Elsevier

Biomaterials

Volume 67, October 2015, Pages 52-64
Biomaterials

Combinatorial polymer matrices enhance in vitro maturation of human induced pluripotent stem cell-derived cardiomyocytes

https://doi.org/10.1016/j.biomaterials.2015.07.004Get rights and content

Abstract

Cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs) hold great promise for modeling human heart diseases. However, iPSC-CMs studied to date resemble immature embryonic myocytes and therefore do not adequately recapitulate native adult cardiomyocyte phenotypes. Since extracellular matrix plays an essential role in heart development and maturation in vivo, we sought to develop a synthetic culture matrix that could enhance functional maturation of iPSC-CMs in vitro. In this study, we employed a library of combinatorial polymers comprising of three functional subunits – poly-ε-caprolacton (PCL), polyethylene glycol (PEG), and carboxylated PCL (cPCL) – as synthetic substrates for culturing human iPSC-CMs. Of these, iPSC-CMs cultured on 4%PEG-96%PCL (each % indicates the corresponding molar ratio) exhibit the greatest contractility and mitochondrial function. These functional enhancements are associated with increased expression of cardiac myosin light chain-2v, cardiac troponin I and integrin alpha-7. Importantly, iPSC-CMs cultured on 4%PEG-96%PCL demonstrate troponin I (TnI) isoform switch from the fetal slow skeletal TnI (ssTnI) to the postnatal cardiac TnI (cTnI), the first report of such transition in vitro. Finally, culturing iPSC-CMs on 4%PEG-96%PCL also significantly increased expression of genes encoding intermediate filaments known to transduce integrin-mediated mechanical signals to the myofilaments. In summary, our study demonstrates that synthetic culture matrices engineered from combinatorial polymers can be utilized to promote in vitro maturation of human iPSC-CMs through the engagement of critical matrix-integrin interactions.

Introduction

Heart disease is the leading cause of death in developed countries, accounting for over 36% of all deaths in the United States [1], yet the mechanistic study of human heart disease beginning at the cellular level has been limited by the lack of suitable human cardiomyocyte models. However, thanks to recent revolutionary advances in cellular reprogramming, and directed differentiation of human induced pluripotent stem cells (iPSCs) into cardiomyocytes (iPSC-CMs), a number of in vitro models of healthy and diseased human cardiac tissues have been developed [2], [3]. Despite these advances, an important concern regarding the use of iPSC-CMs is their functional immaturity relative to primary cardiomyocytes. For instance, early studies indicated that cardiomyocytes generated in vitro from pluripotent stem cells exhibit fetal phenotypes with respect to their physiological performance [4], [5]. Several recent studies have addressed maturation of iPSC-CMs or embryonic stem cell-derived cardiomyocytes (ESC-CMs) [6], [7], [8], [9], [10], but the majority were limited to calcium handling and electrophysiological evaluation. Thus, considerable unmet needs remain for the adequate study of in vitro maturation of iPSC-CMs, particularly at the cell and molecular levels, and factors that modulate it. One such factor influencing cell maturation, including cardiomyocytes, is the tissue microenvironment. In particular, cell-substratum interaction is essential for proper development and maintenance of tissue architecture and function. In many complex organisms, the extracellular matrix (ECM) plays a critical role in cardiomyocyte development, but the full mechanism of its impact remains unknown due to the ECM's heterogeneity in both composition and structural orientation. Yet, despite considerable progress being made to engineer niches that control cellular responses through purpose-specific biomaterial designs (e.g., surface patterning, biomolecule addition) that would encompass some of the native ECM properties, the direct effects of characteristic biochemical and biophysical properties of unmodified materials alone have largely been underexplored. To address the need, we employed a library of copolymer scaffolds with varying physicochemical properties as culture substrates [11]. The copolymer library contained different mole percentages of three components: hydrophilic poly(ethylene glycol) (PEG), hydrophobic poly(ε-caprolacton) (PCL), and negatively-charged carboxylated-PCL (cPCL). Each copolymer subunit was selected for the specific properties it contributed to the resulting copolymer: PCL is a semi-crystalline, biodegradable, and hydrophobic, as well as being FDA-approved in medical devices [12]; PEG is a biocompatible, hydrophilic, and repellent polymer that reduces protein adsorption and cell attachment through steric exclusion [13], [14]; and cPCL facilitates cell attachment to the scaffold surface by providing a negative charge, effectively counteracting the PEG's repellant effects [14]. These combinatorial polymers were electrospun to make fiber mesh scaffolds that mimic ECM fiber structure and orientation, and subsequently used as test culture substrates.

Human iPSCs were differentiated into human iPSC-CMs through a directed differentiation protocol [15]. After 15–30 days of culture on each copolymer scaffold, we examined the effects of the copolymer composition on iPSC-CM phenotype by evaluating beating behavior, mitochondrial function and gene expression profiles. Our results indicate that certain combinatorial polymer scaffolds, especially a 4%PEG-96%PCL copolymer, promote the acquisition of several phenotypic features of mature ventricular myocytes including organized sarcomeres, abundant mitochondria, increased contractility and higher expression of cardiac myosin light chain-2v, cardiac troponin I and integrin alpha-7, each of which have been associated with cardiac/ventricular maturation [16], [17], [18]. Moreover, 4%PEG-96%PCL was associated with enhanced expression of intermediate filament-associated proteins involved in transducing integrin-mediated mechanical signals to the myofilaments. These results suggest the synthetic biomaterial promoted cardiac maturation by mimicking some features of basement membrane-integrin/sarcolemma interactions seen in normal development. In summary, our study suggests that specific chemical compositions of synthetic extracellular substrates can exert profound influence on in vitro maturation of iPSC-CMs.

Section snippets

Reprogramming of human dermal fibroblasts and maintenance of human iPSCs

A human iPSC line (CC2) was generated from a healthy control subject using an episomal approach and validated, as we have previously described, following the work of Dr. Shinya Yamanaka [19], [20], [21]. Maintenance and culture of human iPSCs followed our established methods [19], [20], [21], [22]. Pluripotency was validated by PluriTest, a bioinformatics assay [23], using a teratoma-validated line as a positive control, and normal chromosomal karyotype was confirmed (Genetic Associates,

Generation of human iPSC-CMs

Validated human iPSCs from a healthy control subject, generated via an episomal-based reprogramming method developed by Shinya Yamanaka and colleagues, were used for all studies (Fig. S2a) [19], [20], [21]. Direct differentiation of these iPSCs cultured on Matrigel, outlined in Fig. S2b, successfully yielded iPSC-CMs. Visual inspection of the iPSC-CMs 7–8 days after differentiation showed individual beating cells, and 10–13 days after differentiation the iPSC-CMs formed beating sheets of cells (

Discussion

New breakthroughs in cellular reprogramming to generate human iPSCs, which are capable of differentiating into cardiomyocytes, offer extraordinary opportunities to study human heart diseases at the cellular level. However, despite their vast potential, iPSC-CMs generated with existing methods appear to be immature, resembling embryonic or fetal myocytes; thus, iPSC-CMs do not yet fully recapitulate characteristics of adult human myocytes. In this study, we succeeded in enhancing the maturation

Conclusion

Synthetic combinatorial polymer substrates were found to have distinct effects on in vitro maturation of human iPSC-CMs. Specifically, the 4%PEG-96%PCL scaffold enhanced iPSC-CM contractility and mitochondrial activity. This functional improvement was accompanied by the increased expression of MLC-2v, a marker of mature ventricular myocytes, and cTnI, a cardiac adult marker. While the precise mechanism is unknown, our results suggest that the chemical composition of a synthetic substrate

Acknowledgments

Authors thank Dr. Bjorn Knollmann's laboratory for serving healthy rabbit adult myocytes to us for mitochondrial functional assay. This research work was funded and supported by NIH R01HL104040, NIH UH2 TR000491, NIH R01ES010563, NIH R01ES016931, 1R01 NS078289, VA Merit 101BX000771, NSF CAREER CBET 1056046 and Clinical Translational Science Award No. UL1TR000445 from the National Center for Advancing Translational Sciences. Polymer characterization was conducted through the use of the core

References (75)

  • M.Y. Lee et al.

    Derivation of functional ventricular cardiomyocytes using endogenous promoter sequence from murine embryonic stem cells

    Stem Cell Res.

    (2012)
  • T. Marchini et al.

    Impaired cardiac mitochondrial function and contractile reserve following an acute exposure to environmental particulate matter

    Biochim. Biophys. Acta

    (2013)
  • L.M. Loew et al.

    Imaging in 5 dimensions – time-dependent membrane-potentials in individual mitochondria

    Biophys. J.

    (1993)
  • R.C. Scaduto et al.

    Measurement of mitochondrial membrane potential using fluorescent rhodamine derivatives

    Biophys. J.

    (1999)
  • S.W. Kubalak et al.

    Chamber specification of atrial myosin light chain-2 expression precedes septation during murine cardiogenesis

    J. Biol. Chem.

    (1994)
  • P.K. Bhavsar et al.

    Developmental expression of troponin I isoforms in fetal human heart

    FEBS Lett.

    (1991)
  • C. Al-Jassar et al.

    Mechanistic basis of desmosome-targeted diseases

    J. Mol. Biol.

    (2013)
  • R.O. Hynes

    Cell adhesion: old and new questions

    Trends Biochem. Sci.

    (1999)
  • I. Gyorke et al.

    The role of calsequestrin, triadin, and junctin in conferring cardiac ryanodine receptor responsiveness to luminal calcium

    Biophys. J.

    (2004)
  • A. Bizy et al.

    Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes

    Stem Cell Res.

    (2013)
  • D. Lloyd-Jones et al.

    Heart disease and stroke statistics–2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee

    Circulation

    (2009)
  • G. Wang et al.

    Modeling the mitochondrial cardiomyopathy of barth syndrome with induced pluripotent stem cell and heart-on-chip technologies

    Nat. Med.

    (2014)
  • K. Dolnikov et al.

    Functional properties of human embryonic stem cell-derived cardiomyocytes: intracellular Ca2+ handling and the role of sarcoplasmic reticulum in the contraction

    Stem cells

    (2006)
  • L. Yang et al.

    Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population

    Nature

    (2008)
  • C. Kim et al.

    Non-cardiomyocytes influence the electrophysiological maturation of human embryonic stem cell-derived cardiomyocytes during differentiation

    Stem Cells Dev.

    (2010)
  • J. Liu et al.

    Facilitated maturation of Ca2+ handling properties of human embryonic stem cell-derived cardiomyocytes by calsequestrin expression

    Am. J. Physiol. Cell Physiol.

    (2009)
  • L. Sartiani et al.

    Developmental changes in cardiomyocytes differentiated from human embryonic stem cells: a molecular and electrophysiological approach

    Stem Cells

    (2007)
  • M.K. Gupta et al.

    Combinatorial polymer electrospun matrices promote physiologically-relevant cardiomyogenic stem cell differentiation

    Plos One

    (2011)
  • H.J. Sung et al.

    Poly(ethylene glycol) as a sensitive regulator of cell survival fate on polymeric biomaterials: the interplay of cell adhesion and pro-oxidant signaling mechanisms

    Soft Matter

    (2010)
  • J. Zhang et al.

    Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method

    Circulation Res.

    (2012)
  • A.M. Belkin et al.

    beta 1D integrin displaces the beta 1A isoform in striated muscles: localization at junctional structures and signaling potential in nonmuscle cells

    J. Cell Biol.

    (1996)
  • N.L. Flintoff-Dye et al.

    Role for the alpha 7 beta 1 integrin in vascular development and integrity

    Dev. Dynam.

    (2005)
  • A.A. Aboud et al.

    PARK2 patient neuroprogenitors show increased mitochondrial sensitivity to copper

    Neurobiol. Dis.

    (2014)
  • K. Okita et al.

    A more efficient method to generate integration-free human iPS cells

    Nat. Methods

    (2011)
  • M.D. Neely et al.

    DMH1, a highly selective small molecule BMP inhibitor promotes neurogenesis of hiPSCs: comparison of PAX6 and SOX1 expression during neural induction

    ACS Chem. Neurosci.

    (2012)
  • F.J. Muller et al.

    A bioinformatic assay for pluripotency in human cells

    Nat. Methods

    (2011)
  • B.C. Knollmann et al.

    Casq2 deletion causes sarcoplasmic reticulum volume increase, premature Ca2+ release, and catecholaminergic polymorphic ventricular tachycardia

    J. Clin. Invest.

    (2006)
  • Cited by (0)

    View full text