Publications

2014
59 -, Mosadegh B, Dabiri BE, Lockett MR, Derda R, Campbell P, Parker KK, and Whitesides GM. 2014. “Three-Dimensional Paper-Based Model for Cardiac Ischemia.” Advanced Healthcare Materials, 3, 7, Pp. 1036-43. Publisher's VersionAbstract
In vitro models of ischemia have not historically recapitulated the cellular interactions and gradients of molecules that occur in a 3D tissue. This work demonstrates a paper-based 3D culture system that mimics some of the interactions that occur among populations of cells in the heart during ischemia. Multiple layers of paper containing cells, suspended in hydrogels, are stacked to form a layered 3D model of a tissue. Mass transport of oxygen and glucose into this 3D system can be modulated to induce an ischemic environment in the bottom layers of the stack. This ischemic stress induces cardiomyocytes at the bottom of the stack to secrete chemokines which subsequently trigger fibroblasts residing in adjacent layers to migrate toward the ischemic region. This work demonstrates the usefulness of patterned, stacked paper for performing in vitro mechanistic studies of cellular motility and viability within a model of the laminar ventricle tissue of the heart.
Three-Dimensional Paper-Based Model for Cardiac Ischemia
2013
58 -, Nawroth JC, and Parker KK. 2013. “Design standards for engineered tissues.” Biotechnology Advances, 31, 5, Pp. 632-7. Publisher's VersionAbstract
Traditional technologies are required to meet specific, quantitative standards of safety and performance. In tissue engineering, similar standards will have to be developed to enable routine clinical use and customized tissue fabrication. In this essay, we discuss a framework of concepts leading towards general design standards for tissue-engineering, focusing in particular on systematic design strategies, control of cell behavior, physiological scaling, fabrication modes and functional evaluation.
Design standards for engineered tissues
57 -, Agarwal A, Goss JA, Cho A, McCain ML, and Parker KK. 2013. “Microfluidic heart on a chip for higher throughput pharmacological studies.” Lab on a Chip, 13, 18, Pp. 3599-608. Publisher's VersionAbstract
We present the design of a higher throughput “heart on a chip” which utilizes a semi-automated fabrication technique to process sub millimeter sized thin film cantilevers of soft elastomers. Anisotropic cardiac microtissues which recapitulate the laminar architecture of the heart ventricle are engineered on these cantilevers. Deflection of these cantilevers, termed Muscular Thin Films (MTFs), during muscle contraction allows calculation of diastolic and systolic stresses generated by the engineered tissues. We also present the design of a reusable one channel fluidic microdevice completely built out of autoclavable materials which incorporates various features required for an optical cardiac contractility assay: metallic base which fits on a heating element for temperature control, transparent top for recording cantilever deformation and embedded electrodes for electrical field stimulation of the tissue. We employ the microdevice to test the positive inotropic effect of isoproterenol on cardiac contractility at dosages ranging from 1 nM to 100 μM. The higher throughput fluidic heart on a chip has applications in testing of cardiac tissues built from rare or expensive cell sources and for integration with other organ mimics. These advances will help alleviate translational barriers for commercial adoption of these technologies by improving the throughput and reproducibility of readout, standardization of the platform and scalability of manufacture.
Microfluidic heart on a chip for higher throughput pharmacological studies
56 -, Agarwal A, Farouz Y, Nesmith AP, Deravi LF, McCain ML, and Parker KK. 2013. “Micropatterning Alginate Substrates for In Vitro Cardiovascular Muscle on a Chip.” Advanced Functional Materials, 23, 30, Pp. 3738-46. Publisher's VersionAbstract
Soft hydrogels such as alginate are ideal substrates for building muscle in vitro because they have structural and mechanical properties close to the in vivo extracellular matrix (ECM) network. However, hydrogels are generally not amenable to protein adhesion and patterning. Moreover, muscle structures and their underlying ECM are highly anisotropic, and it is imperative that in vitro models recapitulate the structural anisotropy in reconstructed tissues for in vivo relevance due to the tight coupling between sturcture and function in these systems. We present two techniques to create chemical and structural heterogeneities within soft alginate substrates and employ them to engineer anisotropic muscle monolayers: (i) microcontact printing lines of extracellular matrix proteins on flat alginate substrates to guide cellular processes with chemical cues, and (ii) micromolding of alginate surface into grooves and ridges to guide cellular processes with topographical cues. Neonatal rat ventricular myocytes as well as human umbilical artery vascular smooth muscle cells successfully attach to both these micropatterned substrates leading to subsequent formation of anisotropic striated and smooth muscle tissues. Muscular thin film cantilevers cut from these constructs are then employed for functional characterization of engineered muscular tissues. Thus, micropatterned alginate is an ideal substrate for in vitro models of muscle tissue because it facilitates recapitulation of the anisotropic architecture of muscle, mimics the mechanical properties of the ECM microenvironment, and is amenable to evaluation of functional contractile properties.
Micropatterning Alginate Substrates for In Vitro Cardiovascular Muscle on a Chip
55 -, Deravi LF, Golecki HM, and Parker KK. 2013. “Protein-Based Textiles: Bio-Inspired and Bio-Derived Materials for Medical and Non-Medical Applications.” Journal of Chemical and Biological Interfaces, 1, 1, Pp. 25-34. Publisher's VersionAbstract
The hierarchical structure-dependent function of self-assembling proteins regulates the biochemical and mechanical functions of cells, tissues, and organs. These multi-scale properties make proteins desirable candidates for novel supramolecular materials that require tailored properties and customizable functions. The ability to translate molecular domains of proteins into the bulk production of conformable materials, such as textiles, is restricted by the current limitations in fabrication technologies and the finite abundance of protein starting material. We will review the common features of self-assembling proteins, including their structure-dependent mechanical properties and how these characteristics have inspired techniques for manufacturing protein-based textiles. These technologies coupled with recent advances in recombinant protein synthesis enable the bulk production of fibers and fabrics that emulate the hierarchical function of natural protein networks.
Protein-Based Textiles: Bio-Inspired and Bio-Derived Materials for Medical and Non-Medical Applications
54 -, McCain ML, Sheehy SP, Grosberg A, Goss JA, and Parker KK. 2013. “Recapitulating maladaptive, multiscale remodeling of failing myocardium on a chip.” Proceedings of the National Academy of Sciences of the United States of America, 110, 24, Pp. 9770-9775. Publisher's VersionAbstract
The lack of a robust pipeline of medical therapeutic agents for the treatment of heart disease may be partially attributed to the lack of in vitro models that recapitulate the essential structure-function relationships of healthy and diseased myocardium. We designed and built a system to mimic mechanical overload in vitro by applying cyclic stretch to engineered laminar ventricular tissue on a stretchable chip. To test our model, we quantified changes in gene expression, myocyte architecture, calcium handling, and contractile function and compared our results vs. several decades of animal studies and clinical observations. Cyclic stretch activated gene expression profiles characteristic of pathological remodeling, including decreased α- to β-myosin heavy chain ratios, and induced maladaptive changes to myocyte shape and sarcomere alignment. In stretched tissues, calcium transients resembled those reported in failing myocytes and peak systolic stress was significantly reduced. Our results suggest that failing myocardium, as defined genetically, structurally, and functionally, can be replicated in an in vitro microsystem by faithfully recapitulating the structural and mechanical microenvironment of the diseased heart.
Recapitulating maladaptive, multiscale remodeling of failing myocardium on a chip
2012
53 -, McCain ML, Desplantez T, Geisse NA, Rothen-Rutishauser B, Oberer H, Parker KK, and Kleber AG. 2012. “Cell-to-cell coupling in engineered pairs of rat ventricular cardiomyocytes: relation between Cx43 immunofluorescence and intercellular electrical conductance.” American Journal of Physiology-Heart and Circulatory Physiology, 302, 2, Pp. H443-H450. Publisher's VersionAbstract
Gap junctions are composed of connexin (Cx) proteins, which mediate intercellular communication. Cx43 is the dominant Cx in ventricular myocardium, and Cx45 is present in trace amounts. Cx43 immunosignal has been associated with cell-to-cell coupling and electrical propagation, but no studies have directly correlated Cx43 immunosignal to electrical cell-to-cell conductance, g(j), in ventricular cardiomyocyte pairs. To assess the correlation between Cx43 immunosignal and g(j), we developed a method to determine both parameters from the same cell pair. Neonatal rat ventricular cardiomyocytes were seeded on micropatterned islands of fibronectin. This allowed formation of cell pairs with reproducible shapes and facilitated tracking of cell pair locations. Moreover, cell spreading was limited by the fibronectin pattern, which allowed us to increase cell height by reducing the surface area of the pattern. Whole cell dual voltage clamp was used to record g(j) of cell pairs after 3-5 days in culture. Fixation of cell pairs before removal of patch electrodes enabled preservation of cell morphology and offline identification of patched pairs. Subsequently, pairs were immunostained, and the volume of junctional Cx43 was quantified using confocal microscopy, image deconvolution, and three-dimensional reconstruction. Our results show a linear correlation between g(j) and Cx43 immunosignal within a range of 8-50 nS.
Cell-to-cell coupling in engineered pairs of rat ventricular cardiomyocytes: relation between Cx43 immunofluorescence and intercellular electrical conductance
52 -, Desplantez T, McCain ML, Beauchamp P, Rigoli G, Rothen-Rutishauser B, Parker KK, and Kleber AG. 2012. “Connexin43 ablation in foetal atrial myocytes decreases electrical coupling, partner connexins, and sodium current.” Cardiovascular Research, 94, 1, Pp. 58-65. Publisher's VersionAbstract
Remodelling and regional gradients in expression of connexins (Cx) are thought to contribute to atrial electrical dysfunction and atrial fibrillation. We assessed the effect of interaction between Cx43, Cx40, and Cx45 on atrial cell-to-cell coupling and inward Na current (INa) in engineered pairs of atrial myocytes derived from wild-type mice (Cx43+/+) and mice with genetic ablation of Cx43 (Cx43−/−).
Connexin43 ablation in foetal atrial myocytes decreases electrical coupling, partner connexins, and sodium current.
51 -, Sheehy SP, Grosberg A, and Parker KK. 2012. “The contribution of cellular mechanotransduction to cardiomyocyte form and function.” Biomechanics and Modeling in Mechanobiology, 11, 8, Pp. 1227-1239. Publisher's VersionAbstract
Myocardial development is regulated by an elegantly choreographed ensemble of signaling events mediated by a multitude of intermediates that take a variety of forms. Cellular differentiation and maturation are a subset of vertically integrated processes that extend over several spatial and temporal scales to create a well-defined collective of cells that are able to function cooperatively and reliably at the organ level. Early efforts to understand the molecular mechanisms of cardiomyocyte fate determination focused primarily on genetic and chemical mediators of this process. However, increasing evidence suggests that mechanical interactions between the extracellular matrix (ECM) and cell surface receptors as well as physical interactions between neighboring cells play important roles in regulating the signaling pathways controlling the developmental processes of the heart. Interdisciplinary efforts have made it apparent that the influence of the ECM on cellular behavior occurs through a multitude of physical mechanisms, such as ECM boundary conditions, elasticity, and the propagation of mechanical signals to intracellular compartments, such as the nucleus. In addition to experimental studies, a number of mathematical models have been developed that attempt to capture the interplay between cells and their local microenvironment and the influence these interactions have on cellular self-assembly and functional behavior. Nevertheless, many questions remain unanswered concerning the mechanism through which physical interactions between cardiomyocytes and their environment are translated into biochemical cellular responses and how these signaling modalities can be utilized in vitro to fabricate myocardial tissue constructs from stem cell-derived cardiomyocytes that more faithfully represent their in vivo counterpart. These studies represent a broad effort to characterize biological form as a conduit for information transfer that spans the nanometer length scale of proteins to the meter length scale of the patient and may yield new insights into the contribution of mechanotransduction into heart development and disease.
The contribution of cellular mechanotransduction to cardiomyocyte form and function
50 -, Feinberg AW, Alford PW, Jin H, Ripplinger CM, Werdich AA, Sheehy SP, Grosberg A, and Parker KK. 2012. “Controlling the contractile strength of engineered cardiac muscle by hierarchal tissue architecture.” Biomaterials, 33, 23, Pp. 5732-5741. Publisher's VersionAbstract
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.
Controlling the contractile strength of engineered cardiac muscle by hierarchal tissue architecture
49 -, McCain ML, Lee H, Aratyn-Schaus Y, Kléber AG, and Parker KK. 2012. “Cooperative coupling of cell-matrix and cell-cell adhesions in cardiac muscle.” Proceedings of the National Academy of Sciences of the United States of America, 109, 25, Pp. 9881-9886. Publisher's VersionAbstract
Adhesion between cardiac myocytes is essential for the heart to function as an electromechanical syncytium. Although cell-matrix and cell–cell adhesions reorganize during development and disease, the hierarchical cooperation between these subcellular structures is poorly understood. We reasoned that, during cardiac development, focal adhesions mechanically stabilize cells and tissues during myofibrillogenesis and intercalated disc assembly. As the intercalated disc matures, we postulated that focal adhesions disassemble as systolic stresses are transmitted intercellularly. Finally, we hypothesized that pathological remodeling of cardiac microenvironments induces excessive mechanical loading of intercalated discs, leading to assembly of stabilizing focal adhesions adjacent to the junction. To test our model, we engineered μtissues composed of two ventricular myocytes on deformable substrates of tunable elasticity to measure the dynamic organization and functional remodeling of myofibrils, focal adhesions, and intercalated discs as cooperative ensembles. Maturing μtissues increased systolic force while simultaneously developing into an electromechanical syncytium by disassembling focal adhesions at the cell–cell interface and forming mature intercalated discs that transmitted the systolic load. We found that engineering the microenvironment to mimic fibrosis resulted in focal adhesion formation adjacent to the cell–cell interface, suggesting that the intercalated disc required mechanical reinforcement. In these pathological microenvironments, μtissues exhibited further evidence of maladaptive remodeling, including lower work efficiency, longer contraction cycle duration, and weakened relationships between cytoskeletal organization and force generation. These results suggest that the cooperative balance between cell-matrix and cell–cell adhesions in the heart is guided by an architectural and functional hierarchy established during development and disrupted during disease.
Cooperative coupling of cell-matrix and cell-cell adhesions in cardiac muscle.
48 -, Deravi LF, Su T, Paten JA, Ruberti JW, Bertoldi K, and Parker KK. 2012. “Differential Contributions of Conformation Extension and Domain Unfolding to Properties of Fibronectin Nanotexiles.” Nano Letters, 12, 11, Pp. 5587-5592. Publisher's VersionAbstract
Fibronectin (FN) textiles are built as nanometer-thick fabrics. When uniaxially loaded, these fabrics exhibit a distinct threshold between elastic and plastic deformation with increasing stretch. Fabric mechanics are modeled using an eight-chain network and two-state model, revealing that elastic properties of FN depend on conformational extension of the protein and that plastic deformation depends on domain unfolding. Our results suggest how the molecular architecture of a molecule can be exploited for designer mechanical properties of a bulk material.
Differential Contributions of Conformation Extension and Domain Unfolding to Properties of Fibronectin Nanotexiles
47 -, Beauchamp P, Desplantez T, McCain ML, Li W, Asimaki A, Rigoli G, Parker KK, Saffitz JE, and Kleber AG. 2012. “Electrical Coupling and Propagation in Engineered Ventricular Myocardium With Heterogeneous Expression of Connexin43.” 111.259705Circulation Research, 110, 11, Pp. 1445-1453. Publisher's VersionAbstract
Spatial heterogeneity in connexin (Cx) expression has been implicated in arrhythmogenesis.
Electrical Coupling and Propagation in Engineered Ventricular Myocardium With Heterogeneous Expression of Connexin43.
46 -, Shim J, Grosberg A, Nawroth JC, Parker KK, and Bertoldi K. 2012. “Modeling of cardiac muscle thin films: Pre-stretch, passive and active behavior.” Journal of Biomechanics, 45, 5, Pp. 832-841. Publisher's VersionAbstract
Recent progress in tissue engineering has made it possible to build contractile bio-hybrid materials that undergo conformational changes by growing a layer of cardiac muscle on elastic polymeric membranes. Further development of such muscular thin films for building actuators and powering devices requires exploring several design parameters, which include the alignment of the cardiac myocytes and the thickness/Young’s modulus of elastomeric film. To more efficiently explore these design parameters, we propose a 3-D phenomenological constitutive model, which accounts for both the passive deformation including pre-stretch and the active behavior of the cardiomyocytes. The proposed 3-D constitutive model is implemented within a finite element framework, and can be used to improve the current design of bio-hybrid thin films and help developing bio-hybrid constructs capable of complex conformational changes.
Modeling of cardiac muscle thin films: Pre-stretch, passive and active behavior.
45 -, Grosberg A, Nesmith AP, Goss JA, Brigham MD, McCain ML, and Parker KK. 2012. “Muscle on a chip: In vitro contractility assays for smooth and striated muscle.” Journal of Pharmacological and Toxicological Methods, 65, 3, Pp. 126-135. Publisher's VersionAbstract
article i nfo Introduction: To evaluate the viability of a muscle tissue, it is essential to measure the tissue's contractile performance as well as to control its structure. Accurate contractility data can aid in development of more ef- fective and safer drugs. This can be accomplished with a robust in vitro contractility assay applicable to var- ious types of muscle tissue. Methods: The devices developed in this work were based on the muscular thin film (MTF) technology, in which an elastic film is manufactured with a 2D engineered muscle tissue on one side. The tissue template is made by patterning extracellular matrix with microcontact printing. When muscle cells are seeded on the film, they self-organize with respect to the geometric cues in the matrix to form a tissue. Results: Several assays based on the "MTF on a chip" technology are demonstrated. One such assay incorporates the contractility assay with striated muscle into a fluidic channel. Another assay platform in- corporates the MTFs in a multi-well plate, which is compatible with automated data collection and analysis. Fi- nally, we demonstrate the possibility of analyzing contractility of both striated and smooth muscle simultaneously on the same chip. Discussion: In this work, we assembled an ensemble of contractility assays for striated and smooth muscle based on muscular thin films. Our results suggest an improvement over current methods and an alternative to isolated tissue preparations. Our technology is amenable to both primary har- vests cells and cell lines, as well as both human and animal tissues.
Muscle on a chip: In vitro contractility assays for smooth and striated muscle.
44 -, Kuo P, Lee H, Bray MA, Geisse NA, Huang YT, Adams WJ, Sheehy SP, and Parker KK. 2012. “Myocyte shape regulates lateral registry of sarcomeres and contractility.” American Journal of Pathology, 101, 6, Pp. 2030-7. Publisher's VersionAbstract
The heart actively remodels architecture in response to various physiological and pathological conditions. Gross structural change of the heart chambers is directly reflected at the cellular level by altering the morphological characteristics of individual cardiomyocytes. However, an understanding of the relationship between cardiomyocyte shape and the contractile function remains unclear. By using in vitro assays to analyze systolic stress of cardiomyocytes with controlled shape, we demonstrated that the characteristic morphological features of cardiomyocytes observed in a variety of pathophysiological conditions are correlated with mechanical performance. We found that cardiomyocyte contractility is optimized at the cell length/width ratio observed in normal hearts, and decreases in cardiomyocytes with morphological characteristics resembling those isolated from failing hearts. Quantitative analysis of sarcomeric architecture revealed that the change of contractility may arise from alteration of myofibrillar structure. Measurements of intracellular calcium in myocytes revealed unique characteristics of calcium metabolism as a function of myocyte shape. Our data suggest that cell shape is critical in determining contractile performance of single cardiomyocytes by regulating the intracellular structure and calcium handling ability.
Myocyte shape regulates lateral registry of sarcomeres and contractility
43 -, Dabiri BE, Lee H, and Parker KK. 2012. “A potential role for integrin signaling in mechanoelectrical feedback.” Progress in Biophysics &Molecular Biology, 110, 43864, Pp. 196-203. Publisher's VersionAbstract
Certain forms of heart disease involve gross morphological changes to the myocardium that alter its hemodynamic loading conditions. These changes can ultimately lead to the increased deposition of extracellular matrix (ECM) proteins, such as collagen and fibronectin, which together work to pathologically alter the myocardium's bulk tissue mechanics. In addition to changing the mechanical properties of the heart, this maladaptive remodeling gives rise to changes in myocardium electrical conductivity and synchrony since the tissue's mechanical properties are intimately tied to its electrical characteristics. This phenomenon, called mechanoelectrical coupling (MEC), can render individuals affected by heart disease arrhythmogenic and susceptible to Sudden Cardiac Death (SCD). The underlying mechanisms of MEC have been attributed to various processes, including the action of stretch activated channels and changes in troponin C-Ca(2+) binding affinity. However, changes in the heart post infarction or due to congenital myopathies are also accompanied by shifts in the expression of various molecular components of cardiomyocytes, including the mechanosensitive family of integrin proteins. As transmembrane proteins, integrins mechanically couple the ECM with the intracellular cytoskeleton and have been implicated in mediating ion homeostasis in various cell types, including neurons and smooth muscle. Given evidence of altered integrin expression in the setting of heart disease coupled with the associated increased risk for arrhythmia, we argue in this review that integrin signaling contributes to MEC. In light of the significant mortality associated with arrhythmia and SCD, close examination of all culpable mechanisms, including integrin-mediated MEC, is necessary.
A potential role for integrin signaling in mechanoelectrical feedback
42 -, Nawroth JC, Lee H, Feinberg AW, Ripplinger CM, McCain ML, Grosberg A, Dabiri JO, and Parker KK. 2012. “A tissue-engineered jellyfish with biomimetic propulsion.” Nature Biotechnology, 30, 8, Pp. 792-797. Publisher's VersionAbstract
Reverse engineering of biological form and function requires hierarchical design over several orders of space and time. Recent advances in the mechanistic understanding of biosynthetic compound materials1,2,3, computer-aided design approaches in molecular synthetic biology4,5 and traditional soft robotics6,7, and increasing aptitude in generating structural and chemical microenvironments that promote cellular self-organization8,9,10 have enhanced the ability to recapitulate such hierarchical architecture in engineered biological systems. Here we combined these capabilities in a systematic design strategy to reverse engineer a muscular pump. We report the construction of a freely swimming jellyfish from chemically dissociated rat tissue and silicone polymer as a proof of concept. The constructs, termed 'medusoids', were designed with computer simulations and experiments to match key determinants of jellyfish propulsion and feeding performance by quantitatively mimicking structural design, stroke kinematics and animal-fluid interactions. The combination of the engineering design algorithm with quantitative benchmarks of physiological performance suggests that our strategy is broadly applicable to reverse engineering of muscular organs or simple life forms that pump to survive.
A tissue-engineered jellyfish with biomimetic propulsion
2011
41 -, Patrick W. Alford, Alexander P. Nesmith, Johannes N. Seywerd, Anna Grosberg, and Kevin Kit Parker. 10/12/2011. “Vascular smooth muscle contractility depends on cell shape.” Integrative Biology, 3, 11, Pp. 1063-1070. Publisher's VersionAbstract
The physiologic role of smooth muscle structure in defining arterial function is poorly understood. We aimed to elucidate the relationship between vascular smooth muscle architecture and functional contractile output. Using microcontact printing and muscular thin film technology, we engineered in vitro vascular tissues with strictly defined geometries and tested their contractile function. In all tissues, vascular smooth muscle cells (VSMCs) were highly aligned with in vivo-like spindle architecture, and contracted physiologically in response to stimulation with endothelin-1. However, tissues wherein the VSMCs were forced into exaggerated spindle elongation exerted significantly greater contraction force per unit cross-sectional area than those with smaller aspect ratios. Moreover, this increased contraction did not occur in conjunction with an increase in traditionally measured contractile phenotype markers. These results suggest that cellular architecture within vascular tissues plays a significant role in conferring tissue function and that, in some systems, traditional phenotype characterization is not sufficient to define a functionally contractile population of VSMCs.
Vascular smooth muscle contractility depends on cell shape
40 -, Anna Grosberg, Patrick W. Alford, Megan L. McCain, and Kevin Kit Parker. 10/4/2011. “Ensembles of engineered cardiac tissues for physiological and pharmacological study: Heart on a chip.” Miniaturisation for chemistry, physics, biology, materials science and bioengineering, 11, 24, Pp. 4165-73. Publisher's VersionAbstract
Traditionally, muscle physiology experiments require multiple tissue samples to obtain morphometric, electrophysiological, and contractility data. Furthermore, these experiments are commonly completed one at a time on cover slips of single cells, isotropic monolayers, or in isolated muscle strips. In all of these cases, variability of the samples hinders quantitative comparisons among experimental groups. Here, we report the design of a ‘‘heart on a chip’’ that exploits muscular thin film technology – biohybrid constructs of an engineered, anisotropic ventricular myocardium on an elastomeric thin film – to measure contractility, combined with a quantification of action potential propagation, and cytoskeletal architecture in multiple tissues in the same experiment. We report techniques for real-time data collection and analysis during pharmacological intervention. The chip is an efficient means of measuring structure-function relationships in constructs that replicate the hierarchical tissue architectures of laminar cardiac muscle.
Ensembles of engineered cardiac tissues for physiological and pharmacological study: Heart on a chip

Pages