Skip to main content
Research Article
Originally Published 11 April 2016
Free Access

Extracellular Matrix–Mediated Maturation of Human Pluripotent Stem Cell–Derived Cardiac Monolayer Structure and Electrophysiological Function

Circulation: Arrhythmia and Electrophysiology

Abstract

Background—

Human pluripotent stem cell–derived cardiomyocytes (hPSC-CMs) monolayers generated to date display an immature embryonic-like functional and structural phenotype that limits their utility for research and cardiac regeneration. In particular, the electrophysiological function of hPSC-CM monolayers and bioengineered constructs used to date are characterized by slow electric impulse propagation velocity and immature action potential profiles.

Methods and Results—

Here, we have identified an optimal extracellular matrix for significant electrophysiological and structural maturation of hPSC-CM monolayers. hPSC-CM plated in the optimal extracellular matrix combination have impulse propagation velocities ≈2× faster than previously reported (43.6±7.0 cm/s; n=9) and have mature cardiomyocyte action potential profiles, including hyperpolarized diastolic potential and rapid action potential upstroke velocity (146.5±17.7 V/s; n=5 monolayers). In addition, the optimal extracellular matrix promoted hypertrophic growth of cardiomyocytes and the expression of key mature sarcolemmal (SCN5A, Kir2.1, and connexin43) and myofilament markers (cardiac troponin I). The maturation process reported here relies on activation of integrin signaling pathways: neutralization of β1 integrin receptors via blocking antibodies and pharmacological blockade of focal adhesion kinase activation prevented structural maturation.

Conclusions—

Maturation of human stem cell–derived cardiomyocyte monolayers is achieved in a 1-week period by plating cardiomyocytes on PDMS (polydimethylsiloxane) coverslips rather than on conventional 2-dimensional cell culture formats, such as glass coverslips or plastic dishes. Activation of integrin signaling and focal adhesion kinase is essential for significant maturation of human cardiac monolayers.

WHAT IS KNOWN

Human cardiomyocytes derived from stem cells routinely demonstrate a fetal-like, immature functional and structural phenotype.
Maturation of stem cell–derived cardiomyocytes is necessary to promote the utility of these cells for drug discovery and hypothesis testing.

WHAT THE STUDY ADDS

Human stem cell–derived cardiac monolayers mature rapidly when formed on soft, pliable substrates rather than on rigid cell culture surfaces.
Electric impulse propagation of human stem cell–derived cardiac monolayers can be as fast as ≈50 cm/s; similar to the velocity in the adult cardiac ventricle.
Integrin receptor activation is crucial for significant maturation of human stem cell–derived cardiac monolayers.
The generation of induced human pluripotent stem cell–derived cardiomyocytes (hPSC-CMs) offers a revolutionary platform to study physiological/pathophysiological processes and cardiac arrhythmia mechanisms and for the development of new therapies in vitro.1,2 Experimentally, they offer a human system that can be used to study drug cardiotoxicity and the molecular mechanisms of mono-/poly-genetic diseases.3,4 Recently, we reported on the generation of large (1-cm diameter) electrically coupled hPSC-CM monolayers.5 Despite forming a functional syncytium, impulse propagation remained slow with conduction velocities (CVs) near 25 cm/s, which is 2- to 3-fold slower than normal propagation in adult hearts.6,7 The slow CV of hPSC-CM monolayers may be attributed to the immature electrophysiological phenotype of the hPSC-CMs generated to date.811 At present, this poses a major limitation to the utility of these cells for research and therapeutic purposes.1214
The extracellular matrix (ECM) plays an important role in stem cell fate decisions, normal development, and cardiogenesis.1517 A recent report demonstrated the critical role of the ECM for highly efficient cardiac-directed differentiation of hPSCs.17 In addition to providing structural support for the developing myocardium, the matrix contains important signaling molecules. Bioengineering techniques are currently being developed to create artificial structural matrices using biocompatible synthetic materials that have similar stiffness to the native matrix.1820 For instance, it was recently shown that ECM stiffness could modulate gene expression in neonatal cardiac myocytes.20 Interestingly, the softer ECM produced greater and more highly organized gap junctions (connexin43) than stiff surfaces. Here, we tested the hypothesis that hPSC-CM monolayers can be structurally and functionally matured rapidly by plating them on a soft silicone surface and investigated the role of integrin signaling in the maturation process.

Methods

iCell Human iPSC-CM Monolayers

Cryopreserved vials (liquid nitrogen) of iCell human cardiac myocytes were obtained from Cellular Dynamics International, Inc (Madison, WI). iCell cardiac myocytes are highly purified (>98%) hPSC-derived cells that are cryopreserved after 30 to 31 days of cardiac-directed differentiation. Cells were thawed and subsequently plated on bovine fibronectin-coated (20 μg/mL; Life Technologies, Thermo Fisher Scientific, Waltham, MA) glass coverslips, on bovine fibronectin-coated (20 μg/mL; Life Technologies) transparent polydimethylsiloxane (PDMS membranes, matrigel-coated (500 μg/mL; BD Biosciences, San Jose, CA) glass coverslips, or matrigel-coated (500 μg/mL; BD Biosciences) transparent PDMS membranes at a density of 125 000 cells per monolayer (1500 cardiomyocytes/mm2) in differentiation medium.5,10 In all cases, a 100-μL droplet of cells was plated on each ECM. Differentiation medium (EB20), consisting of 80% DMEM/F12, 0.1 mmol/L nonessential amino acids, 1 mmol/L l-glutamine, 0.1 mmol/L β-mercaptoethanol, 20% FBS, and 10 μmol/L blebbistatin, was used to culture the cells to promote attachment. Each ECM was added as a 100-μL drop of solution in the center of each coverslip; this routinely produced a 1-cm diameter circle of ECM on which the cardiomyocytes were plated after a 30-minute incubation period and aspiration of liquid. PDMS silicone sheeting was obtained from SMI (Specialty Manufacturing, Inc, Saginaw, MI) with 40D (D, Durometer or ≈1000 kPa) hardness and cut to 18×18 mm coverslips. After 24 hours, the media was switched to RPMI (Life Technologies) supplemented with B27 (Life Technologies). The cells were subsequently cultured for 4 to 7 days at 37°C, in 5% CO2 before phenotype analysis. The use of hPSCs and derived cardiomyocytes was approved by the Human Pluripotent Stem Cell Research Oversight (HPSCRO) Committee of the University of Michigan.

Optical Mapping

Optical action potentials (APs) in Figure 1 were recorded using FluoVolt membrane potential probe (F10488; Life Technologies). For calcium transient (CaT) and propagation experiments, monolayers were loaded with the intracellular Ca2+ indicator, rhod-2AM (10 μmol/L; Life Technologies) or fluo-4AM (5 μmol/L; Life Technologies) as indicated. After a 30-minute incubation time, the cells were washed in Hanks’ balanced salt solution (Life Technologies) for an additional 30 minutes before optical mapping recordings. All human cardiac monolayers displayed pacemaker activity, and the spontaneous APs or calcium waves were recorded using a charge-coupled device camera (200 fps, 80×80 pixels; Red-Shirt Little Joe, Scimeasure, Decatur, GA) with the appropriate emission filters and light-emitting diode illumination.21 Movies were filtered in both the time and the space domain, and CV was measured as described previously.22,23
Figure 1. Electric wave propagation in mature human pluripotent stem cell–derived cardiomyocyte monolayers. A, Left, optical activation map of spontaneously initiated electric wave propagation in an iCell cardiomyocyte monolayer cultured on polydimethylsiloxane (PDMS)+matrigel. Right, single pixel signals of optical action potentials recorded from the pacemaker site and a more distal site in the monolayer. We used a charge-coupled device camera and the voltage sensitive dye FluoVolt. B, Action potential impulse propagation velocity slowed as pacing frequency increased. The conduction velocities were as follows: 0.7 Hz= 42.4±2.3 cm/s, 1 Hz=36.4±1.5 cm/s, 1.2 Hz=33.8±0.7 cm/s, 1.5 Hz=29.2±0.8 cm/s, 1.8 Hz=27.0±0.71 cm/s, 2 Hz=25.9±0.9 cm/s, and 2.5 Hz=23.3±0.87 cm/s. C, Action potential duration calculated at 80% repolarization (APD80) shortened as pacing cycle length shortened. APD80 values were as follows: 0.7 Hz=629.9±16.6 ms, 1 Hz=569.2±14.9 ms, 1.2 Hz=511.2±14.4 ms, 1.5 Hz=452.7±9.8 ms, 1.8 Hz=398.1±6.9 ms, 2.0 Hz=363.2±3.3 ms, and 2.5 Hz=293.3±7.3 ms. n=6 monolayers for 0.7 to 1.8 Hz and n=4 for 2.0 and 2.5 Hz. Inset shows representative action potential recordings at different frequencies. All data are presented as mean±SEM.

Biochemical Analysis of hPSC-CM

Details of Western blotting, flow cytometry, immunofluorescence, and image analysis can be found in the expanded Methods in the Data Supplement.

Results

ECM Effects on iCell hPSC-CM Monolayer Impulse Propagation

In Figure 1 (left), we provide the first demonstration that purified iCell cardiomyocytes cultured as monolayers on PDMS+matrigel achieve a high degree of electric maturity, with average AP propagation velocities as high as 55 cm/s. It is important to note, however, that while iCell cardiomyocytes are highly purified; one always encounters mixtures of different cardiomyocyte phenotypes, including atrial-like, ventricular-like, and pacemaker-like myocytes.9,10,17 This is reflected in Figure 1A by the different optical AP configurations (right) in the monolayer. Pacemaker-like cells at the site of impulse initiation undergo slow diastolic depolarization at a steady rate until the threshold potential is reached and an AP is generated. More distally, ventricular-like APs have stable resting membrane potentials and respond to the propagating impulse with rapid upstrokes. In Figure 1B and 1C, we characterized AP propagation velocity and duration over electric pacing frequencies ranging from 0.7 to 2.5 Hz. Figure 1B shows CV restitution as one would expect with faster conduction at lower frequency (greater cycle length) of stimulation. Figure 1C demonstrates the AP duration restitution of mature hPSC-CM monolayers where AP duration gets shorter as pacing frequency increases. Thus, the biomatrix combination of matrigel+PDMS promotes functional electrophysiological maturation of hPSC-CMs in as little as a week.
To establish whether the softer ECM provided by PDMS+matrigel promoted maturation significantly more than stiffer matrices, we tested the combinations presented in Figure 2A by quantifying CaT propagation across the monolayer. First, the rate of spontaneous pacemaker activity was recorded for each of the 4 conditions. There was no difference in the spontaneous activation rate between the groups with the averages being the following: (i) fibronectin+glass=0.25±0.05 Hz, n=10; (ii) fibronectin+PDMS=0.22±0.03 Hz, n=4; (iii) matrigel+glass=0.24±0.09 Hz, n=14; and (iv) matrigel+PDMS=0.2±0.07 Hz, n=9 (mean±SEM; one-way ANOVA; P>0.9999). In Figure 2B, representative color CaT activation maps of wave propagation for each of the experimental conditions are shown. The fastest CV was observed in human cardiac monolayers cultured on matrigel+PDMS (Figure 2B, iv). The quantification of CV in each condition is shown in Figure 2C. The average CVs were as follows: fibronectin+glass=21.6±6.8 cm/s, n=10; fibronectin+PDMS=24.6±6 cm/s, n=4; matrigel+glass=22.0±4.0 cm/s, n=14; and matrigel+PDMS=43.6±7.0 cm/s, n=9 (mean±SEM; 1-way ANOVA; see figure and legend for details). Upper 95% confidence interval for the matrigel+PDMS group is 47.8 cm/s. Point stimulation of monolayers (15–20 V; 5-ms duration; 1 Hz) in each condition was performed in a separate group of experiments to determine differences in CV. The average CVs during 1 Hz pacing were as follows: (i) fibronectin+glass=24.2±1.8 cm/s, n=6; (ii) fibronectin+PDMS=28.1±1.5 cm/s, n=4; (iii) matrigel+glass=28.4±3.2 cm/s, n=5; and (iv) matrigel+PDMS=37.1±1.7 cm/s, n=6 (Figure 2D; mean±SEM). Thus, CV was faster during spontaneous pacemaker activations, as well as during 1 Hz electric pacing, in human cardiac monolayers cultured on matrigel-coated PDMS coverslips. In parallel experiments, iCell cardiomyocytes were plated on laminin or collagen purified from matrigel to test whether these individual components of matrigel would support confluent 2-dimensional (2D) monolayer maturation on PDMS. The results in Figure I in the Data Supplement show that monolayer confluence was not maintained for 7 days in culture on these purified components of matrigel. Optical mapping of AP propagation was continuous in each of the conditions tested (ie, laminin and collagen), suggesting that cellular reorganization was occurring rather than cell death. CV was fastest in monolayers plated on matrigel compared with those on collagen or laminin (Figure IC in the Data Supplement). Furthermore, cardiac troponin I (cTnI), the mature myofilament marker, was only induced when monolayers were plated on matrigel (Figure ID in the Data Supplement). Surface chemistry of cell culture surfaces can also impact on cellular phenotype,24 and the surface chemistry of glass is distinct from PDMS. Therefore, additional experiments were performed where glass coverslips were silanized to make their surface chemistry comparable with PDMS. Results of optical mapping 2D monolayers cultured on silanized glass indicate that the differences in surface chemistry cannot account for the rapid CV that we observed on PDMS (Figure II in the Data Supplement).
Figure 2. Effects of extracellular matrix (ECM) on human pluripotent stem cell–derived cardiomyocyte (hiPSC-CM) monolayer impulse propagation. A, Four different ECM combinations were tested to determine the effects on hPSC-CM monolayer structure and function. B, Activation maps of calcium impulse propagation in the different plating conditions. Each color represents a different activation time with time zero appearing in yellow. C, Quantification of impulse propagation. *Statistical difference where P<0.0001; †statistical difference where P=0.003 analyzed by 1-way ANOVA, Bonferroni multiple comparisons test, n=4 to 14 monolayers per group. D, Quantification of impulse propagation during electric stimulation at 1 Hz. ‡Difference where P=0.0003; §Difference where P=0.01 analyzed by 1-way ANOVA. All data are presented as mean±SEM. PDMS indicates polydimethylsiloxane.

ECM Effects on iCell hPSC-CM Monolayer AP and Single-Cell Electrophysiology

APs are required for propagation of the electric signal that triggers the Ca2+-mediated excitation–contraction coupling.10,13 Therefore, hPSC-CM monolayer APs were recorded and quantified by patch-clamp analysis in current-clamp mode. Properties of the AP, such as the dV/dtmax (V/s; Figure 3A and 3C), the maximum diastolic potential (MDP; Figure 3D), and the threshold potential (takeoff potential; Figure 3E), provide quantitative metrics of the degree of myocyte maturity.9,11,13 We measured these functional parameters in hPSC-CMs cultured on the extremely rigid ECM condition of fibronectin+glass (black bars; Figure 3) and compared the results with hPSC-CMs cultured on the softest ECM condition of matrigel+PDMS (red bars; Figure 3). APs recorded from hPSC-CMs grown on matrigel+PDMS displayed significantly faster upstroke velocities (65.3±8.9 V/s, N=6 monolayers, n=37, versus 146.5±17.7 V/s, N=5 monolayers, n=24; Figure 3C), more hyperpolarized MDPs (−69.9±1.7 mV, N=6 monolayers, n=37, versus −77.5±0.6 mV, N=5 monolayers, n=24; Figure 3D), and more hyperpolarized takeoff potentials (−59.3±1.7 mV, N=6 monolayers, n=37, versus −70.5±1.2 mV, N=5 monolayers, n=24; Figure 3E), all indicative of cardiomyocyte maturation.25
Figure 3. Mature human pluripotent stem cell–derived cardiomyocyte (hPSC-CM) action potential and sodium channel characteristics. A, Representative action potential recordings from monolayers plated on fibronectin on glass (left) and matrigel on polydimethylsiloxane (PDMS) (right). Middle panel shows a faster time scale of the action potential (AP) upstroke. Bottom panel shows the first derivative of the AP upstroke (dV/dt). B–E, AP parameters demonstrate significant electrophysiological maturation of monolayers plated on matrigel on PDMS (red). *Significant difference by the Student t test, P<0.05, N=5 monolayers–6 monolayers with n=24 or n=37 individual cellular recordings. F, Representative sodium current (INa) recordings of hPSC-CMs cultured on fibronectin on glass (black, n=12) and cardiomyocytes cultured on matrigel on PDMS (red, n=19). G, Current–voltage (I-V) relationship of sodium current in each condition shows elevated INa in cardiomyocytes cultured on matrigel-coated PDMS. Comparisons made by the Student t test, P values as indicated. H, Real-time–polymerase chain reaction (RT-PCR) analysis of SCN5a expression. *P<0.05, Student t test. All data are presented as mean±SEM. MDP indicates maximum diastolic potential.
The faster dV/dtmax and faster impulse propagation may be attributed partially to the effect of matrigel+PDMS ECM to increase sodium current (INa) density. Indeed, single-cell patch-clamp analysis of INa revealed significantly elevated current density in cardiomyocytes cultured on PDMS+matrigel compared with cardiomyocytes from the same batch cultured on fibronectin on glass coverslips (Figure 3F and 3G; glass N=4, n=12, and PDMS N=4, n=19). Elevated INa density observed here is consistent with previous data, showing that INa density increases in the maturing heart.26 Figure III in the Data Supplement shows the INa activation/inactivation profiles. The inactivation profile is left-shifted for cardiomyocytes cultured on PDMS+matrigel (V1/2=−79.0±2.45 mV, N=4, n=13, versus −88.0±1.55 mV, N=4, n=12; P=0.009), which suggests increased expression of cardiac sodium channel isoforms (ie, SCN5A and NaV1.5) that is known to be larger in adult than immature embryonic cardiac myocytes.26,27 Real-time–polymerase chain reaction analysis confirmed elevated SCN5A gene (Figure 3H) expression in iCell iPSC-CMs cultured on matrigel-coated PDMS compared with iPSC-CMs cultured on fibronectin-coated glass coverslips.
It has been reported by others that the MDP and spontaneous activity of hPSC-CMs are critically dependent on the density of the rapid component of the delayed rectifier potassium current (IKr) and that IK1 density is extremely low if not absent in hPSC-CMs.28 On the contrary, the hyperpolarized MDP of iCell cardiomyocytes cultured on PDMS+matrigel that we found here suggests that the IK1 density is relatively high. Indeed, patch-clamp analysis revealed elevated IK1 density in cardiomyocytes maintained on PDMS+matrigel biomatrix (Figure 4A and 4B). Western blotting shows expression of Kir2.1 exclusively in cardiomyocytes cultured on PDMS+matrigel compared with the same cells cultured on fibronectin-coated glass coverslips (Figure 4C). To further investigate whether PDMS+matrigel promotes the expression of other potassium currents, we tested the effects of E4031 (100 nmol/L), a selective blocker of IKr on the spontaneous beating rate and CaT duration 80 of hPSC-CM monolayers cultured on matrigel-coated PDMS compared with those cultured on standard rigid, plastic bottom 96-well dishes (also matrigel coated, Figure IV in the Data Supplement and Video I in the Data Supplement illustrate how this multiwell optical mapping platform was used). Small glass coverslips to fit into a 96-well format dish are not readily available, so here we used plastic as the rigid comparator for PDMS. Figure 4D and Figure IV in the Data Supplement each show that the electrophysiology of hPSC-CM monolayers cultured on PDMS, measured by the CaT duration, is less affected by IKr blockade, further supporting the evidence in Figure 4 of elevated inward rectifier potassium current density in hPSC-CMs cultured on matrigel-coated PDMS coverslips. For example, 100 nmol/L E4031 reduced spontaneous beating frequency by ≈50% (0.83±0.02 Hz pre-E4031 versus 0.45±0.05 Hz post-E4031; n=8) in monolayers plated on rigid plastic, whereas the same dose of E4031 modestly reduced beating frequency by ≈20% (1.02±0.04 Hz pre-E4031 versus 0.78±0.07 Hz post-E4031; n=5). This effect on beating frequency is likely because of the effect of E4031 to prolong the AP and CaT durations. Similarly, CaT duration 80 increased ≈3X after E4031 application in hPSC-CMs plated on plastic bottom dishes (605.9±24.3 ms pre-E4031 to 1873.8±278.9 ms post-E4031; n=8), but CaT duration 80 only increased by ≈1.34× in hPSC-CMs plated on PDMS bottom dishes (579.8±13.9 ms pre-E4031 versus 780.1±74.1 ms post-E4031; n=5; Figure IV in the Data Supplement).
Figure 4. Potassium current density (IK1) in human pluripotent stem cell–derived cardiomyocyte (hPSC-CM) single cells. A, Representative IK1 recordings in single hPSC-CM cultured on fibronectin on glass (n=7, black, top) and hPSC-CM cultured on matrigel on polydimethylsiloxane (PDMS) (n=5, red, bottom). B, Current–voltage (I-V) relationship of IK1 in each condition shows significantly elevated current density in hPSC-CM cultured on matrigel on PDMS (red). Comparison by Student t test, P value as indicated. C, Western blot probing for Kir2.1 demonstrates expression only in hPSC-CMs cultured on matrigel on PDMS (lane 1 in each blot, 2 individual monolayers for each condition). D, Intracellular calcium flux measurements in hPSC-CMs cultured on rigid plastic bottom dishes (matrigel coated) show significant impact of E4031 blockade on spontaneous beating frequency and calcium transient duration 80 (CaTD80). Quantification shows greater effect of E4031 on the beat frequency and CaTD80 in immature hPSC-CMs cultured on rigid plastic bottom dishes compared with PDMS bottom dishes. Unpaired t test, for effect on beat frequency: *P=0.000000001, †P=0.01; and for effect on CaTD80: ‡P=0.0000001, §P=0.0003; data expressed as mean±SEM.

ECM Effects on Intercellular Junction Formation

Gap Junctions: Connexin43 Expression

ECM stiffness has been shown to impact rodent neonatal myocyte connexin43 expression at the intercellular gap junctions. Previously, Forte et al20 found that softer substrates impact connexin43 expression and myocyte morphology. Consistent with previous reports using rodent myocytes, here we find that connexin43 expression is elevated in hPSC-CM monolayers cultured on PDMS coated with matrigel compared with monolayers cultured on glass coverslips (Figure 5) or fibronectin-coated PDMS. We determined the effect of the different plating combinations (Figure 2A) on connexin43 expression and subcellular localization. Figure 5A shows the connexin43 expression and localization (red) in hPSC-CM monolayers plated in the various ECM conditions. The greatest connexin43 expression at the intercellular junctions is found in monolayers plated on matrigel+PDMS. This provides yet another molecular mechanism to explain the faster CV found for this biomatrix combination. In Figure 5B, Western blot analysis shows that the amount of connexin43 expression was ≈3× greater when fibronectin+PDMS is compared with matrigel+PDMS (0.07±0.05 AU versus 0.19±0.10 AU; n=5 monolayers; P<0.05). Next, we determined the effect of PDMS to promote connexin43 expression in purified UM22-2 human embryonic stem cell–derived cardiomyocyte (hESC-CM) monolayers. The UM22-2 control hESC-CMs were generated by the matrix sandwich differentiation protocol.17 Immunostaining for α-actinin and connexin43 in hESC-CM monolayers also indicated robust induction of connexin43 expression and localization at the cell–cell borders by PDMS substrate (Figure VA in the Data Supplement). Similar to the iCell connexin43 expression, hESC-CM connexin43 expression outlines the entire cardiomyocytes when cells are cultured on PDMS. Collectively, these results demonstrate that the ECM combination of matrigel+PDMS promotes the development of functional gap junctions available for more efficient intercellular communication and faster impulse propagation in hPSC-CM monolayers.
Figure 5. Extracellular matrix effect on connexin43 (Cx43) expression and cardiomyocyte size. A, Immunostaining of α-actinin (green) and Cx43 (red). DAPI (4′,6-diamidino-2-phenylindole; blue) marks nuclei. Monolayers on matrigel-coated polydimethylsiloxane (PDMS) exhibit the greatest amount of Cx43 at the cell–cell borders. B, Western blotting for Cx43 and total myosin confirms the immunofluorescence results of (A). Quantification of total Cx43 protein expression shows elevated expression in human pluripotent stem cell–derived cardiomyocyte (hiPSC-CM) cultured on matrigel on PDMS (n=5 monolayers in each condition). *Student t test, P<0.05. All data are presented as mean±SEM. C and D, hPSC-CM (iCell) cell size was determined 5 days post thaw by immunofluorescent staining for N-cadherin. There was no difference in hPSC-CM size between the fibronectin+glass group and the matrigel+glass group (938.7±61.2 μm2, n=79 vs 917.8±64.2 μm2; n=66). Myocytes plated on fibronectin+PDMS were larger (1403.4±66.9 μm2; n=132) and myocytes plated on matrigel+PDMS were even larger (2130.3±99.9 μm2; n=130) than those plated on rigid glass coverslips. One-way ANOVA, Bonferroni multiple comparison test; †P<0.0001 and ‡P<0.001.

Terminal Differentiation, Hypertrophic Growth, and cTnI Expression

After birth, myocytes in the heart switch from hyperplastic growth to hypertrophic growth, and this is part of the natural maturation process.11,29,30 Therefore, another marker of maturation of hPSC-CMs is the transition from cardiomyocytes remaining in the cell cycle to myocyte terminal differentiation and hypertrophy. The hPSC-CMs cultured on pliable PDMS were significantly larger in size than cells plated on glass substrates (Figure 5C and 5D). This indicates induction of developmental hypertrophy in hPSC-CMs cultured on soft substrates. Binucleation, another marker of myocyte maturity,30,31 was also apparent in hPSC-CMs cultured on matrigel+PDMS (Figure 6A and 6B; Figure VI in the Data Supplement). The cell cycle marker Ki67 was also used to determine the differentiation state of cardiomyocytes.32 In iCell cardiomyocytes, the percentage of binucleated cells that were also Ki67+ was lower in iPSC-CMs cultured on PDMS+matrigel (Figure 6B). Furthermore, using highly purified BJ-hPSC-CMs developed in our laboratory33(Figure 5B), we quantified the effect of PDMS to reduce the number of cardiomyocytes remaining in the cell cycle (Figure 6C). One key myofilament marker of cardiomyocyte maturation is cTnI expression.11,34 We took this parameter of maturation into consideration here by Western blot analysis of BJ-hPSC-CM expression of cTnI when purified monolayers were cultured on glass or PDMS coverslips coated with matrigel. Significantly more robust cTnI expression was detected relative to GAPDH in purified BJ-hPSC-CMs plated on PDMS coverslips compared with glass coverslips (Figure 6D), thus indicating a greater level of sarcomeric maturation and promotion of developmental isoform switching that is known to occur in the postnatal heart. Furthermore, cTnI sarcomeric incorporation was also apparent in hPSC-CM monolayers cultured on PDMS but not in immature monolayers cultured on Glass coverslips (Figure VC in the Data Supplement).
Figure 6. Molecular markers of maturation. A, Flow cytometry analysis for quantification of the population of binucleated iCell cardiomyocytes. B, The proportion of binucleated cells was significantly greater in the polydimethylsiloxane (PDMS) group compared with matrigel-coated glass (35.24±3.0% vs 21.68±1.08%; n=5 per group; *significant difference; t test; P=0.003). Furthermore, the incidence of Ki67-positive binucleated cells was less in the PDMS group (13.4±1.1% vs 20.5±1.1%; n=5 per group; †significant difference; t test; P=0.002). C (top), Immunostaining for Ki67 (red) and α-actinin (green) shows decreased proliferative activity in BJ-human pluripotent stem cell–derived cardiomyocytes (hPSC-CMs) cultured on PDMS compared with glass coverslips (0.87±0.29 CM/60× field compared with 6.2±0.90 CM/60× field; n=8 and n=10; ‡P=0.0001). C (bottom), Immunostaining for sarcomeric actin (red) and N-cadherin (green) shows hypertrophy and elongation of BJ-iPSC-CMs cultured on PDMS compared with glass coverslips (cell area=3678.59±171.6 μm2 compared with 2071.44±116.7 μm2; n=84 and n=83; §P=0.000003). D, Western blotting for cardiac troponin I (cTnI) protein expression. On glass coverslips, the cTnI/GAPDH ratio=0.39±0.11 AU, and on PDMS coverslips the cTnI/GAPDH ratio=0.6464±0.005 AU; ‖significant difference, P<0.05, unpaired t test. All data mean±SEM.

Integrin Signaling in the Maturation Process

Integrins are transmembrane heterodimeric receptors essential for providing cell-ECM adhesion, cellular structural organization, and transduction of mechanical signals from the ECM into biochemical signals in cardiomyocytes.3537 β1 integrins are abundant in the adult heart and participate in the hypertophic response in rodent ventricular myoyctes.38 Therefore, we hypothesized that integrin signaling underlies the maturation of hPSC-CM monolayers induced by the cell culture condition of plating them on matrigel-coated PDMS coverslips. First, real-time–polymerase chain reaction analysis showed that ITGB1 expression is significantly induced on PDMS coverslips compared with glass coverslips (Figure 7A). In addition, PTK2 gene expression is elevated in hPSC-CM monolayers cultured on PDMS coverslips (Figure 7B). The PTK2 gene encodes the focal adhesion kinase (FAK) intracellular molecule that is a primary mediator of integrin signaling.39 Figure 7C shows the localization of β1 integrin receptors in monolayers; there was elevated expression in monolayers cultured on PDMS.
Figure 7. Integrin signaling via focal adhesion kinase promotes maturation of human pluripotent stem cell–derived cardiomyocyte (hiPSC-CM) monolayers. A, Real-time–polymerase chain reaction (RT-PCR) analysis indicates elevated expression of ITGA5 and ITGB1 integrin receptor genes in mature monolayers (red). RT-PCR performed in triplicate for 5 individual monolayers for each group. B, PTK2 gene expression is elevated in mature monolayers (red). C, β1 integrin localization (red) in hPSC-CM monolayers. More extensive receptor expression is apparent on polydimethylsiloxane (PDMS). D, Nab (neutralizing antibody) again β1 integrin receptors block focal adhesion kinase (FAK) activation and cardiac troponin I (cTnI) expression in iCell CMs. *Significant difference, t test, P<0.05; †t test, P=0.02; n=3 monolayers per group. E, Pharmacological inhibition of FAK activity using FAK inhibitor-14 prevents cTnI protein expression. F, FAK inhibition prevents PDMS induced hypertrophic growth of hPSC-CMs. Cell areas: PDMS control=4410.8±217.3 μm2; 10 μmol/L=2547.7±104.7 μm2; 100 μmol/L=1057.1±59.0 μm2). ‡Significant difference from GLASS (see Figure 7 in the Data Supplement), §significant difference within PDMS group compared with control media, 1-way ANOVA, P<0.0001.
Next, we blocked integrin signaling by 2 different approaches: (1) neutralization of β1 integrin receptor activation using isoform-specific antibody, and (2) pharmacological blockade of FAK activation using FAK inhibitor-14. Importantly, lower dose of FAK inhibitor-14 did not reduce hPSC-CM monolayer confluence significantly (figure VII in the Data Supplement).Western blotting in Figure 7D indicates that FAK activation was prevented by Nab (neutralizing antibody) and also that cTnI protein expression was attenuated. Collectively this indicates that FAK activation via β1 integrin activation contributes to the expression of mature myofilament markers, such as cTnI. Furthermore, purified hPSC-CMs (iCell) cultured on PDMS in the presence of 10 μmol/L FAK inhibitor-14 failed to express cTnI and also expressed less β-MyHC (β-myosin heavy chain) (Figure 7E; Figure VIII in the Data Supplement). This suggests that FAK activation underlies expression of mature myofilament markers. In addition, we determined the role of FAK activation in the hypertrophic response of hPSC-CMs to the soft cell culture environment on PDMS (Figure 7F; Figure VII in the Data Supplement). FAK inhibitor-14 prevented hypertrophic growth of hPSC-CMs observed on PDMS coverslips. Interestingly, hPSC-CMs grown on glass coverslips were only affected at the higher dose of FAK inhibitor-14 (100 μmol/L). In both conditions (glass and PDMS), 100 μmol/L FAK inhibitor-14 reduced monolayer confluence and hPSC-CM expansion in culture (Figures VII and VIII in the Data Supplement). Finally, the effects of integrin receptor activation were next studied using a monoclonal antibody that stabilizes the dimerization of α5β1 integrins and promotes FAK activation (MAB1969). Figure 8 summarizes the Western blot analysis of hPSC-CM monolayers (iCell) treated with the activating α5β1 integrin monoclonal antibody or FAK inhibitor-14. FAK activation (phosphorylation) was induced by this specific antibody (Figure 8B) and so was cardiac sodium channel protein expression (Nav1.5; Figure 8C).
Figure 8. Integrin activation promotes Nav1.5 sodium channel expression. A, Treatment of human pluripotent stem cell–derived cardiomyocyte (hiPSC-CM) monolayers chronically with a mouse monoclonal antibody for the α5β1 integrin receptor heterodimer (fibronectin receptor) causes increase in total focal adhesion kinase (FAK) expression and activation (B). C, Sodium channel (Nav1.5) expression is also induced by integrin activation via α5β1 integrin receptor antibody treatment. Data are expressed as whiskers plots (mean, maximum and minimum values are shown), ANOVA was used to test for significance with *P=0.016 (A), †P=0.001 (B), ‡P=0.003 (C) to indicate difference.

Discussion

We have demonstrated the following in hPSC-CM 2D monolayers: First, culturing the monolayers on a soft PDMS membrane (40D, durometers) coated with matrigel but not fibronectin alone, increases the impulse CV to values ≤48 cm/s, which is 2× faster than previously reported for human iPSC-CM monolayers.5 Second, PDMS+matrigel ECM promotes electrophysiological maturation of the hPSC-CM single cell characterized by increased inward rectifier potassium and sodium inward current densities, giving rise to a well-polarized MDP and faster AP upstroke velocity, respectively. Third, formation of intercellular gap junctions and mechanical junctions is promoted by the soft PDMS+matrigel ECM. Fourth, hPSC-CM hypertrophy and mature myofilament isoform expression are induced when plated on pliable PDMS rather than on rigid glass coverslips. Fifth, integrin β1 and α5 receptor gene expression is induced in the maturation process, and FAK activity is required for maturation. Remarkably, the electrophysiological and structural maturation of iCell cardiomyocytes plated on the optimal biomatrix combination as reported here occurs in as little as 1 week after replating cryopreserved cardiomyocytes. This represents a major advance over previous reports that have demonstrated modest maturation of stem cell–derived cardiomyocytes during a period of ≤9 months.34,40,41 Importantly, the process described here does not require gene transfer or other genetic modifications that may artificially mimic the developmental process. Furthermore, we have validated the maturation observed on PDMS in cardiomyocytes derived from multiple hPSC lines, including other hPSC lines and one hESC line.
Mounting evidence has made it apparent that combining matrigel with softer synthetic biomaterials is a way to produce more mature hPSC and ESC-derived human cardiomyocytes that will be more useful for disease modeling, drug testing, and cardiac regeneration applications.42,43 This study is unique in that we have studied maturation of electrically and mechanically connected human stem cell–derived cardiac monolayers that function as a syncytium. Importantly, we show that confluent monolayer culture promotes more electrophysiological maturation than single-cell culture approaches on soft matrigel substrates. In 2005, Baharvand et al44 reported the importance of using either a native cardiac ECM called cardiogel or a matrigel when culturing mouse ESC-derived cardiomyocytes. In that study, Baharvand et al reported modest maturation of murine cardiomyocytes cultured on both cardiogel and matrigel ECM and also showed that cardiomyocytes grown on matrigel respond in a more mature cardiomyocyte manner to carbachol administration. Thus, it seems that the complex composition of matrigel ECM consisting of collagen, laminin, fibronectin, and other components may provide a better maturation ECM than the slightly more defined native cardiac ECM. Despite the higher level of maturation that we have reported here using matrigel-coated PDMS coverslips, the use of matrigel does impose limitations. Matrigel is derived from a mouse Engelbreth-Holm-Swarm sarcoma cell line.45 It contains not only a random array of ECM proteins but also a variety of growth factors. Thus, it is ill defined and may vary from batch to batch. Despite this concern, the maturation process reported here has been repeatable using over a dozen different batches of matrigel.
Previous studies have used 3D cell culture techniques to mature human stem cell–derived cardiomyocytes. For example, Nunes et al8 attempted to mature human stem cell–derived cardiomyocytes by creating a 3D biowire platform. Although they reported modest maturation and cardiomyocyte alignment, CV of those biowires was slow, with the most mature constructs having a CV of only ≈15 cm/s.8 Here, we have generated mature 2D hPSC-CM monolayers with CVs of ≤55 cm/s and immature monolayers with impulse propagation velocity of ≈25 cm/s, both of which are faster and more mature than in the biowire platform. Furthermore, here we have shown maturation in single hPSC-CM ion channel densities (IK1, INa), ion channel expression (Kir2.1, SCN5A), AP profiles (rapid dV/dt, hyperpolarized diastolic potentials and threshold potentials), and structural maturation in monolayers (cTnI expression, connexin43 expression, and hypertrophy). In particular, Figure 5 demonstrates that immature hPSC-CM monolayers depend almost entirely on IKr repolarizing current, and this is congruent with previous reports.28 However, mature hPSC-CM monolayers cultured on PDMS express both Kir2.1 channels and robust IK1 density (Figure 4) and respond to the IKr blocker E4031 in a more modest manner that resembles responses of adult myocardium (Figure 4).
Importantly, our findings demonstrate the ability of hPSC-CMs to terminally differentiate, form gap junctions, and undergo myofilament isoform switches that may promote greater tension development and responsiveness to autonomic input. This may have impact on the development of autologous cardiac regeneration approaches using hPSC-CMs. Terminal differentiation of hPSC-CMs is indicated by binucleation and significant reduction of Ki67-positive cardiomyocyte nuclei (Figure 6). This suggests that hPSC-CMs have the capacity to exit the cell cycle in the right environment and may not be tumorigenic when used for cardiac regeneration therapies. There is also a significant induction of connexin43 containing gap junctions when hPSC-CMs are put into the optimal maturation environment (Figure 5). Induction of connexin43 expression suggests that hPSC-CMs have the capacity to electrically integrate into native myocardium if used for the development of autologous cardiac regeneration therapies. We demonstrate robust induction of cTnI in mature hPSC-CMs (Figure 6) and that expression of mature myofilament proteins is dependent on β1 integrin receptor activation and FAK activity (Figure 7). Thus, when in the proper environment, hPSC-CMs have the capacity for myofibrillar isoform switching including expression of cTnI, a critical sarcomeric component that modulates cardiac contractility on a beat–beat basis.46,47
Integrin signaling is a dynamic and complex process in cardiomyocytes depending on the ECM components and mechanical activity.48 The expression and dimerization of integrin receptors can be triggered by specific extracellular protein components and specific ECM proteins (eg, fibronectin versus laminin) to impact cardiomyocyte gene expression and function. Systematic study of integrin isoforms and dimers will be required to determine the optimal ECM for maturation of PSC-CMs. Our results indicate the capacity of hPSC and hESC-CMs to mature in vitro; however, for cell transplantation, the optimal state of maturation to use for cardiac regeneration that will lead to the best outcomes requires further investigation.

Supplemental Material

File (circae_circae-2015-003638-t_supp1.pdf)
File (circae_circae-2015-003638-t_supp2.avi)

References

1.
Anson BD, Kolaja KL, Kamp TJ. Opportunities for use of human iPS cells in predictive toxicology. Clin Pharmacol Ther. 2011;89:754–758. doi: 10.1038/clpt.2011.9.
2.
Kamp TJ, Lyons GE. On the road to iPS cell cardiovascular applications. Circ Res. 2009;105:617–619. doi: 10.1161/CIRCRESAHA.109.205740.
3.
Itzhaki I, Maizels L, Huber I, Gepstein A, Arbel G, Caspi O, Miller L, Belhassen B, Nof E, Glikson M, Gepstein L. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll Cardiol. 2012;60:990–1000. doi: 10.1016/j.jacc.2012.02.066.
4.
Malan D, Friedrichs S, Fleischmann BK, Sasse P. Cardiomyocytes obtained from induced pluripotent stem cells with long-QT syndrome 3 recapitulate typical disease-specific features in vitro. Circ Res. 2011;109:841–847. doi: 10.1161/CIRCRESAHA.111.243139.
5.
Lee P, Klos M, Bollensdorff C, Hou L, Ewart P, Kamp TJ, Zhang J, Bizy A, Guerrero-Serna G, Kohl P, Jalife J, Herron TJ. Simultaneous voltage and calcium mapping of genetically purified human induced pluripotent stem cell-derived cardiac myocyte monolayers. Circ Res. 2012;110:1556–1563. doi: 10.1161/CIRCRESAHA.111.262535.
6.
Noujaim SF, Kaur K, Milstein M, Jones JM, Furspan P, Jiang D, Auerbach DS, Herron T, Meisler MH, Jalife J. A null mutation of the neuronal sodium channel NaV1.6 disrupts action potential propagation and excitation-contraction coupling in the mouse heart. FASEB J. 2012;26:63–72. doi: 10.1096/fj.10-179770.
7.
David M., Harrild CSH. A computer model of normal conduction in the human atria. Circ Res. 2000;87:e25–e36.
8.
Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y, Zhang B, Jiang J, Massé S, Gagliardi M, Hsieh A, Thavandiran N, Laflamme MA, Nanthakumar K, Gross GJ, Backx PH, Keller G, Radisic M. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat Methods. 2013;10:781–787. doi: 10.1038/nmeth.2524.
9.
Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ, Kolaja KL, Swanson BJ, January CT. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am J Physiol Heart Circ Physiol. 2011;301:H2006–H2017. doi: 10.1152/ajpheart.00694.2011.
10.
Zhang J, Wilson GF, Soerens AG, Koonce CH, Yu J, Palecek SP, Thomson JA, Kamp TJ. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009;104:e30–e41. doi: 10.1161/CIRCRESAHA.108.192237.
11.
Yang X, Pabon L, Murry CE. Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Res. 2014;114:511–523. doi: 10.1161/CIRCRESAHA.114.300558.
12.
Priori SG. Induced pluripotent stem cell-derived cardiomyocytes and long QT syndrome: is personalized medicine ready for prime time? Circ Res. 2011;109:822–824. doi: 10.1161/CIRCRESAHA.111.253724.
13.
Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ Res. 2012;111:344–358. doi: 10.1161/CIRCRESAHA.110.227512.
14.
Knollmann BC. Induced pluripotent stem cell-derived cardiomyocytes: boutique science or valuable arrhythmia model? Circ Res. 2013;112:969–76; discussion 976. doi: 10.1161/CIRCRESAHA.112.300567.
15.
Guilak F, Cohen DM, Estes BT, Gimble JM, Liedtke W, Chen CS. Control of stem cell fate by physical interactions with the extracellular matrix. Cell Stem Cell. 2009;5:17–26. doi: 10.1016/j.stem.2009.06.016.
16.
Rozario T, DeSimone DW. The extracellular matrix in development and morphogenesis: a dynamic view. Dev Biol. 2010;341:126–140. doi: 10.1016/j.ydbio.2009.10.026.
17.
Zhang J, Klos M, Wilson GF, Herman AM, Lian X, Raval KK, Barron MR, Hou L, Soerens AG, Yu J, Palecek SP, Lyons GE, Thomson JA, Herron TJ, Jalife J, Kamp TJ. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method. Circ Res. 2012;111:1125–1136. doi: 10.1161/CIRCRESAHA.112.273144.
18.
Liau B, Zhang D, Bursac N. Functional cardiac tissue engineering. Regen Med. 2012;7:187–206. doi: 10.2217/rme.11.122.
19.
McCain ML, Lee H, Aratyn-Schaus Y, Kléber AG, Parker KK. Cooperative coupling of cell-matrix and cell-cell adhesions in cardiac muscle. Proc Natl Acad Sci U S A. 2012;109:9881–9886. doi: 10.1073/pnas.1203007109.
20.
Forte G, Pagliari S, Ebara M, Uto K, Tam JK, Romanazzo S, Escobedo-Lucea C, Romano E, Di Nardo P, Traversa E, Aoyagi T. Substrate stiffness modulates gene expression and phenotype in neonatal cardiomyocytes in vitro. Tissue Eng Part A. 2012;18:1837–1848. doi: 10.1089/ten.TEA.2011.0707.
21.
Lee P, Bollensdorff C, Quinn TA, Wuskell JP, Loew LM, Kohl P. Single-sensor system for spatially resolved, continuous, and multiparametric optical mapping of cardiac tissue. Heart Rhythm. 2011;8:1482–1491. doi: 10.1016/j.hrthm.2011.03.061.
22.
Hou L, Deo M, Furspan P, Pandit SV, Mironov S, Auerbach DS, Gong Q, Zhou Z, Berenfeld O, Jalife J. A major role for HERG in determining frequency of reentry in neonatal rat ventricular myocyte monolayer. Circ Res. 2010;107:1503–1511. doi: 10.1161/CIRCRESAHA.110.232470.
23.
Campbell K, Calvo CJ, Mironov S, Herron T, Berenfeld O, Jalife J. Spatial gradients in action potential duration created by regional magnetofection of hERG are a substrate for wavebreak and turbulent propagation in cardiomyocyte monolayers. J Physiol. 2012;590:6363–6379. doi: 10.1113/jphysiol.2012.238758.
24.
Trantidou T, Rao C, Barrett H, Camelliti P, Pinto K, Yacoub MH, Athanasiou T, Toumazou C, Terracciano CM, Prodromakis T. Selective hydrophilic modification of Parylene C films: a new approach to cell micro-patterning for synthetic biology applications. Biofabrication. 2014;6:025004. doi: 10.1088/1758-5082/6/2/025004.
25.
Robertson C, Tran DD, George SC. Concise review: maturation phases of human pluripotent stem cell-derived cardiomyocytes. Stem Cells. 2013;31:829–837. doi: 10.1002/stem.1331.
26.
Davies MP, An RH, Doevendans P, Kubalak S, Chien KR, Kass RS. Developmental changes in ionic channel activity in the embryonic murine heart. Circ Res. 1996;78:15–25.
27.
Domínguez JN, de la Rosa A, Navarro F, Franco D, Aránega AE. Tissue distribution and subcellular localization of the cardiac sodium channel during mouse heart development. Cardiovasc Res. 2008;78:45–52. doi: 10.1093/cvr/cvm118.
28.
Doss MX, Di Diego JM, Goodrow RJ, Wu Y, Cordeiro JM, Nesterenko VV, Barajas-Martínez H, Hu D, Urrutia J, Desai M, Treat JA, Sachinidis A, Antzelevitch C. Maximum diastolic potential of human induced pluripotent stem cell-derived cardiomyocytes depends critically on I(Kr). PLoS One. 2012;7:e40288. doi: 10.1371/journal.pone.0040288.
29.
Laflamme MA, Murry CE. Heart regeneration. Nature. 2011;473:326–335. doi: 10.1038/nature10147.
30.
Li F, Wang X, Capasso JM, Gerdes AM. Rapid transition of cardiac myocytes from hyperplasia to hypertrophy during postnatal development. J Mol Cell Cardiol. 1996;28:1737–1746. doi: 10.1006/jmcc.1996.0163.
31.
Katzberg AA, Farmer BB, Harris RA. The predominance of binucleation in isolated rat heart myocytes. Am J Anat. 1977;149:489–499. doi: 10.1002/aja.1001490406.
32.
Walsh S, Pontén A, Fleischmann BK, Jovinge S. Cardiomyocyte cell cycle control and growth estimation in vivo–an analysis based on cardiomyocyte nuclei. Cardiovasc Res. 2010;86:365–373. doi: 10.1093/cvr/cvq005.
33.
Bizy A, Guerrero-Serna G, Hu B, Ponce-Balbuena D, Willis BC, Zarzoso M, Ramirez RJ, Sener MF, Mundada LV, Klos M, Devaney EJ, Vikstrom KL, Herron TJ, Jalife J. Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes. Stem Cell Res. 2013;11:1335–1347. doi: 10.1016/j.scr.2013.09.003.
34.
Bedada Fikru B, Chan Sunny S-K, Metzger Stefania K, Zhang L, Zhang J, Garry Daniel J, Kamp Timothy J, Kyba M, Metzger Joseph M. Acquisition of a quantitative, stoichiometrically conserved ratiometric marker of maturation status in stem cell-derived cardiac myocytes. Stem Cell Reports. 2014;3:594–605.
35.
Ross RS, Borg TK. Integrins and the myocardium. Circ Res. 2001;88:1112–1119.
36.
Israeli-Rosenberg S, Chen C, Li R, Deussen DN, Niesman IR, Okada H, Patel HH, Roth DM, Ross RS. Caveolin modulates integrin function and mechanical activation in the cardiomyocyte. FASEB J. 2015;29:374–384. doi: 10.1096/fj.13-243139.
37.
Israeli-Rosenberg S, Manso AM, Okada H, Ross RS. Integrins and integrin-associated proteins in the cardiac myocyte. Circ Res. 2014;114:572–586. doi: 10.1161/CIRCRESAHA.114.301275.
38.
Ross RS, Pham C, Shai SY, Goldhaber JI, Fenczik C, Glembotski CC, Ginsberg MH, Loftus JC. Beta1 integrins participate in the hypertrophic response of rat ventricular myocytes. Circ Res. 1998;82:1160–1172.
39.
Cheng Z, DiMichele LA, Rojas M, Vaziri C, Mack CP, Taylor JM. Focal adhesion kinase antagonizes doxorubicin cardiotoxicity via p21(Cip1.). J Mol Cell Cardiol. 2014;67:1–11. doi: 10.1016/j.yjmcc.2013.12.002.
40.
Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 2013;22:1991–2002. doi: 10.1089/scd.2012.0490.
41.
Sartiani L, Bettiol E, Stillitano F, Mugelli A, Cerbai E, Jaconi ME. Developmental changes in cardiomyocytes differentiated from human embryonic stem cells: a molecular and electrophysiological approach. Stem Cells. 2007;25:1136–1144. doi: 10.1634/stemcells.2006-0466.
42.
Feaster TK, Cadar AG, Wang L, Williams CH, Chun YW, Hempel JE, Bloodworth N, Merryman WD, Lim CC, Wu JC, Knollmann BC, Hong CC. Matrigel mattress: a method for the generation of single contracting human-induced pluripotent stem cell-derived cardiomyocytes. Circ Res. 2015;117:995–1000. doi: 10.1161/CIRCRESAHA.115.307580.
43.
Ribeiro AJ, Ang YS, Fu JD, Rivas RN, Mohamed TM, Higgs GC, Srivastava D, Pruitt BL. Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. Proc Natl Acad Sci U S A. 2015;112:12705–12710. doi: 10.1073/pnas.1508073112.
44.
Baharvand H, Azarnia M, Parivar K, Ashtiani SK. The effect of extracellular matrix on embryonic stem cell-derived cardiomyocytes. J Mol Cell Cardiol. 2005;38:495–503. doi: 10.1016/j.yjmcc.2004.12.011.
45.
Lee GY, Kenny PA, Lee EH, Bissell MJ. Three-dimensional culture models of normal and malignant breast epithelial cells. Nat Methods. 2007;4:359–365. doi: 10.1038/nmeth1015.
46.
Herron TJ, Korte FS, McDonald KS. Power output is increased after phosphorylation of myofibrillar proteins in rat skinned cardiac myocytes. Circ Res. 2001;89:1184–1190.
47.
Kentish JC, McCloskey DT, Layland J, Palmer S, Leiden JM, Martin AF, Solaro RJ. Phosphorylation of troponin I by protein kinase A accelerates relaxation and crossbridge cycle kinetics in mouse ventricular muscle. Circ Res. 2001;88:1059–1065.
48.
Traister A, Li M, Aafaqi S, Lu M, Arab S, Radisic M, Gross G, Guido F, Sherret J, Verma S, Slorach C, Mertens L, Hui W, Roy A, Delgado-Olguín P, Hannigan G, Maynes JT, Coles JG. Integrin-linked kinase mediates force transduction in cardiomyocytes by modulating SERCA2a/PLN function. Nat Commun. 2014;5:4533. doi: 10.1038/ncomms5533.

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation: Arrhythmia and Electrophysiology
Go to Circulation: Arrhythmia and Electrophysiology
Circulation: Arrhythmia and Electrophysiology
PubMed: 27069088

History

Received: 5 May 2013
Accepted: 16 March 2016
Published in print: April 2016
Published online: 11 April 2016

Permissions

Request permissions for this article.

Keywords

  1. cardiac electrophysiology
  2. cell culture techniques
  3. hardness
  4. induced pluripotent stem cells
  5. integrin alpha5beta1
  6. myocytes, cardiac
  7. troponin I

Subjects

Authors

Affiliations

Todd J. Herron, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Andre Monteiro Da Rocha, DVM, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Katherine F. Campbell, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Daniela Ponce-Balbuena, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
B. Cicero Willis, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Guadalupe Guerrero-Serna, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Qinghua Liu, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Matt Klos, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Hassan Musa, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Manuel Zarzoso, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Alexandra Bizy, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Jamie Furness, BS
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Justus Anumonwo, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
Sergey Mironov, PhD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).
José Jalife, MD
From the Center for Arrhythmia Research, Department of Internal Medicine, Cardiovascular Research Center, University of Michigan, Ann Arbor, MI (T.J.H., A.M.D.R., K.C., D.P.-B., B.C.W., G.G.-S., Q.L., J.F., J.A., S.M., J.J.); Department of Medicine, University of California San Diego, La Jolla, CA (M.K.); Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH (H.M.); Shanxi Medical University, Zhejiang, China (Q.L.); and University of Valencia, Valencia, Spain (M.Z., A.B.).

Notes

Correspondence to Todd J. Herron, PhD, Center for Arrhythmia Research, 2800 Plymouth Road, Bldg 26, 2L01N, Ann Arbor, MI 48109. E-mail [email protected]

Disclosures

None.

Sources of Funding

Cellular Dynamics International (CDI) Innovative Research Grant (Dr Herron), National Institutes of Health grants P01-HL039707 and P01-HL087226 (Dr Jalife), the Leducq Foundation (Dr Jalife), the Lefkofsky Family Foundation (Dr Herron), and the State of Michigan Economic Development Fund (U-M Michigan Translational Research and Commercialization for Life Sciences Program [U-M MTRAC], Drs Herron and Jalife) all contributed to this work.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. iPSC-cardiomyocytes in the preclinical prediction of candidate pharmaceutical toxicity, Frontiers in Pharmacology, 15, (2024).https://doi.org/10.3389/fphar.2024.1308217
    Crossref
  2. hiPSC-derived cardiomyocytes as a model to study the role of small-conductance Ca2+-activated K+ (SK) ion channel variants associated with atrial fibrillation, Frontiers in Cell and Developmental Biology, 12, (2024).https://doi.org/10.3389/fcell.2024.1298007
    Crossref
  3. Maturation of pluripotent stem cell-derived cardiomyocytes: limitations and challenges from metabolic aspects, Stem Cell Research & Therapy, 15, 1, (2024).https://doi.org/10.1186/s13287-024-03961-4
    Crossref
  4. hiPSC-CM electrophysiology: impact of temporal changes and study parameters on experimental reproducibility, American Journal of Physiology-Heart and Circulatory Physiology, 327, 1, (H12-H27), (2024).https://doi.org/10.1152/ajpheart.00631.2023
    Crossref
  5. Engineered cocultures of iPSC-derived atrial cardiomyocytes and atrial fibroblasts for modeling atrial fibrillation, Science Advances, 10, 3, (2024).https://doi.org/10.1126/sciadv.adg1222
    Crossref
  6. Structure-Activity Relationship and Voltage Dependence for the Drug–Drug Interaction between Amiodarone Analogs and MNI-1 at the L-type Cav Channel, Journal of Pharmacology and Experimental Therapeutics, 389, 2, (229-242), (2024).https://doi.org/10.1124/jpet.123.001858
    Crossref
  7. Human sodium current voltage‐dependence at physiological temperature measured by coupling a patch‐clamp experiment to a mathematical model, The Journal of Physiology, 602, 4, (633-661), (2024).https://doi.org/10.1113/JP285162
    Crossref
  8. Stem cell‐derived cardiomyocyte heterogeneity confounds electrophysiological insights, The Journal of Physiology, (2024).https://doi.org/10.1113/JP284618
    Crossref
  9. Histone demethylase KDM5 regulates cardiomyocyte maturation by promoting fatty acid oxidation, oxidative phosphorylation, and myofibrillar organization, Cardiovascular Research, 120, 6, (630-643), (2024).https://doi.org/10.1093/cvr/cvae014
    Crossref
  10. Engineered heart tissue: Design considerations and the state of the art, Biophysics Reviews, 5, 2, (2024).https://doi.org/10.1063/5.0202724
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Extracellular Matrix–Mediated Maturation of Human Pluripotent Stem Cell–Derived Cardiac Monolayer Structure and Electrophysiological Function
Circulation: Arrhythmia and Electrophysiology
  • Vol. 9
  • No. 4

Purchase access to this journal for 24 hours

Circulation: Arrhythmia and Electrophysiology
  • Vol. 9
  • No. 4
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Share article link

Share

Comment Response