Elsevier

Biomaterials

Volume 32, Issue 7, March 2011, Pages 1856-1864
Biomaterials

The significance of pore microarchitecture in a multi-layered elastomeric scaffold for contractile cardiac muscle constructs

https://doi.org/10.1016/j.biomaterials.2010.11.032Get rights and content

Abstract

Multi-layered poly(glycerol-sebacate) (PGS) scaffolds with controlled pore microarchitectures were fabricated, combined with heart cells, and cultured with perfusion to engineer contractile cardiac muscle constructs. First, one-layered (1L) scaffolds with accordion-like honeycomb shaped pores and elastomeric mechanical properties were fabricated by laser microablation of PGS membranes. Second, two-layered (2L) scaffolds with fully interconnected three dimensional pore networks were fabricated by oxygen plasma treatment of 1L scaffolds followed by stacking with off-set laminae to produce a tightly bonded composite. Third, heart cells were cultured on scaffolds with or without interstitial perfusion for 7 days. The laser-microablated PGS scaffolds exhibited ultimate tensile strength and strain-to-failure higher than normal adult rat left ventricular myocardium, and effective stiffnesses ranging from 220 to 290 kPa. The 7-day constructs contracted in response to electrical field stimulation. Excitation thresholds were unaffected by scaffold scale up from 1L to 2L. The 2L constructs exhibited reduced apoptosis, increased expression of connexin-43 (Cx-43) and matrix metalloprotease-2 (MMP-2) genes, and increased Cx-43 and cardiac troponin-I proteins when cultured with perfusion as compared to static controls. Together, these findings suggest that multi-layered, microfabricated PGS scaffolds may be applicable to myocardial repair applications requiring mechanical support, cell delivery and active implant contractility.

Introduction

Cardiovascular disease is the leading cause of death in developed countries [1] and congenital heart disease, which affects approximately one percent of newborns world-wide, is associated with high morbidity [2]. The functional consequences of myocardial infarction (MI) and other heart defects in which muscle fibers and collagen networks are disrupted are loss of myocardial elasticity, compliance and pumping action [3]. Current myocardial regeneration strategies, while promising [4], are unable to recreate the robust mechanical and contractile properties of normal heart muscle. In particular, an effective graft for myocardial repair is a critical unmet need, where combining elasticity and strength without compromising heart cell viability and contractility have proved challenging [5], [6], [7].

In the prototypical tissue engineering approach, three dimensional (3D) scaffolds provide the delivery vehicle for transplanting large numbers of viable cells toward a goal of tissue regeneration [8], [9]. Numerous 3D biomaterials have been explored as cardiac tissue engineering scaffolds, including non-woven poly(glycolic acid) (PGA) mesh [10], [11], [12], collagen gel [13], [14], collagen foam [15], [16], [17], [18], [19], alginate foam [20], [21], chitosan foam [22], knitted poly(lactic acid) [23], knitted hyaluronan ester [24], poly-4-hydroxybutyrate foam [25], poly(lactic acid)/poly(glycolic-co-lactic acid) (PLLA/PLGA) foam [26], and composites of natural and synthetic polymers [27]. However, these scaffolds are either thermoplastic polymers, which tend to be stiffer than normal soft tissues, degrade by bulk hydrolysis, and fail under long-term cyclic loading [28], or naturally occurring materials with intrinsic variability, immunogenicity, and mechanical strength concerns [29]. Langer and colleagues [30] developed a tough bioresorbable elastomer, poly(glycerol-sebacate) (PGS), that degraded predominately by surface hydrolysis [31] and has been tested in various tissue engineering applications [32], [33], [34] including myocardial repair. The mechanical properties of the PGS elastomer, both in the context of non-porous membranes [7], [35], [36] and porous scaffolds [37], [38], could be tailored to match those of normal heart muscle. Recently, one-layered (1L) PGS scaffolds with in-plane pore anisotropy, i.e., rectangular and accordion-like honeycomb pores produced by laser microablation of ∼250 μm thick PGS membranes [37], were shown to guide the alignment of cultured neonatal rat heart cells [37] and C2C12 myoblasts [39].

Alternatives to the cell-scaffold paradigm include “scaffold-free” approaches based on transplanting cell–cell or cell-extracellular matrix (ECM) grafts. As examples, engraftment and vascularization were demonstrated for heart cell patches comprised of human embryonic stem cell-derived cardiomyocytes, endothelial cells, and fibroblasts [40] and electrical and vascular integration were demonstrated after implantation of thin (∼100 μm) heart cell sheets comprised of interconnected cardiomyocytes [41]. However, scalability remains a major limitation of scaffold-free approaches [9], [13], [42], [43]. Other alternative approaches include “cell-free” biomaterials for myocardial repair. However, biomaterials used for congenital heart defect repair in pediatric patients are limited by lack of potential for growth and remodeling [44], [45], and although cell-free, non-porous PGS membranes were recently shown to reduce post-infarction myocardial hypertrophy in rodents, these implants could not assist contractile function, suggesting a role for cell-PGS implants in future approaches [35].

In the present study, multi-layered elastomeric PGS scaffolds with controlled pore microarchitectures were fabricated and combined with heart cells to engineer contractile cardiac muscle constructs in vitro. Excitation threshold, gene expression, and cardiac specific marker proteins were assessed under different conditions of cell seeding and cultivation, in particular scaffold coating with laminin (LN) to promote heart cell attachment [11], [38], [46] and interstitial perfusion to promote heart cell viability [12], [18], [19], [20], [47].

Section snippets

Methods

Fig. 1 provides an overview of methods used to microfabricate and demonstrate the multi-layered PGS scaffold.

Scaffold microfabrication

To produce 1L scaffolds with accordion-like honeycomb pores in PGS membranes, we adapted our previously described method [37] for use with a frequency quintupled 213 nm Nd:YAG laser. A program was written in Visual Basic for Applications to generate sequences of coordinates and laser parameters, and suitably formatted for uploading into the software controlling this laser (DigiLaz II, v.3.1.0; CETAC Technologies) such that a specified in-plane pore microarchitecture could be automatically

Discussion

Tissue engineered cardiac muscle remains challenged by cell sourcing, mass transport, and scaffold limitations [4], [7]. Recent advances in PGS microfabrication have permitted the design of biodegradable, elastomeric scaffolds with precisely defined pore microarchitectures amenable to both cardiomyogenesis and predictive mathematical modeling. Toward scaling-up our previous 1L accordion-like honeycomb PGS scaffolds for cardiac tissue engineering [37], laser-microablated PGS membranes were

Conclusion

Multi-layered elastomeric PGS scaffolds with controlled pore microarchitectures were fabricated by laser ablation and oxygen plasma-mediated lamination, seeded with heart cells, and cultured with interstitial perfusion. The laser-microablated PGS exhibited UTS and ɛf higher than normal rat left ventricular myocardium and stiffnesses ranging from 220 to 290 kPa. Heart cell culture on these scaffolds yielded cardiac muscle constructs. Excitation thresholds were unaffected by scaffold scale up

Acknowledgments

This work was funded by the American Recovery and Reinvestment Act (ARRA), Award 1-R01-HL086521-01A2 (to LEF) from the National Heart, Lung and Blood Institute (NHLBI). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NHLBI or NIH. We are indebted to R. Langer for general advice, J. Wang and J. Hsiao for help with polymer synthesis, processing, and SEM, N. Watson for help with microscopy, M.G. Moretti and G. Talo and E. Kim for

References (68)

  • C.A. Sundback et al.

    Biocompatibility analysis of poly(glycerol sebacate) as a nerve guide material

    Biomaterials

    (2005)
  • J.W. Nichol et al.

    Co-culture induces alignment in engineered cardiac constructs via MMP-2 expression

    Biochem Biophys Res Commun

    (2008)
  • J.G. Jacot et al.

    Substrate stiffness affects the functional maturation of neonatal rat ventricular myocytes

    Biophys J

    (2008)
  • A. Jean et al.

    Finite element analysis of an accordion-like honeycomb scaffold for cardiac tissue engineering

    J Biomech

    (2010)
  • D. Lloyd-Jones et al.

    Heart disease and stroke statistics–2010 update: a report from the American heart association

    Circulation

    (2010)
  • J.A. Martin et al.

    Births: final data for 2003

    Natl Vital Stat Rep

    (2005)
  • G.M. Fomovsky et al.

    Evolution of scar structure, mechanics, and ventricular function after myocardial infarction in the rat

    Am J Physiol Heart Circ Physiol

    (2010)
  • K.R. Chien et al.

    Cardiogenesis and the complex biology of regenerative cardiovascular medicine

    Science

    (2008)
  • D. Boethig et al.

    Mid term course after pediatric right ventricular outflow tract reconstruction: a comparison of homografts, porcine xenografts and contegras

    Eur J Cardiothorac Surg

    (2005)
  • R. Langer et al.

    Tissue engineering

    Science

    (1993)
  • N. Bursac et al.

    Cardiac muscle tissue engineering: toward an in vitro model for electrophysiological studies

    Am J Physiol

    (1999)
  • M. Papadaki et al.

    Tissue engineering of functional cardiac muscle: molecular, structural, and electrophysiological studies

    Am J Physiol Heart Circ Physiol

    (2001)
  • R.L. Carrier et al.

    Perfusion improves tissue architecture of engineered cardiac muscle

    Tissue Eng

    (2002)
  • T. Eschenhagen et al.

    Engineering myocardial tissue

    Circ Res

    (2005)
  • W.H. Zimmermann et al.

    Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts

    Nat Med

    (2006)
  • R.K. Li et al.

    Survival and function of bioengineered cardiac grafts

    Circulation

    (1999)
  • P. Akhyari et al.

    Mechanical stretch regimen enhances the formation of bioengineered autologous cardiac muscle grafts

    Circulation

    (2002)
  • M. Radisic et al.

    High-density seeding of myocyte cells for cardiac tissue engineering

    Biotechnol Bioeng

    (2003)
  • M. Radisic et al.

    Medium perfusion enables engineering of compact and contractile cardiac tissue

    Am J Physiol Heart Circ Physiol

    (2004)
  • M. Cheng et al.

    Insulin-like growth factor-I and slow, bi-directional perfusion enhance the formation of tissue-engineered cardiac grafts

    Tissue Eng Part A

    (2009)
  • T. Dvir et al.

    Activation of the ERK1/2 cascade via pulsatile interstitial fluid flow promotes cardiac tissue assembly

    Tissue Eng

    (2007)
  • T. Dvir et al.

    A novel perfusion bioreactor providing a homogenous milieu for tissue regeneration

    Tissue Eng

    (2006)
  • N.R. Blan et al.

    Design and fabrication of heart muscle using scaffold-based tissue engineering

    J Biomed Mater Res A

    (2008)
  • K. Matsubayashi et al.

    Improved left ventricular aneurysm repair with bioengineered vascular smooth muscle grafts

    Circulation

    (2003)
  • Cited by (75)

    • Development of modified and multifunctional poly(glycerol sebacate) (PGS)-based biomaterials for biomedical applications

      2021, European Polymer Journal
      Citation Excerpt :

      The two-layered (2L) scaffolds with fully interconnected three-dimensional pore networks were fabricated by oxygen plasma treatment of monolayer scaffolds followed by stacking with off-set laminae to produce a tightly bonded composite. The 2L constructs exhibited reduced apoptosis, increased expression of connexin-43 (Cx-43) and matrix metalloprotease-2 (MMP-2) genes, and thereby upregulating Cx-43 and troponin I (TnI) proteins when cultured with interstitial perfusion [81]. Meanwhile, inspired by the concept of bionic, Annabi et al. combined PGS/PCL microfibers and micro-fabricated PGS in a tri-layered construction [87].

    View all citing articles on Scopus
    View full text