1. Introduction
1.1. Treatment for ischemic heart diseases: State-of-the-art
Cardiovascular diseases, especially ischemic heart disease, are the leading cause of mortality in the United States [1]. In the past decades, pharmacological therapy, such as β-blocker, Angiotensin Converting Enzyme Inhibitor (ACEI)/Angiotensin Receptor Blocker(ARB) have been shown to ameliorate cardiac dysfunction and limit heart remodeling following myocardial infarction [2]. Moreover, the development of coronary artery bypass graft (CABG) can recanalize occlusive coronary and salvage the remaining surviving myocardium [3]. More recently, with the development of percutaneous coronary intervention (PCI), retrograde approach through collaterals has been introduced for percutaneous recanalization of chronic total occlusion (CTO) of the coronary arteries, a new option for the patient with CTO beyond CABG [4]. Unfortunately, even with great advances of these modern technologies, myocardial infarctions will eventually develop into decompensated chronic heart failure. Heart transplantation is currently the last resort for the patient with end-stage heart failure. However, this therapeutic option is limited by donor organ shortage and eventual organ rejection [5]. Therefore, new therapies are required to prevent the progression of pathological remodeling and cell death, as well as to induce tissue recovery in the ischemic heart. Regenerative cardiovascular medicine becomes a holy grail with the goal to replace and repair the damaged myocardium and reverse heart dysfunction. In the past two decades, a variety of stem cells have been investigated by scientists to achieve this goal [6]. Recently developed reprogramming technology, induced pluripotent stem cells (iPSC) has become an alternative source for embryonic stem cells (ESC) without the ethical drawbacks [7], showing their powerful potential to differentiate into desired cell type [8]. A new discipline termed stem cell engineering has recently emerged aiming to reconstruct the damaged heart by integrating progenitor/stem cell biology and bioengineering technology [9]. In this chapter, the recent the recent advances and challenges of progenitor/stem cell engineering for the therapy of ischemic heart disease will be discussed from a translational perspective.
2. Proper cell sources
Appropriate cell sources for stem cell engineering must meet the following requirements: 1) The cell must be electromechanically coupled with the host heart tissue. 2) They must survive in the hostile environment created by ischemic stress [10]. In this regard, several cell types have been tried. Skeletal myoblast was the pioneering attempt but it was found to be limited to contract synchronously with the host myocardium [11]. Bone marrow derived stem cells or hematopoietic stem cells are easily obtained in clinical setting but their potential of cardiogenic differentiation is still being debated [10]. The recent discovery of multipotent cardiac progenitors have been proven to give rise to cardiomyocytes, endothelial, and smooth muscle cells, forming the basic “components” for heart reconstruction [12]. The attempt to discover endogenous cardiac progenitors showed some of those express c-Kit [13, 14] or Sca-1 [14] markers. Transplantation of c-Kit+ cell resulted in neovascularization and cardiomyogenesis in the infarcted heart [13]. Moreover, the cardiosphere obtained from human heart tissue contained a mixed population of c-Kit+ and Sca-1+ cells and could regenerate infarcted heart [15]. These encouraging results led to the initiation of several phase 1 clinical trials: ALCADIA ((AutoLogous Human CArdiac-Derived Stem Cell to Treat Ischemic cArdiomyopathy, NCT00981006), SCIPIO (Cardiac Stem Cell Infusion in Patients with Ischemic Cardiomyopathy) (NCT00474461), and CADUCEUS (NCT00893360). Preliminary data from the SCIPIO and CADUCEUS (CArdiosphere-Derived aUtologous Stem CElls to Reverse ventricUlar dysfunction) trials have been recently published in the
Furthermore, stimulation of the adult progenitor pool with epicardial origin after an acute myocardial infarction (AMI) has been reported with some progress [18-20]. The successful production of iPSC by transducing pluripotent-regulated transcriptional factors has made it a powerful weapon for
3. Engineering approach
Direct injection of cell suspension into infarcted area or peri-infarcted area is still the main progenitor/stem cell treatment used for cardiac repair. Nevertheless, the poor survival of stem cells in the harsh environment (hypoxia, inflammatory cytokines, etc.) limits the reparative function of progenitor/stem cells in ischemic heart [28].
Combining progenitor/stem cell biology and bioengineering, tissue engineering holds great promise to generate viable three dimensional heart tissue with vasculature prior to engraftment to the heart, as an integral part of the host. This tissue graft should display contractile and electromechanical coupled properties, contributing to the improvement of heart function. The recent evidence from large animals indicated that human ES-derived cardiomyocytes electrically coupled and suppressed arrhythmias in injured hearts. This provided support for the continued development of human stem cell derived tissue graft for cardiac repair [29]. The conventional approach involved seeding cell on scaffolds and culture
Eschenhagen and Zimmermann constructed an engineering heart tissue (EHT) by seeding neonatal rat cardiomyocytes and a mix of collagen I, extracellular matrix proteins (Matrigel) into a lattices or circular molds. Upon spontaneous remodeling of the liquid reconstitution mixture and cyclic mechanical, EHT spontaneously and synchronously contracted after one to two weeks of cultivation, which highlights the great importance of physical stimulation on the maintenance of the physical and mechanical function of EHT [30]. This pioneering work for the first time showed EHT could ameliorate cardiac function post MI.
3.1. Scaffold free tissue construct
The use of cell sheets provides a simple scaffold-free approach by seeding cardiac cells on poly (N-isopropylacrylamide)-grafted polystyrene dishes and then lowering the temperature to 20ºC, thus inducing the detachment of intact cell monolayers without enzymatic digestion. Using this method, a 1-mm-thick cell patch can be created by serial stacking of multiple monolayer sheets [31]. A recent study by Murry group using ESC-derived cardiomyocytes reported another scaffold free approach, demonstrating cell aggregation is sufficient to generate functional EHT also showing endothelial cell and fibroblast are required for the survival and integration of EHT and in host myocardium [32]. Further studies are needed to optimize the proportion of cardiomyocyte, endothelial cells, and fibroblast for maximal performance of EHT.
3.2. Construction of myocardial tissue/heart using decellularized native tissue
In order to create decellularized scaffolds, Taylor and her team perfused rat hearts with detergents to remove the cells and leave a complex architecture of acellular extracellular matrix (ECM) behind. [33]. This native scaffold was reseeded with cardiomyocytes and endothelial cells taken from rats. They then placed these constructs in bioreactors that simulated blood pressure, electrical stimulation, and other aspects of cardiac physiology to assure integration of the scaffold and seeded cells. Although approximately only 2% of normal contractile activity was acquired from this approach, this is a successful proof-of-concept trial and might be the ultimate biomimetic method for constructing an intact human heart.
3.3. Porous scaffolds
By using electrical stimulation, Vunjak-Novakovic
3.4. Biological and synthetic polymers
Collagen is the first biological polymer used for fabrication of three dimensional tissues. It was reported that neonatal rat cardiomyocytes spontaneously contracted when cultivated in gelatin coated scaffold. Although the implanted cardiomyocytes survived in infarcted heart, LVEF is not significantly improved after long term observation. Furthermore, Zimmermann’s group engineered contractile 3-D heart tissue, in which cardiomyocytes encapsulated with ring--shaped hydrogels (collagen and Matrigel) showing reduction of ventricle dilatation, significant ventricular wall thickening and improvement of the fractional shortening (FS) [30]. The improvement of cardiac function, myofibril organization indicated that mechanical stimulation is important for maturation of myocardial structure. Leor et al. also reported that 3-D alginate scaffolds seeded with fetal rat cardiomyocytes attenuated left ventricular dilatation and deterioration of the heart function after myocardial infarction [37].
4. Progenitor/stem cell niche engineering
As we discussed above, the laboratories of Deepak Srivastava and Eric Olson were successful in reprogramming cardiac fibroblasts
4.1. Matrix rigidity
Progenitor/stem cell engineering involves coordination of selective proliferation of precursor/stem cells and differentiation into target somatic cells (cardiomyocytes, smooth muscle cell, and endothelial cells). Mechanical cues influence proliferation, differentiation, migration, and spatial morphological organization. These cues include the rigidity of the surrounding matrix or cell adhesion substratum. Thus, better understanding of the role of matrix rigidity is critical for optimization of the regimes of mechanical conditioning of cultured tissue constructs. Based on a pioneering study which discovered mechanosensitive transcriptional mechanism in 2009 [40], Kshitiz
4.2. Exosome secretion
Recent studies have suggested four potential mechanisms for how exogenous-culture-expanded MSC may contribute to cardiovascular repair: transdifferentiation, cell fusion with a native cell, stimulation of endogenous cardiac progenitor/stem cells via direct cell-cell communication or paracrine mechanism [42]. Transdifferentiation of MSC into cardiomyocytes is not inefficient in current regimes [10]. Cell fusion is a rare event[43]. As aforementioned, the observed salutary effects of progenitor/stem cell on cardiac repair probably resulted from paracrine mechanism. And the cardiogenic differentiation of CSC stimulated by MSC processed a limited capacity [44]. By antibody array and Liquid Chromatography with Tandem Mass Spectrometry Detection (LC-MS/MS), compelling evidences have shown MSC could secrete a wide spectrum of trophic proteins that could induce proliferation and differentiation of CPC and angiogenesis [45]. Interestingly, in 2007, the paper published on
5. Conclusions
Progenitor/stem cell engineering has presented as an exciting and promising avenue for the treatment of ischemic heart diseases. Regeneration of damaged heart by progenitor/stem cell engineering is becoming a fact rather than fiction. The translation of experimental discovery in progenitor/stem cell engineering into clinical application should be accelerated and large scale clinical trials should be initiated in the patients with ischemic heart diseases. Therefore, the collaboration of progenitor/stem cell biologists, bioengineers, and physicians is possibly the future modality in personalized regenerative medicine.
Acknowledgments
Funding sources
This work was supported by NIH grants, HL089824, HL081859, HL110740 (Y. Wang).
Mianna Armstrong for technical assistance.
References
- 1.
Heart disease and stroke statistics--2011 update: a report from the American Heart Association. Circulation. (Journal Article).Roger V. L Go A. S Lloyd-jones D. M Adams R. J Berry J. D Brown T. M et al 2011 e18 209 - 2.
Adding ACEIs and/or ARBs to Standard Therapy for Stable Ischemic Heart Disease: Benefits and Harms Book Chapter).2007 - 3.
Heart failure in 2011: Heart failure therapy--technology to the fore. Nat Rev Cardiol. (Journal Article; Review).Mcmurray J. J 2012 9 2 73 4 - 4.
Successful recanalization of chronic total occlusions is associated with improved long-term survival. JACC Cardiovasc Interv. (Journal Article).Jones D. A Weerackody R Rathod K Behar J Gallagher S Knight C. J et al 2012 5 4 380 8 - 5.
ACCF/AHA/HFSA 2011 survey results: current staffing profile of heart failure programs, including programs that perform heart transplant and mechanical circulatory support device implantation. J Card Fail. (Comparative Study; Journal Article; Multicenter Study).Jessup M Albert N. M Lanfear D. E Lindenfeld J Massie B. M Walsh M. N et al 2011 17 5 349 58 - 6.
Towards regenerative therapy for cardiac disease. Lancet. (Journal Article; Research Support, Non-U.S. Gov’t; Review).Ptaszek L. M Mansour M Ruskin J. N Chien K. R 2012 379 9819 933 42 - 7.
Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. (Journal Article; Research Support, Non-U.S. Gov’t).Takahashi K Tanabe K Ohnuki M Narita M Ichisaka T Tomoda K et al 2007 131 5 861 72 - 8.
Concise review: Induced pluripotent stem cells versus embryonic stem cells: close enough or yet too far apart? Stem Cells. (Journal Article; Research Support, Non-U.S. Gov’t; Review).Bilic J Izpisua B. J 2012 30 1 33 41 - 9.
The current status of engineering myocardial tissue. Stem Cell Rev. (Journal Article; Review).Sui R Liao X Zhou X Tan Q 2011 7 1 172 80 - 10.
Heart regeneration. Nature. (Journal Article; Research Support, N.I.H., Extramural; Review).Laflamme M. A Murry C. E 2011 473 7347 326 35 - 11.
Skeletal myoblasts for heart regeneration and repair: state of the art and perspectives on the mechanisms for functional cardiac benefits. Curr Pharm Des. (Journal Article; Review).Formigli L Zecchi-orlandini S Meacci E Bani D 2010 16 8 915 28 - 12.
Regeneration next: toward heart stem cell therapeutics. Cell Stem Cell. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t; Review).Hansson E. M Lindsay M. E Chien K. R 2009 5 4 364 77 - 13.
Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. (Journal Article; Research Support, U.S. Gov’t, P.H.S.).Beltrami A. P Barlucchi L Torella D Baker M Limana F Chimenti S et al 2003 114 6 763 76 - 14.
Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. (In Vitro; Journal Article; Research Support, Non-U.S. Gov’t; Research Support, U.S. Gov’t, P.H.S.).Oh H Bradfute S. B Gallardo T. D Nakamura T Gaussin V Mishina Y et al 2003 100 21 12313 8 - 15.
Relative roles of direct regeneration versus paracrine effects of human cardiosphere-derived cells transplanted into infarcted mice. Circ Res. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Chimenti I Smith R. R Li T. S Gerstenblith G Messina E Giacomello A et al 2010 106 5 971 80 - 16.
S, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. Lancet. (Clinical Trial, Phase I; Journal Article; Randomized Controlled Trial; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Bolli R Chugh A. R D Amario D Loughran J. H Stoddard M. F Ikram 2011 378 9806 1847 57 - 17.
Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet. (Clinical Trial, Phase I; Journal Article; Multicenter Study; Randomized Controlled Trial; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Makkar R. R Smith R. R Cheng K Malliaras K Thomson L. E Berman D et al 2012 379 9819 895 904 - 18.
Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Zhou B Ma Q Rajagopal S Wu S. M Domian I Rivera-feliciano J et al 2008 454 7200 109 13 - 19.
De novo cardiomyocytes from within the activated adult heart after injury. Nature. (Journal Article; Research Support, Non-U.S. Gov’t).Smart N Bollini S Dube K. N Vieira J. M Zhou B Davidson S et al 2011 474 7353 640 4 - 20.
Adult mouse epicardium modulates myocardial injury by secreting paracrine factors. J Clin Invest. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Zhou B Honor L. B He H Ma Q Oh J. H Butterfield C et al 2011 121 5 1894 904 - 21.
Production of de novo cardiomyocytes: human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Burridge P. W Keller G Gold J. D Wu J. C 2012 10 1 16 28 - 22.
Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature. (Journal Article; Research Support, N.I.H., Extramural).Yang L Soonpaa M. H Adler E. D Roepke T. K Kattman S. J Kennedy M et al 2008 453 7194 524 8 - 23.
Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Laflamme M. A Chen K. Y Naumova A. V Muskheli V Fugate J. A Dupras S. K et al 2007 25 9 1015 24 - 24.
Mouse and human induced pluripotent stem cells as a source for multipotent Isl1+ cardiovascular progenitors. Faseb J. (Journal Article; Research Support, Non-U.S. Gov’t).Moretti A Bellin M Jung C. B Thies T. M Takashima Y Bernshausen A et al 2010 24 3 700 11 - 25.
Protecting against wayward human induced pluripotent stem cells with a suicide gene. Biomaterials. (Journal Article; Research Support, Non-U.S. Gov’t).Cheng F Ke Q Chen F Cai B Gao Y Ye C et al 2012 33 11 3195 204 - 26.
In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Qian L Huang Y Spencer C. I Foley A Vedantham V Liu L et al 2012 485 7400 593 8 - 27.
Heart repair by reprogramming non-myocytes with cardiac transcription factors. Nature. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Song K Nam Y. J Luo X Qi X Tan W Huang G. N et al 2012 485 7400 599 604 - 28.
Perspectives on stem cell therapy for cardiac regeneration. Advances and challenges. Circ J. (Journal Article; Research Support, Non-U.S. Gov’t).Choi S. H Jung S. Y Kwon S. M Baek S. H 2012 76 6 1307 12 - 29.
Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature. (JOURNAL ARTICLE).Shiba Y Fernandes S Zhu W. Z Filice D Muskheli V Kim J et al 2012 - 30.
Tissue engineering of a differentiated cardiac muscle construct. Circ Res. (Journal Article; Research Support, Non-U.S. Gov’t).Zimmermann W. H Schneiderbanger K Schubert P Didie M Munzel F Heubach J. F et al 2002 90 2 223 30 - 31.
Fabrication of functional three-dimensional tissues by stacking cell sheets in vitro. Nat Protoc. (Journal Article; Research Support, Non-U.S. Gov’t).Haraguchi Y Shimizu T Sasagawa T Sekine H Sakaguchi K Kikuchi T et al 2012 7 5 850 8 - 32.
Scaffold-free human cardiac tissue patch created from embryonic stem cells. Tissue Eng Part A. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Stevens K. R Pabon L Muskheli V Murry C. E 2009 15 6 1211 22 - 33.
Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nat Med. (Journal Article; Research Support, Non-U.S. Gov’t).Ott H. C Matthiesen T. S Goh S. K Black L. D Kren S. M Netoff T. I et al 2008 14 2 213 21 - 34.
Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proc Natl Acad Sci U S A. (Journal Article; Research Support, Non-U.S. Gov’t; Research Support, U.S. Gov’t, Non-P.H.S.; Research Support, U.S. Gov’t, P.H.S.).Radisic M Park H Shing H Consi T Schoen F. J Langer R et al 2004 101 52 18129 34 - 35.
Electrical stimulation of human embryonic stem cells: cardiac differentiation and the generation of reactive oxygen species. Exp Cell Res. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Serena E Figallo E Tandon N Cannizzaro C Gerecht S Elvassore N et al 2009 315 20 3611 9 - 36.
Electrical stimulation systems for cardiac tissue engineering. Nat Protoc. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Tandon N Cannizzaro C Chao P. H Maidhof R Marsano A Au H. T et al 2009 4 2 155 73 - 37.
Intracoronary injection of in situ forming alginate hydrogel reverses left ventricular remodeling after myocardial infarction in Swine. J Am Coll Cardiol. (Journal Article; Research Support, Non-U.S. Gov’t).Leor J Tuvia S Guetta V Manczur F Castel D Willenz U et al 2009 54 11 1014 23 - 38.
Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Ieda M Fu J. D Delgado-olguin P Vedantham V Hayashi Y Bruneau B. G et al 2010 142 3 375 86 - 39.
Inefficient reprogramming of fibroblasts into cardiomyocytes using Gata4, Mef2c, and Tbx5. Circ Res. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Chen J. X Krane M Deutsch M. A Wang L Rav-acha M Gregoire S et al 2012 111 1 50 5 - 40.
A mechanosensitive transcriptional mechanism that controls angiogenesis. Nature. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t; Research Support, U.S. Gov’t, Non-P.H.S.).Mammoto A Connor K. M Mammoto T Yung C. W Huh D Aderman C. M et al 2009 457 7233 1103 8 - 41.
Kshitiz Hubbi ME, Ahn EH, Downey J, Afzal J, Kim DH, et al. Matrix rigidity controls endothelial differentiation and morphogenesis of cardiac precursors. Sci Signal. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).2012 a41. - 42.
Mesenchymal stem cell therapy for heart disease. Vascul Pharmacol. (Journal Article; Research Support, Non-U.S. Gov’t).Gnecchi M Danieli P Cervio E 2012 57 1 48 55 - 43.
Mesenchymal stem cells overexpressing Akt dramatically repair infarcted myocardium and improve cardiac function despite infrequent cellular fusion or differentiation. Mol Ther. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Noiseux N Gnecchi M Lopez-ilasaca M Zhang L Solomon S. D Deb A et al 2006 14 6 840 50 - 44.
Mechanisms of action of mesenchymal stem cells in cardiac repair: potential influences on the cardiac stem cell niche. Nat Clin Pract Cardiovasc Med. (Journal Article; Research Support, N.I.H., Extramural; Research Support, Non-U.S. Gov’t).Mazhari R Hare J. M 2007 Suppl 1:S21 6 - 45.
Proteomic analysis of tumor necrosis factor-alpha-induced secretome of human adipose tissue-derived mesenchymal stem cells. J Proteome Res. (Journal Article; Research Support, Non-U.S. Gov’t).Lee M. J Kim J Kim M. Y Bae Y. S Ryu S. H Lee T. G et al 2010 9 4 1754 62 - 46.
Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. (Journal Article; Research Support, Non-U.S. Gov’t).Valadi H Ekstrom K Bossios A Sjostrand M Lee J. J Lotvall J. O 2007 9 6 654 9 - 47.
Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. (Journal Article; Research Support, Non-U.S. Gov’t).Alvarez-erviti L Seow Y Yin H Betts C Lakhal S Wood M. J 2011 29 4 341 5