagnosis, therapeutic management, and post-MI care. Late gadolinium enhancement-cardiac magnetic resonance (LGE-CMR) is regarded as the gold standard for precise infarct tissue localization in MI patients. A fundamental limitation of LGE-CMR is the invasive intravenous introduction of gadolinium-based contrast agents that present potential high-risk toxicity, particularly for individuals with underlying chronic kidney diseases. Herein, we develop a completely non-invasive methodology that identifies the location and extent of an infarct region in the left ventricle via a machine learning (ML) model using only cardiac strains as inputs. In this transformative approach, we demonstrate the remarkable performance of a multi-fidelity ML model that combines rodent-based in-silico-generated training data (low-fidelity) with very limited patient-specific human data (high-fidelity) in predicting LGE ground truth. Our results offer a new paradigm for developing feasible prognostic tools by augmenting synthetic simulation-based data with very small amounts of in-vivo human data. More broadly, the proposed approach can significantly assist with addressing biomedical challenges in healthcare where human data are limited.

9.
bioRxiv ; 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38854032

RESUMEN

Aims: Pulmonary hypertension (PH) results in an increase in RV afterload, leading to RV dysfunction and failure. The mechanisms underlying maladaptive RV remodeling are poorly understood. In this study, we investigated the multiscale and mechanistic nature of RV free wall (RVFW) biomechanical remodeling and its correlations with RV function adaptations. Methods and Results: Mild and severe models of PH, consisting of hypoxia (Hx) model in Sprague-Dawley (SD) rats (n=6 each, Control and PH) and Sugen-hypoxia (SuHx) model in Fischer (CDF) rats (n=6 each, Control and PH), were used. Organ-level function and tissue-level stiffness and microstructure were quantified through in-vivo and ex-vivo measures, respectively. Multiscale analysis was used to determine the association between fiber-level remodeling, tissue-level stiffening, and organ-level dysfunction. Animal models with different PH severity provided a wide range of RVFW stiffening and anisotropy alterations in PH. Decreased RV-pulmonary artery (PA) coupling correlated strongly with stiffening but showed a weaker association with the loss of RVFW anisotropy. Machine learning classification identified the range of adaptive and maladaptive RVFW stiffening. Multiscale modeling revealed that increased collagen fiber tautness was a key remodeling mechanism that differentiated severe from mild stiffening. Myofiber orientation analysis indicated a shift away from the predominantly circumferential fibers observed in healthy RVFW specimens, leading to a significant loss of tissue anisotropy. Conclusion: Multiscale biomechanical analysis indicated that although hypertrophy and fibrosis occur in both mild and severe PH, certain fiber-level remodeling events, including increased tautness in the newly deposited collagen fibers and significant reorientations of myofibers, contributed to excessive biomechanical maladaptation of the RVFW leading to severe RV-PA uncoupling. Collagen fiber remodeling and the loss of tissue anisotropy can provide an improved understanding of the transition from adaptive to maladaptive remodeling. Translational perspective: Right ventricular (RV) failure is a leading cause of mortality in patients with pulmonary hypertension (PH). RV diastolic and systolic impairments are evident in PH patients. Stiffening of the RV wall tissue and changes in the wall anisotropy are expected to be major contributors to both impairments. Global assessments of the RV function remain inadequate in identifying patients with maladaptive RV wall remodeling primarily due to their confounded and weak representation of RV fiber and tissue remodeling events. This study provides novel insights into the underlying mechanisms of RV biomechanical remodeling and identifies the adaptive-to-maladaptive transition across the RV biomechanics-function spectrum. Our analysis dissecting the contribution of different RV wall remodeling events to RV dysfunction determines the most adverse fiber-level remodeling to RV dysfunction as new therapeutic targets to curtail RV maladaptation and, in turn, RV failure in PH.

10.
bioRxiv ; 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38895261

RESUMEN

The quantification of cardiac motion using cardiac magnetic resonance imaging (CMR) has shown promise as an early-stage marker for cardiovascular diseases. Despite the growing popularity of CMR-based myocardial strain calculations, measures of complete spatiotemporal strains (i.e., three-dimensional strains over the cardiac cycle) remain elusive. Complete spatiotemporal strain calculations are primarily hampered by poor spatial resolution, with the rapid motion of the cardiac wall also challenging the reproducibility of such strains. We hypothesize that a super-resolution reconstruction (SRR) framework that leverages combined image acquisitions at multiple orientations will enhance the reproducibility of complete spatiotemporal strain estimation. Two sets of CMR acquisitions were obtained for five wild-type mice, combining short-axis scans with radial and orthogonal long-axis scans. Super-resolution reconstruction, integrated with tissue classification, was performed to generate full four-dimensional (4D) images. The resulting enhanced and full 4D images enabled complete quantification of the motion in terms of 4D myocardial strains. Additionally, the effects of SRR in improving accurate strain measurements were evaluated using an in-silico heart phantom. The SRR framework revealed near isotropic spatial resolution, high structural similarity, and minimal loss of contrast, which led to overall improvements in strain accuracy. In essence, a comprehensive methodology was generated to quantify complete and reproducible myocardial deformation, aiding in the much-needed standardization of complete spatiotemporal strain calculations.

11.
Adv Sci (Weinh) ; : e2400476, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696618

RESUMEN

Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.

12.
Artículo en Inglés | MEDLINE | ID: mdl-37584008

RESUMEN

Calculating cardiac strains through speckle tracking echocardiography (STE) has shown promise as prognostic markers linked to functional indices and disease outcomes. However, the presence of acoustic shadowing often challenges the accuracy of STE in small animals such as rodents. The shadowing arises due to the complex anatomy of rodents, with operator dexterity playing a significant role in image quality. The effects of the semi-transparent shadows are further exacerbated in right ventricular (RV) imaging due to the thinness and rapid motion of the RV free wall (RVFW). The movement of the RVFW across the shadows distorts speckle tracking and produces unnatural and non-physical strains. The objective of this study was to minimize the effects of shadowing on STE by distinguishing "out-of-shadow" motion and identifying speckles in and out of shadow. Parasternal 2D echocardiography was performed, and short-axis B-mode (SA) images of the RVFW were acquired for a rodent model of pulmonary hypertension (n = 1). Following image acquisition, a denoising algorithm using edge-enhancing anisotropic diffusion (EED) was implemented, and the ensuing effects on strain analysis were visualized using a custom STE pipeline. Speckles in the shadowed regions were identified through a correlation between the filtered image and the original acquisition. Thus, pixel movement across the boundary was identified by enhancing the distinction between the shadows and the cardiac wall, and non-physical strains were suppressed. The strains obtained through STE showed expected patterns with enhanced circumferential contractions in the central region of the RVFW in contrast to smaller and nearly uniform strains derived from the unprocessed images.

13.
Comput Biol Med ; 163: 107134, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37379617

RESUMEN

Impaired relaxation of cardiomyocytes leads to diastolic dysfunction in the left ventricle. Relaxation velocity is regulated in part by intracellular calcium (Ca2+) cycling, and slower outflux of Ca2+ during diastole translates to reduced relaxation velocity of sarcomeres. Sarcomere length transient and intracellular calcium kinetics are integral parts of characterizing the relaxation behavior of the myocardium. However, a classifier tool that can separate normal cells from cells with impaired relaxation using sarcomere length transient and/or calcium kinetics remains to be developed. In this work, we employed nine different classifiers to classify normal and impaired cells, using ex-vivo measurements of sarcomere kinematics and intracellular calcium kinetics data. The cells were isolated from wild-type mice (referred to as normal) and transgenic mice expressing impaired left ventricular relaxation (referred to as impaired). We utilized sarcomere length transient data with a total of n = 126 cells (n = 60 normal cells and n = 66 impaired cells) and intracellular calcium cycling measurements with a total of n = 116 cells (n = 57 normal cells and n = 59 impaired cells) from normal and impaired cardiomyocytes as inputs to machine learning (ML) models for classification. We trained all ML classifiers with cross-validation method separately using both sets of input features, and compared their performance metrics. The performance of classifiers on test data showed that our soft voting classifier outperformed all other individual classifiers on both sets of input features, with 0.94 and 0.95 area under the receiver operating characteristic curves for sarcomere length transient and calcium transient, respectively, while multilayer perceptron achieved comparable scores of 0.93 and 0.95, respectively. However, the performance of decision tree, and extreme gradient boosting was found to be dependent on the set of input features used for training. Our findings highlight the importance of selecting appropriate input features and classifiers for the accurate classification of normal and impaired cells. Layer-wise relevance propagation (LRP) analysis demonstrated that the time to 50% contraction of the sarcomere had the highest relevance score for sarcomere length transient, whereas time to 50% decay of calcium had the highest relevance score for calcium transient input features. Despite the limited dataset, our study demonstrated satisfactory accuracy, suggesting that the algorithm can be used to classify relaxation behavior in cardiomyocytes when the potential relaxation impairment of the cells is unknown.


Asunto(s)
Calcio , Sarcómeros , Ratones , Animales , Contracción Miocárdica , Miocitos Cardíacos , Aprendizaje Automático
14.
Artículo en Inglés | MEDLINE | ID: mdl-37565032

RESUMEN

There are several lung diseases that lead to alterations in regional lung mechanics, including acute respiratory distress syndrome. Such alterations can lead to localized underventilation of the affected areas resulting in the overdistension of the surrounding healthy regions. They can also lead to the surrounding alveoli expanding unevenly or distorting. Therefore, the quantification of the regional deformation in the lungs offers insights into identifying the regions at risk of lung injury. Although few recent studies have developed image processing techniques to quantify the regional volumetric deformation in the lung from dynamic imaging, the presence and extent of distortional deformation in the lung, and its correlation with volumetric deformation, remain poorly understood. In this study, we present a method that uses the four-dimensional displacement field obtained from image registration to quantify both regional volumetric and distortional deformation in the lung. We used dynamic computed tomography scans in a healthy rat over the course of one respiratory cycle in free breathing. Non-rigid image registration was performed to quantify voxel displacement during respiration. The deformation gradient was calculated using the displacement field and its determinant was used to quantify regional volumetric deformation. Regional distortion was calculated as the ratio of maximum to minimum principal stretches using the isochoric part of the Cauchy green tensor. We found an inverse correlation between volumetric strains and distortion indicating that poorly expanding alveoli tend to experience larger distortion. The combination of regional volumetric strains and distortion may serve as high-fidelity biomarkers to identify the regions at risk of most adverse lung injuries.

15.
Acta Biomater ; 162: 240-253, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36963596

RESUMEN

The myocardium possesses an intricately designed microarchitecture to produce an optimal cardiac contraction. The contractile behavior of the heart is generated at the sarcomere level and travels across several length scales to manifest as the systolic function at the organ level. While passive myocardial behavior has been studied extensively, the translation of active tension produced at the fiber level to the organ-level function is not well understood. Alterations in cardiac systolic function are often key sequelae in structural heart diseases, such as myocardial infarction and systolic heart failure; thus, characterization of the contractile behavior of the heart across multiple length scales is essential to improve our understanding of mechanisms collectively leading to depressed systolic function. In this study, we present a methodology to characterize the active behavior of left ventricle free wall (LVFW) myocardial tissues in mice. Combined with active tests in papillary muscle fibers and conventional in vivo contractility measurement at the organ level in an animal-specific manner, we establish a multiscale active characterization of the heart from fiber to organ. In addition, we quantified myocardial architecture from histology to shed light on the directionality of the contractility at the tissue level. The LVFW tissue activation-relaxation behavior under isometric conditions was qualitatively similar to that of the papillary muscle fiber bundle. However, the maximum stress developed in the LVFW tissue was an order of magnitude lower than that developed by a fiber bundle, and the time taken for active forces to plateau was 2-3 orders of magnitude longer. Although the LVFW tissue exhibited a slightly stiffer passive response in the circumferential direction, the tissues produced significantly larger active stresses in the longitudinal direction during active testing. Also, contrary to passive viscoelastic stress relaxation, active stresses relaxed faster in the direction with larger peak stresses. The multiscale experimental pipeline presented in this work is expected to provide crucial insight into the contractile adaptation mechanisms of the heart with impaired systolic function. STATEMENT OF SIGNIFICANCE: Heart failure cause significant alterations to the contractile-relaxation behavior of the yocardium. Multiscale characterization of the contractile behavior of the myocardium is essential to advance our understanding of how contractility translates from fiber to organ and to identify the multiscale mechanisms leading to impaired cardiac function. While passive myocardial behavior has been studied extensively, the investigation of tissue-level contractile behavior remains critically scarce in the literature. To the best of our knowledge, our study here is the first to investigate the contractile behavior of the left ventricle at multiple length scales in small animals. Our results indicate that the active myocardial wall is a function of transmural depth and relaxes faster in the direction with larger peak stresses.


Asunto(s)
Ventrículos Cardíacos , Corazón , Ratones , Animales , Corazón/fisiología , Miocardio , Contracción Miocárdica , Sístole
16.
Funct Imaging Model Heart ; 13958: 34-43, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37465393

RESUMEN

Myocardial infarction (MI) results in cardiac myocyte death and often initiates the formation of a fibrotic scar in the myocardium surrounded by a border zone. Myocyte loss and collagen-rich scar tissue heavily influence the biomechanical behavior of the myocardium which could lead to various cardiac diseases such as systolic heart failure and arrhythmias. Knowledge of how myocyte and collagen micro-architecture changes affect the passive mechanical behavior of the border zone remains limited. Computational modeling provides us with an invaluable tool to identify and study the mechanisms driving the biomechanical remodeling of the myocardium post-MI. We utilized a rodent model of MI and an image-based approach to characterize the three-dimensional (3-D) myocyte and collagen micro-architecture at various timepoints post-MI. Left ventricular free wall (LVFW) samples were obtained from infarcted hearts at 1-week and 4-week post-MI (n = 1 each). Samples were labeled using immunoassays to identify the extracellular matrix (ECM) and myocytes. 3-D reconstructions of the infarct border zone were developed from confocal imaging and meshed to develop high-fidelity micro-anatomically accurate finite element models. We performed a parametric study using these models to investigate the influence of collagen undulation on the passive micromechanical behavior of the myocardium under a diastolic load. Our results suggest that although parametric increases in collagen undulation elevate the strain amount experienced by the ECM in both early- and late-stage MI, the sensitivity of myocytes to such increases is reduced from early to late-stage MI. Our 3-D micro-anatomical modeling holds promise in identifying mechanisms of border zone maladaptation post-MI.

17.
J Mech Behav Biomed Mater ; 142: 105788, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37060716

RESUMEN

We have previously demonstrated the importance of myofiber-collagen mechanical interactions in modeling the passive mechanical behavior of right ventricle free wall (RVFW) myocardium. To gain deeper insights into these coupling mechanisms, we developed a high-fidelity, micro-anatomically realistic 3D finite element model of right ventricle free wall (RVFW) myocardium by combining high-resolution imaging and supercomputer-based simulations. We first developed a representative tissue element (RTE) model at the sub-tissue scale by specializing the hyperelastic anisotropic structurally-based constitutive relations for myofibers and ECM collagen, and equi-biaxial and non-equibiaxial loading conditions were simulated using the open-source software FEniCS to compute the effective stress-strain response of the RTE. To estimate the model parameters of the RTE model, we first fitted a 'top-down' biaxial stress-strain behavior with our previous structurally based (tissue-scale) model, informed by the measured myofiber and collagen fiber composition and orientation distributions. Next, we employed a multi-scale approach to determine the tissue-level (5 x 5 x 0.7 mm specimen size) RVFW biaxial behavior via 'bottom-up' homogenization of the fitted RTE model, recapitulating the histologically measured myofiber and collagen orientation to the biaxial mechanical data. Our homogenization approach successfully reproduced the tissue-level mechanical behavior of our previous studies in all biaxial deformation modes, suggesting that the 3D micro-anatomical arrangement of myofibers and ECM collagen is indeed a primary mechanism driving myofiber-collagen interactions.


Asunto(s)
Ventrículos Cardíacos , Miocardio , Estrés Mecánico , Miocardio/patología , Corazón , Colágeno , Fenómenos Biomecánicos
18.
Ann Biomed Eng ; 51(4): 846-863, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36394778

RESUMEN

Myocardial infarction (MI) results in cardiac myocyte death and the formation of a fibrotic scar in the left ventricular free wall (LVFW). Following an acute MI, LVFW remodeling takes place consisting of several alterations in the structure and properties of cellular and extracellular components with a heterogeneous pattern across the LVFW. The normal function of the heart is strongly influenced by the passive and active biomechanical behavior of the LVFW, and progressive myocardial structural remodeling can have a detrimental effect on both diastolic and systolic functions of the LV leading to heart failure. Despite important advances in understanding LVFW passive remodeling in the setting of MI, heterogeneous remodeling in the LVFW active properties and its relationship to organ-level LV function remain understudied. To address these gaps, we developed high-fidelity finite-element (FE) rodent computational cardiac models (RCCMs) of MI using extensive datasets from MI rat hearts representing the heart remodeling from one-week (1-wk) to four-week (4-wk) post-MI timepoints. The rat-specific models (n = 2 for each timepoint) integrate detailed imaging data of the heart geometry, myocardial fiber architecture, and infarct zone determined using late gadolinium enhancement prior to terminal measurements. The computational models predicted a significantly higher level of active tension in remote myocardium in early post-MI hearts (1-wk post-MI) followed by a return to near the control level in late-stage MI (3- and 4-wk post-MI). The late-stage MI rats showed smaller myofiber ranges in the remote region and in-silico experiments using RCCMs suggested that the smaller fiber helicity is consistent with lower contractile forces needed to meet the measured ejection fractions in late-stage MI. In contrast, in-silico experiments predicted that collagen fiber transmural orientation in the infarct region has little influence on organ-level function. In addition, our MI RCCMs indicated that reduced and potentially positive circumferential strains in the infarct region at end-systole can be used to infer information about the time-varying properties of the infarct region. The detailed description of regional passive and active remodeling patterns can complement and enhance the traditional measures of LV anatomy and function that often lead to a gross and limited assessment of cardiac performance. The translation and implementation of our model in patient-specific organ-level simulations offer to advance the investigation of individualized prognosis and intervention for MI.


Asunto(s)
Ventrículos Cardíacos , Infarto del Miocardio , Ratas , Animales , Medios de Contraste , Roedores , Gadolinio , Miocardio , Simulación por Computador , Remodelación Ventricular
19.
Funct Imaging Model Heart ; 13958: 74-83, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37671365

RESUMEN

The myocardium is composed of a complex network of contractile myofibers that are organized in such a way as to produce efficient contraction and relaxation of the heart. The myofiber architecture in the myocardium is a key determinant of cardiac motion and the global or organ-level function of the heart. Reports of architectural remodeling in cardiac diseases, such as pulmonary hypertension and myocardial infarction, potentially contributing to cardiac dysfunction call for the inclusion of an architectural marker for an improved assessment of cardiac function. However, the in-vivo quantification of three-dimensional myo-architecture has proven challenging. In this work, we examine the sensitivity of cardiac strains to varying myofiber orientation using a multiscale finite-element model of the LV. Additionally, we present an inverse modeling approach to predict the myocardium fiber structure from cardiac strains. Our results indicate a strong correlation between fiber orientation and LV kinematics, corroborating that the fiber structure is a principal determinant of LV contractile behavior. Our inverse model was capable of accurately predicting the myocardial fiber range and regional fiber angles from strain measures. A concrete understanding of the link between LV myofiber structure and motion, and the development of non-invasive and feasible means of characterizing the myocardium architecture is expected to lead to advanced LV functional metrics and improved prognostic assessment of structural heart disease.

20.
Circ Heart Fail ; 16(2): e009768, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36748476

RESUMEN

BACKGROUND: Global indices of right ventricle (RV) function provide limited insights into mechanisms underlying RV remodeling in pulmonary hypertension (PH). While RV myocardial architectural remodeling has been observed in PH, its effect on RV adaptation is poorly understood. METHODS: Hemodynamic assessments were performed in 2 rodent models of PH. RV free wall myoarchitecture was quantified using generalized Q-space imaging and tractography analyses. Computational models were developed to predict RV wall strains. Data from animal studies were analyzed to determine the correlations between hemodynamic measurements, RV strains, and structural measures. RESULTS: In contrast to the PH rats with severe RV maladaptation, PH rats with mild RV maladaptation showed a decrease in helical range of fiber orientation in the RV free wall (139º versus 97º; P=0.029), preserved global circumferential strain, and exhibited less reduction in right ventricular-pulmonary arterial coupling (0.029 versus 0.017 mm/mm Hg; P=0.037). Helical range correlated positively with coupling (P=0.036) and stroke volume index (P<0.01). Coupling correlated with global circumferential strain (P<0.01) and global radial strain (P<0.01) but not global longitudinal strain. CONCLUSIONS: Data analysis suggests that adaptive RV architectural remodeling could improve RV function in PH. Our findings suggest the need to assess RV architecture within routine screenings of PH patients to improve our understanding of its prognostic and therapeutic significance in PH.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión Pulmonar , Disfunción Ventricular Derecha , Animales , Ratas , Hemodinámica , Ventrículos Cardíacos , Adaptación Fisiológica , Función Ventricular Derecha , Remodelación Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA