12 research outputs found

    Biohybrid thin films for measuring contractility in engineered cardiovascular muscle.

    No full text
    In vitro cardiovascular disease models need to recapitulate tissue-scale function in order to provide in vivo relevance. We have developed a new method for measuring the contractility of engineered cardiovascular smooth and striated muscle in vitro during electrical and pharmacological stimulation. We present a growth theory-based finite elasticity analysis for calculating the contractile stresses of a 2D anisotropic muscle tissue cultured on a flexible synthetic polymer thin film. Cardiac muscle engineered with neonatal rat ventricular myocytes and paced at 0.5 Hz generated stresses of 9.2 +/- 3.5 kPa at peak systole, similar to measurements of the contractility of papillary muscle from adult rats. Vascular tissue engineered with human umbilical arterial smooth muscle cells maintained a basal contractile tone of 13.1 +/- 2.1 kPa and generated another 5.1 +/- 0.8 kPa when stimulated with endothelin-1. These data suggest that this method may be useful in assessing the efficacy and safety of pharmacological agents on cardiovascular tissue.</p

    Nuclear morphology and deformation in engineered cardiac myocytes and tissues.

    No full text
    Cardiac tissue engineering requires finely-tuned manipulation of the extracellular matrix (ECM) microenvironment to optimize internal myocardial organization. The myocyte nucleus is mechanically connected to the cell membrane via cytoskeletal elements, making it a target for the cellular response to perturbation of the ECM. However, the role of ECM spatial configuration and myocyte shape on nuclear location and morphology is unknown. In this study, printed ECM proteins were used to configure the geometry of cultured neonatal rat ventricular myocytes. Engineered one- and two-dimensional tissue constructs and single myocyte islands were assayed using live fluorescence imaging to examine nuclear position, morphology and motion as a function of the imposed ECM geometry during diastolic relaxation and systolic contraction. Image analysis showed that anisotropic tissue constructs cultured on microfabricated ECM lines possessed a high degree of nuclear alignment similar to that found in vivo; nuclei in isotropic tissues were polymorphic in shape with an apparently random orientation. Nuclear eccentricity was also increased for the anisotropic tissues, suggesting that intracellular forces deform the nucleus as the cell is spatially confined. During systole, nuclei experienced increasing spatial confinement in magnitude and direction of displacement as tissue anisotropy increased, yielding anisotropic deformation. Thus, the nature of nuclear displacement and deformation during systole appears to rely on a combination of the passive myofibril spatial organization and the active stress fields induced by contraction. Such findings have implications in understanding the genomic consequences and functional response of cardiac myocytes to their ECM surroundings under conditions of disease.</p

    Metabolic differences in engineered cardiac tissues on gels of different stiffness.

    No full text
    <p>(A) We measured oxygen consumption rates of NRVM cultured on soft (1 kPa), normal (13 kPa), and stiff (90 kPa) gels during a standard mitochondria stress test. We assessed basal respiration rate (B), ATP production (C), spare respiratory capacity (D), and nonmitochondrial respiration (E) for each substrate. Results are given as mean ± s.e.m. with sample size equal to 12, 15, and 22 for soft, normal, and stiff gels, respectively. The symbol * implies significant differences (p < 0.05) compared with the group of the same color.</p

    Contractile structure and function of cardiac tissues engineered on gels of varying stiffness.

    No full text
    <p>Using α-actinin immunographs from NRVM engineered on soft (1 kPa, A-i), normal (13 kPa, A-ii), and stiff (90 kPa, A-iii) gels we assessed differences in sarcomere length (SL) and sarcomere packing density (SPD, B). Scale bar 50 μm. Further, we performed TFM on diamond-shaped NRVM tissues engineered on soft (C-i), normal (C-ii), and stiff (C-iii) gels (peak systolic stress shown) to calculate (D) maximum deformation, peak stress, and total contractile work (strain energy). Results are given as mean ± s.e.m. and N = 3, 4, and 7 for soft, normal, and stiff gels respectively. The symbol * implies significant differences (p < 0.05) compared with the group of the same color.</p

    Cardiomyocytes operate optimally on substrates of physiological stiffness possibly due to a link between metabolism and contractile structure and function.

    No full text
    <p>Based on our data, we speculated that substrate stiffness might regulate the balance of energy production and utilization in cardiac tissues as follows. Cardiomyocytes on soft gels (A) need abundant ATP (white spheres) derived by mitochondria (orange organelles) to promote the contraction of sarcomeres (Z-disks in black) via cross-bridge cycling (brown lines). This might lead to very inefficient mechanical work as only a small stress (red arrows) is required to deform the soft gel substantially. Conversely, NRVM cultured on normal (B) and stiff (C) substrates have well organized contractile cytoskeletons and need a limited amount of ATP to fuel sarcomere contraction. Since the same amount of contractile force causes a smaller displacement of stiffer gels, the product between force and displacement (work) is maximum on gels of physiological stiffness.</p

    Micropatterned cardiac tissues on soft gels for traction force microscopy.

    No full text
    <p>(A) We cast PA gels (i) sandwiching the pre-polymer solution between activated and non-activated glass (ii) before stamping fibronectin (iii) to promote cell adhesion on the gel surface (iv). With this versatile method, we engineered neonate rat ventricular myocytes (NRVM) into diamond-shaped mini tissues (B) featuring cells aligned along the major axis of the diamond (C) thanks to a micro-contact printed brick-wall pattern of fibronectin (D). By tracking the displacement of fluorescent beads embedded in the in the soft gel during cell relaxation (E) and contraction (F), we used traction force microscopy to obtain displacement (G), and stress (H) maps at the tissue level. Importantly, since we could electrically stimulate the diamond-shaped tissues, we could measure displacement, stress, and contractile work as a function of beating frequency (I). Scale bars: 100 μm.</p

    Hierarchical architecture influences calcium dynamics in engineered cardiac muscle.

    No full text
    Changes in myocyte cell shape and tissue structure are concurrent with changes in electromechanical function in both the developing and diseased heart. While the anisotropic architecture of cardiac tissue is known to influence the propagation of the action potential, the influence of tissue architecture and its potential role in regulating excitation-contraction coupling (ECC) are less well defined. We hypothesized that changes in the shape and the orientation of cardiac myocytes induced by spatial arrangement of the extracellular matrix (ECM) affects ECC. To test this hypothesis, we isolated and cultured neonatal rat ventricular cardiac myocytes on various micropatterns of fibronectin where they self-organized into tissues with varying degrees of anisotropy. We then measured the morphological features of these engineered myocardial tissues across several hierarchical dimensions by measuring cellular aspect ratio, myocyte area, nuclear density and the degree of cytoskeletal F-actin alignment. We found that when compared with isotropic tissues, anisotropic tissues have increased cellular aspect ratios, increased nuclear densities, decreased myocyte cell areas and smaller variances in actin alignment. To understand how tissue architecture influences cardiac function, we studied the role of anisotropy on intracellular calcium ([Ca(2+)](i)) dynamics by characterizing the [Ca(2+)](i)-frequency relationship of electrically paced tissues. When compared with isotropic tissues, anisotropic tissues displayed significant differences in [Ca(2+)](i) transients, decreased diastolic baseline [Ca(2+)](i) levels and greater [Ca(2+)](i) influx per cardiac cycle. These results suggest that ECM cues influence tissue structure at cellular and subcellular levels and regulate ECC.</p

    Functional differences in engineered myocardium from embryonic stem cell-derived versus neonatal cardiomyocytes.

    No full text
    <p>Stem cell-derived cardiomyocytes represent unique tools for cell- and tissue-based regenerative therapies, drug discovery and safety, and studies of fundamental heart-failure mechanisms. However, the degree to which stem cell-derived cardiomyocytes compare to mature cardiomyocytes is often debated. We reasoned that physiological metrics of engineered cardiac tissues offer a means of comparison. We built laminar myocardium engineered from cardiomyocytes that were differentiated from mouse embryonic stem cell-derived cardiac progenitors or harvested directly from neonatal mouse ventricles, and compared their anatomy and physiology in vitro. Tissues assembled from progenitor-derived myocytes and neonate myocytes demonstrated similar cytoskeletal architectures but different gap junction organization and electromechanical properties. Progenitor-derived myocardium had significantly less contractile stress and slower longitudinal conduction velocity than neonate-derived myocardium, indicating that the developmental state of the cardiomyocytes affects the electromechanical function of the resultant engineered tissue. These data suggest a need to establish performance metrics for future stem cell applications.</p

    Controlling the contractile strength of engineered cardiac muscle by hierarchal tissue architecture.

    No full text
    <p>The heart is a muscular organ with a wrapping, laminar structure embedded with neural and vascular networks, collagen fibrils, fibroblasts, and cardiac myocytes that facilitate contraction. We hypothesized that these non-muscle components may have functional benefit, serving as important structural alignment cues in inter- and intra-cellular organization of cardiac myocytes. Previous studies have demonstrated that alignment of engineered myocardium enhances calcium handling, but how this impacts actual force generation remains unclear. Quantitative assays are needed to determine the effect of alignment on contractile function and muscle physiology. To test this, micropatterned surfaces were used to build 2-dimensional myocardium from neonatal rat ventricular myocytes with distinct architectures: confluent isotropic (serving as the unaligned control), confluent anisotropic, and 20 μm spaced, parallel arrays of multicellular myocardial fibers. We combined image analysis of sarcomere orientation with muscular thin film contractile force assays in order to calculate the peak sarcomere-generated stress as a function of tissue architecture. Here we report that increasing peak systolic stress in engineered cardiac tissues corresponds with increasing sarcomere alignment. This change is larger than would be anticipated from enhanced calcium handling and increased uniaxial alignment alone. These results suggest that boundary conditions (heterogeneities) encoded in the extracellular space can regulate muscle tissue function, and that structural organization and cytoskeletal alignment are critically important for maximizing peak force generation.</p

    ANG II treatment leads to decreased contractile function.

    No full text
    <p>(A,) MTF Chip. (A,i) Photograph of experimental setup (Scale bar: 1 cm). (A ii-iii) Schematic of the MTF chip in diastole (ii) and systole positions (iii). Microcontact printing was used to pattern cells to resemble the structure of heart tissue architecture mimicked on the MTF chip (Scale bar: 20 μm). (A,iv) Fibronectin patterns on surface of thin film drive tissue architecture (Scale bar: 20 μm). (A,v) Brightfield image of engineered cardiac tissues on the MTF. (B-D) (i) Diastole and (ii) peak systole of muscular thin films. Blue outline represents the original film length, red line represents the x-projection of the radius of curvature of the film (Scale bar: 500 μm). (E) Representative stress traces generated from x-projections of films (F) ANG II treatment leads to a decrease in contractile stress generation. Control n = 85 tissues, 5 nM n = 15 tissues, 100 nM n = 69 tissues, 3 harvests. Tissues were paced at 2Hz (mean ± SEM, * indicates p < 0.05 vs control, # indicates p < 0.05 vs. 100 nM).</p
    corecore