Abstract
Synthetic patches and fixed grafts currently used in the repair of congenital heart defects are nonliving, noncontractile, and not electrically responsive, leading to increased risk of complication, reoperation, and sudden cardiac death. Studies suggest that tissue-engineered patches made from living, functional cells could grow with the patient, facilitate healing, and help recover cardiac function. In this paper, we review the research into possible sources of cardiomyocytes and other cardiac cells, including embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, adipose-derived stem cells, umbilical cord blood cells, amniotic fluid-derived stem cells, and cardiac progenitor cells. Each cell source has advantages, but also has technical hurdles to overcome, including heterogeneity, functional maturity, immunogenicity, and pathogenicity. Additionally, biomaterials used as patch materials will need to attract and support desired cells and induce minimal immune responses.
Keywords: stem cells, progenitor cells, congenital heart defects, cardiac tissue engineering
Congenital Heart Defects
Congenital heart defects (CHD) are the most common birth defects (3 per 1000 live births) and are the leading cause of birth defect-related death.1,2 CHD can range from life threatening to benign. Benign CHD, such as septal defects, the most common CHD (>50% of all CHD), are often underdiagnosed because of lack of symptoms,3 and only an estimated 20% of patients with septal defects will require surgical intervention.4 Other CHD, such as hypoplastic left heart syndrome (2.7% of all CHD) or Tetralogy of Fallot (4% of all CHD) have life-threatening symptoms and require surgical intervention.3 When treating patients with CHD, surgical repairs often require patches to close septal defects or enlarge stenosed structures, for example, ∼50% of Tetralogy of Fallot repairs include a patch for the right ventricular outflow tract.5 Currently, polymers (polytetrafluoroethylene [PTFE], Dacron, etc.), fixed xenografts (eg, bovine pericardium), and fixed pericardium autografts are the most common patch materials used in CHD repairs.6 However, because these materials are synthetic or treated to remove living cells, they do not grow with the patient nor are they electromechanically integrated; moreover, the patches may become fibrotic at the site of implantation and have mismatched mechanical properties compared to the surrounding tissue. As CHD are often treated early in development, additional surgeries are sometimes required to replace overgrown patches, and risks of arrhythmia and sudden cardiac death are increased in patients with patch implants.7,8 The 25-year survival rate of patients who underwent repair with patch implantation is 80% compared to 93% in patients who underwent repairs without patch implantation.9 Without cells, the patch cannot grow with the patient, potentially requiring further surgeries to replace it as it is outgrown; 14% of Tetralogy of Fallot patients require reoperation on the right ventricular outflow tract.10 The lack of contractility and mismatched mechanical properties could lead to loss of cardiac function and output. The lack of electrical conductivity could potentially cause a reentrant loop of depolarization around the patch area, leading to a self-sustained depolarization pattern, which could lead to arrhythmia, fibrillation, and further complications. The combination of these various shortcomings may contribute to the elevated risk of sudden cardiac death in patients implanted with patches, which is 25 to 100 times the norm.11 A tissue-engineered patch that contains cardiac cells with contractility and conductivity matching the native tissue could grow with the patient and recover cardiac function.12,13 This patch could be seeded with cardiac cells or cardiac progenitor cells (CPC), potentially derived from the patient’s own stem cells, in a bioengineered material designed to incorporate with native tissue and facilitate healing.
Cardiac Development and Homeostasis
Cardiomyocytes (CM) are derived from the mesodermal lineage through the interaction between several signaling factors, including activin, bone morphogenetic proteins (BMP), and transforming growth factor β (TGF-β).14,15 Through the activation of mesendoderm differentiation by activin, BMP signals, and inhibition of the canonical Wnt pathway, the cardiac lineage is derived, as indicated by the expression of Nkx2.5, an early cardiac transcription factor.14,15 Nkx2.5 further initiates the expression of another early cardiac transcription factor, GATA4, and these transcription factors upregulate the expression of key components of the CM, including cardiac actin filaments, myosin heavy chains, and cardiac troponin.14 Myofibrillogenesis is the formation and organization of the sarcomere during CM differentiation and maturation. Beginning in the precardiac mesoderm stage, actin and myosin filaments independently form and give rise to stress fibers. Initially, these fibers are composed mostly of nonmuscle actin and myosin subunits. As differentiation occurs, cardiac-specific actin and myosin subunits begin to replace their nonmuscle counterparts. Formation of anchoring proteins as well as filament alignment takes place as the cells begin to contract. As the cells mature, the myofilaments increase in alignment and most nonmuscle filament proteins are replaced.16
Supporting cells, including cardiac fibroblasts and connective tissues, are also important in maintaining cardiac functions. Cardiac fibroblasts comprise the largest population (>50%) of cells in the myocardium.17,18 They help stabilize cardiac tissue by maintaining extracellular matrix (ECM) homeostasis, providing electromechanical support, and participating in wound healing.19 Fibroblasts are responsible for secreting collagen types I, III, IV, and IV, generating the collagen-rich ECM that provides mechanical stability to cardiac tissues.17,20 Fibroblasts also produce growth factors that regulate cardiac cells, including TGF-β, angiotensin II, IL-6, and IL-1β.21 Cardiac fibroblasts provide passive resistive force to the cardiac tissue, and the electrical connection between cardiac fibroblasts and CM facilitates the propagation of action potentials throughout the cardiac tissue.17 Cardiac fibroblasts participate in wound healing responses such as fibrosis when ischemia or other damage occurs in the heart.22 Cardiac fibroblasts can be identified by their expression of discoidin domain receptor 2, a collagen receptor.17 Vascular tissues such as endothelial cells (EC) and smooth muscle cells (SMC) are also found in cardiac tissue; they are a small portion of the 70% non-CM cell population in the heart.17,18 They primarily participate in the vascularization necessary to support cardiac tissues, providing a pathway for oxygen, nutrients, and signaling cytokines.23
Application of Stem Cells for Chd Treatment
A tissue-engineered patch encapsulated with CM and support cells with properties and functions similar to native tissues would integrate with healthy tissues to restore functions while avoiding the risk of arrhythmia.24,25 However, because native CM do not normally proliferate, alternative sources for CM are needed. Several sources of CM have been investigated in current literature, including CM derived from embryonic stem cells (ESC),26,27 induced pluripotent stem cells (iPSC),28,29 mesenchymal stem cells (MSC),30 adipose-derived stem cells (ADSC),31 umbilical cord blood cells (UCBC),32 amniotic fluid-derived stem cells (AFSC),33 and CPC34–36 (Table 1). As potential sources of CM in a cardiac patch for pediatric CHD repair, the cells need to be minimally allergenic while being able to develop sufficient force generation and electrical coupling.
Table 1.
Summary of CM cell sources discussed, ESC, iPSC, MSC, ADSC, UCBC, AFSC, and CPC.
CELL TYPE | CHARACTERISTICS | CARDIOGENESIS CAPABILITY | SOURCES |
---|---|---|---|
ESC | Embroid body, pluripotent | Cardiogenesis induced by activin A and BMP 4 or small molecule inhibitors creates spontaneously contracting CM, maturing phenotype | 26 29 39 40 41 |
iPSC | Embroid body, pluripotent, can be generated autologouly, | Cardiogenesis induced by activin A and BMP 4 or small molecule inhibitors creates spontaneously contracting CM | 39 40 48 49 |
MSC | MSC lineage markers (CD29, CD44, CD71, CD90, CD106, CD120a, CD124, CD166), found in various niches including bone marrow and peripheral blood, can be isolated autologously | Cardiac marker expression, electromechanical connections, calcium flux, aid recovery of cardiac function post infarct | 30 64 65 66 |
ADSC | Identified in adipose tissue, express MSC lineage markers, can be isolated autologously | Cardiac marker expression, contractile cells | 74 76 |
UCBC | Identified in umbilical cord blood, can be isolated autologously | Aid recovery of cardiac function post infarct, endothelial cell source | 32 80 81 82 83 |
AFSC | Identified in amniotic fluid, express MSC lineage markers, c-Kit+, can be isolated autologously | Cardiac marker expression, electromechanical connections, endothelial cell source | 33 91 92 93 94 95 |
CPC | Identified by various markers (Isl-1, Flk-1, c-Kit, Nanog, Sca-1), can be generated through cardiosphere culture, found in side population cells, can be isolated autologously | Cardiac marker expression, spontaneously contracting CM in murine species, aid recovery of cardiac function post infarct | 34 35 36 106 114 115 |
Embryonic stem cells
ESC with the capability to differentiate into all three germ layers have been heavily investigated in the last two decades.37,38 Early research had shown that spontaneously beating CM can be found in embryoid body culture of ESC with low yield (<10%).26 Kattman et al showed that optimized treatment of activin A and BMP4 can differentiate ESC to beating CM with better yield (>50%).39 Recently, groups have shown that small molecule inhibitors of Wnt signaling can induce highly efficient (>85%) CM differentiation in embryonic-like stem cells.29,40 However, the leftover undifferentiated ESC could form teratomas.37,38 ESC-derived CM are functionally immature; myofilaments in ESC-derived CM do not align in a preferential direction, thus do not generate the level of force found in mature CM. However, Lundy et al recently reported that late-stage (80–120 days of differentiation) ESC-derived CM exhibited improved filament organization and force generation compared to early-stage (20–40 days of differentiation) ESC-derived CM.41 Additionally, fibrin gel containing CD15+, Isl1+ ESC-derived progenitor cells is currently being tested for use in ischemic heart failure recovery in an ongoing clinical trial.42,43 Variability between different lines of ESC may also affect the differentiation efficiency of ESC and cause heterogeneity in the resulting cells.44 Furthermore, ESC would not be autologous to the patient, resulting in risks of immune reactions.45
Induced pluripotent stem cells
iPSC were first generated from differentiated cells by the Yamanaka group with the transfection of a combination of transcription factors, including Oct3/4, Sox2, Nanog, Klf4, and Myc.46,47 These transcription factors act to reprogram differentiated cells to revert back to a pluripotent stage resembling ESC. iPSC reprogramed from human fibroblasts have been shown to possess the ability to form teratoma containing all three germ layers when injected in mice.47
iPSC have been shown to differentiate into CM through protocols similar to ESC-derived CM.48 Namely, iPSC can be induced to differentiate toward mesoderm lineage through inhibiting GSK3 or embryoid body formation. Further inhibition of the canonical Wnt signaling pathway in these mesoderm fated iPSC will lead to differentiation of functional CM. Recently, highly efficient CM differentiation protocols have been identified using both growth factor Wnt pathway manipulation39,49 and small molecule inhibition of the Wnt pathway.29,40 Introducing chemically defined small molecule inhibitors of Wnt at specific time points, the Palecek lab was able to induce greater than 85% pure iPSC-CM.40
iPSC are readily available from commercial sources, typically generated from fibroblasts.50 The advantage of iPSC over ESC is that iPSC could be generated autologously from one’s own tissue; iPSC can even be generated from cells isolated from patient’s amniotic fluid.51,52 Patient iPSC derived CM can also be used as in vitro models of cardiac disorders.53,54 However, iPSC pose the same potential for teratoma formation in patients if undifferentiated cells remain with the desired differentiated ones. While there are many advantages of personalized iPSC lines, patient to patient variability may be a concern, requiring additional consideration in controlling the differentiation efficiency and heterogeneity in the resulting cells. Additionally, studies of iPSC showed different immunogenic responses in their syngeneic hosts depending on the implantation site.55–57
Mesenchymal stem cells
MSC are adult stem cells identified by the expression of CD29, CD44, CD71, CD90, CD106, CD120a, CD124, and CD166 with the presence of integrins α1, α5, and β1.58–60 MSC have been isolated from various niches, including bone marrow and peripheral blood.61–63 The ability of MSC to differentiate into CM has been disputed. Studies have observed MSC expressing cardiac lineage proteins such as cardiac troponin and GATA4 when injected in cardiac infarct models.64,65 Groups have also shown that MSC could establish electromechanical connections with native cells in host cardiac tissue in animal models.30,66 Xu et al found rhythmic calcium flux and potential electrical activities in MSC cultures treated with 5-azacytidine to prevent DNA methylation.30 However, consistent generation of functional MSC-derived CM has yet to be shown. MSC have been shown to differentiate into endothelial and perivascular phenotypes and have been shown to improve cardiac function in animal cardiac infarct models.67–69 While MSC could be isolated autologously, the frequency and differentiation potential of MSC varies between patients and decreases with age and morbidity.70,71 While MSC were shown to be nonimmunogenic in experiments in vitro, recent studies have shown that MSC could still be immunogenic in vivo, suggesting that MSC may be less immunoprivileged than previous literatures suggested.57,72
Adipose-derived stem cells
ADSC are cells similar to MSC isolated from adipose tissue.73–75 ADSC can differentiate into various phenotypes similar to MSC.75 Mouse ADSC have been reported to generate spontaneous beating cells expressing cardiac genes.74 Human ADSC have been shown to upregulate cardiac genes when cultured on laminin76 and generate small populations of beating cells when exposed to rat CM extracts.31 ADSC could be isolated autologously. However, similar to MSC, the consistency and purity of ADSC are highly variable, and the methods for generating CM from ADSC have yet to demonstrate clinically relevant numbers.
Umbilical cord blood cells
UCBC are a group of heterogeneous cells found in the umbilical cord that have been shown to possess multipotent differentiation potential.77–79 Populations of UCBC have been shown to help recover cardiac function when implanted on infarction sites in animal models.80,81 UCBC can serve as an endothelial source.82,83 However, differentiating UCBC into CM phenotypes resulted in limited functioning CM.32 Owing to the heterogeneic nature of UCBC, strict selection methods are required to limit variability and improve reproducibility of results.79
Amniotic fluid-derived stem cells
Amniotic fluid (AF) provides protection for the developing embryo by dampening movements to the amniotic cavity. AF contains a mixture of several cell types from the fetus.84,85 A population of the cells found in the AF was found to express Oct-4, a transcription factor found in pluripotent stem cells.86,87 Another population of the cells found in the AF was identified to express c-kit (or CD117), a surface marker found in multipotent stem cells in adults.88 These c-kit positive cells, referred to as AFSC, have been shown to possess osteogenic, myogenic, and adipogenic potential similar to MSC, but do not form teratomas in mice.85,89
AFSC, as a potential therapeutic component, offer many advantages, especially in congenital defect repairs. When defects are diagnosed during pregnancy, AF could be collected and AFSC could be isolated. These AFSC would be autologous and pose lower risk of immune rejection than allogeneic sources. AFSC can be isolated from as early as the first trimester and expanded to clinically relevant numbers for perinatal or neonatal surgical applications.85 Methods of inducing CM differentiation in AFSC have been investigated, which include coculture mixing of AFSC with cardiac cells of various sources.33 When cocultured directly with neonatal rat ventricular myocytes (NRVM), AFSC have been shown to express cardiac troponin I.33 When cocultured indirectly with NRVM, AFSC have been shown to form functional gap junctions and improve cell–cell connectivity.90 AFSC have yet to be shown to generate spontaneously contracting CM.
AFSC can be differentiated into EC with the commercially available media Endothelial Growth Medium 2 (EGM-2) supplemented with basic Fibroblast Growth Factor (bFGF) or Vascular Endothelial Growth Factor (VEGF).91–93 A viable method of high-efficiency AFSC endothelial differentiation has been developed by a member of our lab94 and formed comparable vessel structures to human umbilical vein EC control in fibrin/PEG hydrogels.95
Cardiac progenitor cells
Even though most CM are terminally differentiated in the developed heart, efforts have been made to identify and study CPC. CPC are proliferating cells with the ability to differentiate into cardiac cells when triggered by cytokine signals or other factors.96,97 Some CPC are identified by their ability to elute Hoechst dye; these cells are referred to as “side population” cells.98,99 The surface markers Isl1,100,101 Flk-1,102 c-Kit,36 and Nanog103 were also used to identify CPC from human patient samples.100,101 CPC are found to be activated by electrical stimulation in vitro104 and cardiac infarction in adult patients.36 Promising methods of identifying and differentiating CPCs have been identified with rodent sources.105 Cardiac cells expressing Sca-1 have been identified in cardiac niches in neonatal and adult murine hearts that can aid in regenerating cardiac function post infarction and potentially differentiate into CM.36,106–108 Groups have even generated functional beating cells from murine CPC.108,109 Additionally, studies have reported that murine CPC can help improve cardiac functions in postinfarction models.110 However, experiments with human CPC have yet to show beating CM in vitro.
Alternatively, phase-bright cells migrating out of explants of cardiac tissue samples have also been shown to form dense colonies or cardiospheres.111 These cardiosphere-derived cells (CDC) contained potential CPC that expressed markers of pluripotency and were able to divide in in vitro and in vivo conditions.111 CDC were able to differentiate into multiple germ layers in vitro and even exhibited myocardial differentiation when implanted in rat heart.34 Mishra et al have shown that CDC can be isolated from samples taken from pediatric CHD patients.35 They noted that age correlated negatively with the amount of CDC harvest and their proliferation abilities.35
A study has identified a population of c-kit positive cardiac cells that has shown CM differentiation in infarcted areas.36 When isolated from human patients and injected into immune compromised mice, CPC were found to have developed connections to the native mouse CM and begin to differentiate into different cardiac lineages.112,113 Other studies with Hoechst stain eluding “side population” CPC also have shown cardiac differentiation capabilities.105 CDC containing CPC have been shown to be multipotent in vitro.34 Implanted CDC in a rodent cardiac infarct model have been shown to differentiate into CM, fibroblast, and vascular cells, and CDC derived CM were able to integrate with host CM.35 Compared to control, CDC implantation led to improved cardiac tissue regeneration and functional recovery.35 Matsuura et al have shown beating cells derived from mouse CPC cultures.108 The presence and quality of CPC decrease with age; therefore, CPC may be a more relevant source of generating autologous cardiac cells for pediatric applications as greater amount of CPC could be isolated from younger patients. A variety of niches for potential CPC have been identified. Therefore, extensive characterization and strict selection criteria must be established prior to clinically relevant application.114
Using Stem Cells in Conjunction with Biomaterials
Cell-loaded biomaterials
Stem cells could be incorporated into biomaterials and function as tissue replacements. Tissue-engineered systems can be tested in various in vivo models of cardiovascular defects such as the mouse angiogenesis model,115 rat Left Ventricle (LV) ischemia model,16 rabbit cardiac infarct model,116 baboon infarct model,117 and hemodialysis model.118 Engineered tissues could have passive functions: scaffolds containing bone marrow MSC have been tested as functional vascular grafts with successful implants in ongoing clinical trials12,119 and fibrin hydrogels containing skeletal myoblasts have been shown to reduce the thinning of the ventricular cell walls postinjury in a rat LV ischemia model.16 A vascular autograft composed of autologous fibroblast and EC has been shown to achieve 76% patency in patients in a clinical trial.120 Tissue-engineered grafts could also provide active functions. Shin et al examined a gelatin methacrylate gel containing carbon nanotube and seeded with NRVM reported that it was able to act as a bioactuator, generating forces in vitro when stimulated with electrical signals.121 Bio-materials loaded with stem cells could also be used as stabilizing or regenerative agents. Wang et al reported that injectable poly (ethylene glycol)-poly (caprolactone) copolymer (PEG-PCL) hydrogels containing bone marrow MSC have significantly higher vessel growth in the infarcted areas compared to control in a rabbit cardiac infarct model.116 However, because of the multipotent nature of stem cells and because biomaterials are derived from native tissue or containing heterogeneous natural molecules, the resulting material could have variable behavior.122 One method of reducing variability and toxicity of biomaterials is to use cells of definite phenotype or predifferentiated stem cells.123 At a cost of plasticity and differentiation potentials, the behavior of the material could be more controllable.
Biomaterials that recruit cells
Biomaterials can also be designed to recruit native cells. By adjusting the mechanical properties, such as elasticity, compressibility, density, and degradation rate, biomaterials could be selective in the adhesion and proliferation of specific types of cells.124,125 Recently, a silk-fibronectin hydrogel has been shown to recruit native endothelial population by enhancing the bioactivity of a porous synthetic material (PDLLA) by adding silk-fibronectin, resulting in increased vascular ingrowth.126 Biomaterials could also be designed to promote the migration and proliferation of specific types of cells through chemical means with preloaded growth factors, biomolecules, or cells.125,127 A MSC-loaded hydrogel has been shown to induce angiogenesis in vivo with significantly higher invading vessel segments compared to acellular controls when injected subcutaneously in a mouse model.115
Biomaterials conditioned with cells
Furthermore, bio-materials could be pretreated with cells, and then decellularized. The resulting material would benefit from the remodeling and ECM deposited by the cells, improving biocompatibility and cell recruiting abilities of the material. Removing the cells reduces the concern of controlling cell behaviors postimplantation and host immune responses. Syedain et al generated arterial grafts with tubular fibrin gels conditioned with human fibroblast and underwent mechanical stretching; the authors reported that the decellularized grafts withstood burst pressures at 50% or higher of ovine femoral arteries control.128 A study has shown that tissue-engineered fibrin heart valves conditioned with fibroblasts have similar mechanical properties compared to allograft controls and have improved recellularization potential in vitro.129 Weber et al reported that similar fibrin gel heart valves conditioned with human fibroblast and dynamically stretched showed improved in vivo recellularization in a baboon model compared to decellularized allograft controls.117 Dahl et al generated a tissue-engineered vascular graft with SMC cultured in PGA; after decellularization, the graft was examined in a baboon hemodialysis model and found to have 83% patency and lower T-cell adhesion compared to PTFE control after 24 weeks.118
Future Considerations
Several stem or progenitor cell sources have been investigated for their ability to generate cardiac cells. ESC and iPSC have been shown to generate functional CM.39,40,49 MSC expressed CM phenotype markers when injected in myocardium in in vivo models.30,64,65 CPC have been shown to express a CM phenotype in in vitro experiments, and murine CPC have been shown to generate spontaneously beating cells.34,35,108,109 Other stem cell sources, such as ADSC, UCBC, and AFSC, have been able to express CM phenotype markers when induced by mature CM in vitro or in vivo.31–33 Current studies have yet to produce CM with mature phenotype capable of generating force at the level of native CM.41 Generating CM at clinically relevant numbers remains another challenge.18 Purity and immunogenicity of the resulting CM should also be closely monitored.44,45 With respect to CHD and pediatric patients, one should also consider the lasting effects of materials and cells that may remain in the patients for years, which may increase risk of certain side effects such as the formation of teratomas in pluripotent stem cell-derived cells37,38 or the toxicity of degradation byproducts.130 The pediatric heart is also at a different stage developmentally compared to the adult heart, which may result in different immune responses and regenerative potentials.17,18 Material design and choice of cells will require consideration of these challenges. Designing biomaterials to improve the proliferation and function of stem cells should be considered.121,125 Conditioning biomaterials with stem cells is a different approach that has seen promising results recently.117,128,129 So far, promising results have been shown in generating cardiovascular tissues from various stem cell sources in combination with biomaterials; however, many challenges still have to be met in tissue engineering constructs for surgical repairs of CHD.
Footnotes
ACADEMIC EDITOR: Karen Pulford, Editor in Chief.
PEER REVIEW: Six peer reviewers contributed to the peer review report. Reviewers’ reports totaled 1,006 words, excluding any confidential comments to the academic editor.
FUNDING: Funding was provided by the American Heart Association (14BGIA18750004 to JGJ) and the National Science Foundation (CAREER CBET1055942 to JGJ). The authors confirm that the funder had no influence over the study design, content of the article, or selection of this journal.
COMPETING INTERESTS: Authors disclose no potential conflicts of interest.
Paper subject to independent expert blind peer review. All editorial decisions made by independent academic editor. Upon submission manuscript was subject to anti-plagiarism scanning. Prior to publication all authors have given signed confirmation of agreement to article publication and compliance with all applicable ethical and legal requirements, including the accuracy of author and contributor information, disclosure of competing interests and funding sources, compliance with ethical requirements relating to human and animal study participants, and compliance with any copyright requirements of third parties. This journal is a member of the Committee on Publication Ethics (COPE).
Author Contributions
Wrote the first draft of the manuscript: YG. Contributed to the writing of the manuscript: YG, JGJ. Agree with manuscript results and conclusions: YG, JGJ. Jointly developed the structure and arguments for the paper: YG, JGJ. Made critical revisions and approved final version: JGJ. Both authors reviewed and approved of the final manuscript.
REFERENCES
- 1.Hamilton BE, Hoyert DL, Martin JA, Strobino DM, Guyer B. Annual summary of vital statistics: 2010–2011. Pediatrics. 2013;131:548–58. doi: 10.1542/peds.2012-3769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Murphy S, Xu J, Kochanek K. Deaths: final data for 2010. [Accessed May 12, 2014];Natl Vital Stat Reports. 2013 60(4) Available at: http://www.birthbythenumbers.org/wp-content/uploads/2012/12/prelim-deaths-2011.pdf. [PubMed] [Google Scholar]
- 3.Hoffman JIE, Kaplan S. The incidence of congenital heart disease. J Am Coll Cardiol. 2002;39:1890–900. doi: 10.1016/s0735-1097(02)01886-7. [DOI] [PubMed] [Google Scholar]
- 4.Hoffman JIE, Kaplan S, Liberthson RR. Prevalence of congenital heart disease. Am Heart J. 2004;147:425–39. doi: 10.1016/j.ahj.2003.05.003. [DOI] [PubMed] [Google Scholar]
- 5.Murphy JG, Gersh BJ, Mair DD, et al. Long-term outcome in patients undergoing surgical repair of Tetralogy of Fallot. N Engl J Med. 1993;329:593–9. doi: 10.1056/NEJM199308263290901. [DOI] [PubMed] [Google Scholar]
- 6.Jonas R. Comprehensive Surgical Management of Congenital Heart Disease. London, UK: Hodder Arnold; 2004. [Google Scholar]
- 7.Du ZD, Hijazi ZM, Kleinman CS, Silverman NH, Larntz K. Comparison between transcatheter and surgical closure of secundum atrial septal defect in children and adults: results of a multicenter nonrandomized trial. J Am Coll Cardiol. 2002;39(11):1836–44. doi: 10.1016/s0735-1097(02)01862-4. [DOI] [PubMed] [Google Scholar]
- 8.Steeds RP, Oakley D. Predicting late sudden death from ventricular arrhythmia in adults following surgical repair of Tetralogy of Fallot. QJM. 2004;97:7–13. doi: 10.1093/qjmed/hch004. [DOI] [PubMed] [Google Scholar]
- 9.Klinner W, Reichart B, Pfaller M, Hatz R. Late results after correction of Tetralogy of Fallot necessitating outflow tract reconstruction. Comparison with results after correction without outflow tract patch. Thorac Cardiovasc Surg. 1984;32:244–7. doi: 10.1055/s-2007-1023394. [DOI] [PubMed] [Google Scholar]
- 10.Knott-Craig CJ, Elkins RC, Lane MM, Holz J, McCue C, Ward KE. A 26-year experience with surgical management of Tetralogy of Fallot: Risk analysis for mortality or late reintervention. Ann Thorac Surg. 1998;66:506–11. doi: 10.1016/s0003-4975(98)00493-7. [DOI] [PubMed] [Google Scholar]
- 11.Silka MJ, Hardy BG, Menashe VD, Morris CD. A population-based prospective evaluation of risk of sudden cardiac death after operation for common congenital heart defects. J Am Coll Cardiol. 1998;32:245–51. doi: 10.1016/s0735-1097(98)00187-9. [DOI] [PubMed] [Google Scholar]
- 12.Shin’oka T, Matsumura G, Hibino N, et al. Midterm clinical result of tissue- engineered vascular autografts seeded with autologous bone marrow cells. J Thorac Cardiovasc Surg. 2005;129:1330–8. doi: 10.1016/j.jtcvs.2004.12.047. [DOI] [PubMed] [Google Scholar]
- 13.Sakai T, Li RK, Weisel RD, et al. The fate of a tissue-engineered cardiac graft in the right ventricular outflow tract of the rat. J Thorac Cardiovasc Surg. 2001;121:932–42. doi: 10.1067/mtc.2001.113600. [DOI] [PubMed] [Google Scholar]
- 14.Kirby ML, Waldo K. Cardiac Development. New York, NY: Oxford University Press; 2007. [Google Scholar]
- 15.Sadler TW. Langman’s Essential Medical Embryology. Philadelphia: Lippincott Williams & Wilkins; 2005. p. 150. [Google Scholar]
- 16.Christman K, Fok H. Fibrin glue alone and skeletal myoblasts in a fibrin scaffold preserve cardiac function after myocardial infarction. Tissue Eng. 2004;10(3) doi: 10.1089/107632704323061762. Available at: http://online.liebertpub.com/ [DOI] [PubMed] [Google Scholar]
- 17.Camelliti P, Borg TK, Kohl P. Structural and functional characterisation of cardiac fibroblasts. Cardiovasc Res. 2005;65:40–51. doi: 10.1016/j.cardiores.2004.08.020. [DOI] [PubMed] [Google Scholar]
- 18.Adler CP, Costabel U. Cell number in human heart in atrophy, hypertrophy, and under the influence of cytostatics. Recent Adv Stud Cardiac Struct Metab. 1975;6:343–55. [PubMed] [Google Scholar]
- 19.Brown RD, Ambler SK, Mitchell MD, Long CS. The cardiac fibroblast: therapeutic target in myocardial remodeling and failure. Annu Rev Pharmacol Toxicol. 2005;45:657–87. doi: 10.1146/annurev.pharmtox.45.120403.095802. [DOI] [PubMed] [Google Scholar]
- 20.Brilla CG, Zhou G, Matsubara L, Weber KT. Collagen metabolism in cultured adult rat cardiac fibroblasts: response to angiotensin II and aldosterone. J Mol Cell Cardiol. 1994;26:809–20. doi: 10.1006/jmcc.1994.1098. [DOI] [PubMed] [Google Scholar]
- 21.Souders CA, Bowers SLK, Baudino TA. Cardiac fibroblast: the renaissance cell. Circ Res. 2009;105:1164–76. doi: 10.1161/CIRCRESAHA.109.209809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Frangogiannis NG, Smith CW, Entman ML. The inflammatory response in myocardial infarction. Cardiovasc Res. 2002;53:31–47. doi: 10.1016/s0008-6363(01)00434-5. [DOI] [PubMed] [Google Scholar]
- 23.Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–57. doi: 10.1038/35025220. [DOI] [PubMed] [Google Scholar]
- 24.Zisch AH, Lutolf MP, Hubbell JA. Biopolymeric delivery matrices for angiogenic growth factors. Cardiovasc Pathol. 2003;12:295–310. doi: 10.1016/s1054-8807(03)00089-9. [DOI] [PubMed] [Google Scholar]
- 25.Vallée JP, Hauwel M, Lepetit-Coiffé M, et al. Embryonic stem cell-based cardiopatches improve cardiac function in infarcted rats. Stem Cells Transl Med. 2012;1:248–60. doi: 10.5966/sctm.2011-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kehat I, Kenyagin-Karsenti D, Snir M, et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108:407–14. doi: 10.1172/JCI12131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Xu C. Characterization and enrichment of cardiomyocytes derived from human embryonic stem cells. Circ Res. 2002;91:501–8. doi: 10.1161/01.res.0000035254.80718.91. [DOI] [PubMed] [Google Scholar]
- 28.Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
- 29.Zhang J, Wilson GF, Soerens AG, et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009;104:e30–41. doi: 10.1161/CIRCRESAHA.108.192237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Xu W, Zhang X, Qian H, et al. Mesenchymal stem cells from adult human bone marrow differentiate into a cardiomyocyte phenotype in vitro. Exp Biol Med (Maywood) 2004;229:623–31. doi: 10.1177/153537020422900706. [DOI] [PubMed] [Google Scholar]
- 31.Gaustad KG, Boquest AC, Anderson BE, Gerdes AM, Collas P. Differentiation of human adipose tissue stem cells using extracts of rat cardiomyocytes. Biochem Biophys Res Commun. 2004;314:420–7. doi: 10.1016/j.bbrc.2003.12.109. [DOI] [PubMed] [Google Scholar]
- 32.Martin-Rendon E, Sweeney D, Lu F, Girdlestone J, Navarrete C, Watt SM. 5-Azacytidine-treated human mesenchymal stem/progenitor cells derived from umbilical cord, cord blood and bone marrow do not generate cardiomyocytes in vitro at high frequencies. Vox Sang. 2008;95:137–48. doi: 10.1111/j.1423-0410.2008.01076.x. [DOI] [PubMed] [Google Scholar]
- 33.Chiavegato A, Bollini S, Pozzobon M, et al. Human amniotic fluid-derived stem cells are rejected after transplantation in the myocardium of normal, ischemic, immuno-suppressed or immuno-deficient rat. J Mol Cell Cardiol. 2007;42(4):746–59. doi: 10.1016/j.yjmcc.2006.12.008. [DOI] [PubMed] [Google Scholar]
- 34.Itzhaki-Alfia A, Leor J, Raanani E, et al. Patient characteristics and cell source determine the number of isolated human cardiac progenitor cells. Circulation. 2009;120:2559–66. doi: 10.1161/CIRCULATIONAHA.109.849588. [DOI] [PubMed] [Google Scholar]
- 35.Mishra R, Vijayan K, Colletti EJ, et al. Characterization and functionality of cardiac progenitor cells in congenital heart patients. Circulation. 2011;123:364–73. doi: 10.1161/CIRCULATIONAHA.110.971622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Oh H, Bradfute SB, Gallardo TD, et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. 2003;100:12313–8. doi: 10.1073/pnas.2132126100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol. 2000;18:399–404. doi: 10.1038/74447. [DOI] [PubMed] [Google Scholar]
- 38.Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–7. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
- 39.Kattman SJ, Witty AD, Gagliardi M, et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell. 2011;8:228–40. doi: 10.1016/j.stem.2010.12.008. [DOI] [PubMed] [Google Scholar]
- 40.Lian X, Hsiao C, Wilson G, et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci U S A. 2012;109:E1848–57. doi: 10.1073/pnas.1200250109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lundy SD, Zhu W-Z, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Puymirat E, Geha R, Tomescot A, et al. Can mesenchymal stem cells induce tolerance to cotransplanted human embryonic stem cells? Mol Ther. 2009;17:176–82. doi: 10.1038/mt.2008.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Assistance Publique – Hôpitaux de Paris Transplantation of human embryonic stem cell-derived progenitors in severe heart failure (ESCORT) Clin Bethesda Natl Libr Med. 2013. [Accessed February 20, 2015]. Available at: https://clinicaltrials.gov/ct2/show/NCT02057900?term=NCT02057900&rank=1.
- 44.Osafune K, Caron L, Borowiak M, et al. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol. 2008;26:313–5. doi: 10.1038/nbt1383. [DOI] [PubMed] [Google Scholar]
- 45.Drukker M, Katz G, Urbach A, et al. Characterization of the expression of MHC proteins in human embryonic stem cells. Proc Natl Acad Sci U S A. 2002;99:9864–9. doi: 10.1073/pnas.142298299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322:949–53. doi: 10.1126/science.1164270. [DOI] [PubMed] [Google Scholar]
- 47.Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
- 48.Mauritz C, Schwanke K, Reppel M, et al. Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation. 2008;118:507–17. doi: 10.1161/CIRCULATIONAHA.108.778795. [DOI] [PubMed] [Google Scholar]
- 49.Narazaki G, Uosaki H, Teranishi M, et al. Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells. Circulation. 2008;118:498–506. doi: 10.1161/CIRCULATIONAHA.108.769562. [DOI] [PubMed] [Google Scholar]
- 50.Lancaster MA, Renner M, Martin C, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2012;501:373–9. doi: 10.1038/nature12517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Li C, Zhou J, Shi G, et al. Pluripotency can be rapidly and efficiently induced in human amniotic fluid-derived cells. Hum Mol Genet. 2009;18:4340–9. doi: 10.1093/hmg/ddp386. [DOI] [PubMed] [Google Scholar]
- 52.Galende E, Karakikes I, Edelmann L, et al. Amniotic fluid cells are more efficiently reprogrammed to pluripotency than adult cells. Cell Reprogram. 2010;12:117–25. doi: 10.1089/cell.2009.0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Josowitz R, Carvajal-Vergara X, Lemischka IR, Gelb BD. Induced pluripotent stem cell-derived cardiomyocytes as models for genetic cardiovascular disorders. Curr Opin Cardiol. 2011;26:223–9. doi: 10.1097/HCO.0b013e32834598ad. [DOI] [PubMed] [Google Scholar]
- 54.Matsa E, Rajamohan D, Dick E, et al. Drug evaluation in cardiomyocytes derived from human induced pluripotent stem cells carrying a long QT syndrome type 2 mutation. Eur Heart J. 2011;32:952–62. doi: 10.1093/eurheartj/ehr073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Fu X. The immunogenicity of cells derived from induced pluripotent stem cells. Cell Mol Immunol. 2014;11:14–6. doi: 10.1038/cmi.2013.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhao T, Zhang Z-N, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature. 2011;474:212–5. doi: 10.1038/nature10135. [DOI] [PubMed] [Google Scholar]
- 57.Mukonoweshuro B, Brown CJ, Fisher J, Ingham E. Immunogenicity of undifferentiated and differentiated allogeneic mouse mesenchymal stem cells. J Tissue Eng. 2014;5 doi: 10.1177/2041731414534255. 2041731414534255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36:568–84. doi: 10.1016/j.biocel.2003.11.001. [DOI] [PubMed] [Google Scholar]
- 59.Pittenger MF. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
- 60.Majumdar MK, Keane-Moore M, Buyaner D, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci. 2003;10:228–41. doi: 10.1007/BF02256058. [DOI] [PubMed] [Google Scholar]
- 61.Kadner A, Hoerstrup SP, Zund G, et al. A new source for cardiovascular tissue engineering: human bone marrow stromal cells. Eur J Cardiothorac Surg. 2002;21:1055–60. doi: 10.1016/s1010-7940(02)00079-9. [DOI] [PubMed] [Google Scholar]
- 62.Perry TE, Kaushal S, Sutherland FW, et al. Bone marrow as a cell source for tissue engineering heart valves. Ann Thorac Surg. 2003;75:761–7. doi: 10.1016/s0003-4975(02)03776-1. [DOI] [PubMed] [Google Scholar]
- 63.Roufosse CA, Direkze NC, Otto WR, Wright NA. Circulating mesenchymal stem cells. Int J Biochem Cell Biol. 2004;36:585–97. doi: 10.1016/j.biocel.2003.10.007. [DOI] [PubMed] [Google Scholar]
- 64.Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105:93–8. doi: 10.1161/hc0102.101442. [DOI] [PubMed] [Google Scholar]
- 65.Kawada H, Fujita J, Kinjo K, et al. Nonhematopoietic mesenchymal stem cells can be mobilized and differentiate into cardiomyocytes after myocardial infarction. Blood. 2004;104:3581–7. doi: 10.1182/blood-2004-04-1488. [DOI] [PubMed] [Google Scholar]
- 66.Labovsky V, Hofer EL, Feldman L, et al. Cardiomyogenic differentiation of human bone marrow mesenchymal cells: role of cardiac extract from neonatal rat cardiomyocytes. Differentiation. 2010;79:93–101. doi: 10.1016/j.diff.2009.10.001. [DOI] [PubMed] [Google Scholar]
- 67.Gojo S, Gojo N, Takeda Y, et al. In vivo cardiovasculogenesis by direct injection of isolated adult mesenchymal stem cells. Exp Cell Res. 2003;288:51–9. doi: 10.1016/s0014-4827(03)00132-0. [DOI] [PubMed] [Google Scholar]
- 68.Silva GV, Litovsky S, Assad JA, et al. Mesenchymal stem cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a canine chronic ischemia model. Circulation. 2005;111:150–6. doi: 10.1161/01.CIR.0000151812.86142.45. [DOI] [PubMed] [Google Scholar]
- 69.Gnecchi M, He H, Noiseux N, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J. 2006;20:661–9. doi: 10.1096/fj.05-5211com. [DOI] [PubMed] [Google Scholar]
- 70.Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. Stem Cells. 2003;21:105–10. doi: 10.1634/stemcells.21-1-105. [DOI] [PubMed] [Google Scholar]
- 71.D’Ippolito G, Schiller PC, Ricordi C, Roos BA, Howard GA. Age-related osteogenic potential of mesenchymal stromal stem cells from human vertebral bone marrow. J Bone Miner Res. 1999;14:1115–22. doi: 10.1359/jbmr.1999.14.7.1115. [DOI] [PubMed] [Google Scholar]
- 72.Sudres M, Norol F, Trenado A, et al. Bone marrow mesenchymal stem cells suppress lymphocyte proliferation in vitro but fail to prevent graft-versus-host disease in mice. J Immunol. 2006;176:7761–7. doi: 10.4049/jimmunol.176.12.7761. [DOI] [PubMed] [Google Scholar]
- 73.Fraser JK, Zhu M, Wulur I, Alfonso Z. Adipose-derived stem cells. Methods Mol Biol. 2008;449:59–67. doi: 10.1007/978-1-60327-169-1_4. [DOI] [PubMed] [Google Scholar]
- 74.Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100:1249–60. doi: 10.1161/01.RES.0000265074.83288.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C. Adipose-derived stem cells: isolation, expansion and differentiation. Methods. 2008;45:115–20. doi: 10.1016/j.ymeth.2008.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Van Dijk A, Niessen HWM, Zandieh Doulabi B, Visser FC, Van Milligen FJ. Differentiation of human adipose-derived stem cells towards cardiomyocytes is facilitated by laminin. Cell Tissue Res. 2008;334:457–67. doi: 10.1007/s00441-008-0713-6. [DOI] [PubMed] [Google Scholar]
- 77.Newcomb JD, Willing AE, Sanberg PR. Umbilical cord blood cells. Methods Mol Biol. 2009;549:119–36. doi: 10.1007/978-1-60327-931-4_9. [DOI] [PubMed] [Google Scholar]
- 78.Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109:235–42. doi: 10.1046/j.1365-2141.2000.01986.x. [DOI] [PubMed] [Google Scholar]
- 79.Broxmeyer HE, Douglas GW, Hangoc G, et al. Human umbilical cord blood as a potential source of transplantable hematopoietic stem/progenitor cells. Proc Natl Acad Sci U S A. 1989;86:3828–32. doi: 10.1073/pnas.86.10.3828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hirata Y, Sata M, Motomura N, et al. Human umbilical cord blood cells improve cardiac function after myocardial infarction. Biochem Biophys Res Commun. 2005;327:609–14. doi: 10.1016/j.bbrc.2004.12.044. [DOI] [PubMed] [Google Scholar]
- 81.Leor J, Guetta E, Feinberg MS, et al. Human umbilical cord blood-derived CD133+ cells enhance function and repair of the infarcted myocardium. Stem Cells. 2006;24:772–80. doi: 10.1634/stemcells.2005-0212. [DOI] [PubMed] [Google Scholar]
- 82.Ingram DA, Mead LE, Tanaka H, et al. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood. 2004;104:2752–60. doi: 10.1182/blood-2004-04-1396. [DOI] [PubMed] [Google Scholar]
- 83.Murohara T, Ikeda H, Duan J, et al. Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. J Clin Invest. 2000;105:1527–36. doi: 10.1172/JCI8296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Bossolasco P, Montemurro T, Cova L, et al. Molecular and phenotypic characterization of human amniotic fluid cells and their differentiation potential. Cell Res. 2006;16:329–36. doi: 10.1038/sj.cr.7310043. [DOI] [PubMed] [Google Scholar]
- 85.De Coppi P, Bartsch G, Jr, Siddiqui MM, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007;25(1):100–6. doi: 10.1038/nbt1274. [DOI] [PubMed] [Google Scholar]
- 86.Seimiya M, Watanabe Y, Kurosawa Y. Identification of POU-class homeobox genes in a freshwater sponge and the specific expression of these genes during differentiation. Eur J Biochem. 1997;243:27–31. doi: 10.1111/j.1432-1033.1997.0027a.x. [DOI] [PubMed] [Google Scholar]
- 87.Prusa AR. Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Hum Reprod. 2003;18:1489–93. doi: 10.1093/humrep/deg279. [DOI] [PubMed] [Google Scholar]
- 88.Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of Kit-ligand. Cell. 2002;109:625–37. doi: 10.1016/s0092-8674(02)00754-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Tsai MS, Lee JL, Chang YJ, Hwang SM. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod. 2004;19:1450–6. doi: 10.1093/humrep/deh279. [DOI] [PubMed] [Google Scholar]
- 90.Connell JP, Augustini E, Moise KJ, Johnson A, Jacot JG. Formation of functional gap junctions in amniotic fluid-derived stem cells induced by transmembrane coculture with neonatal rat cardiomyocytes. J Cell Mol Med. 2013;17(6):774–81. doi: 10.1111/jcmm.12056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Delo DM, De Coppi P, Bartsch G, Atala A. Amniotic fluid and placental stem cells. Methods Enzymol. 2006;419:426–38. doi: 10.1016/S0076-6879(06)19017-5. [DOI] [PubMed] [Google Scholar]
- 92.Yeh Y-C, Wei H-J, Lee W-Y, et al. Cellular cardiomyoplasty with human amniotic fluid stem cells: in vitro and in vivo studies. Tissue Eng Part A. 2010;16:1925–36. doi: 10.1089/ten.TEA.2009.0728. [DOI] [PubMed] [Google Scholar]
- 93.Zhang P, Baxter J, Vinod K, Tulenko TN, Di Muzio PJ. Endothelial differentiation of amniotic fluid-derived stem cells: synergism of biochemical and shear force stimuli. Stem Cells Dev. 2009;18:1299–308. doi: 10.1089/scd.2008.0331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Benavides OM, Petsche JJ, Moise KJ, Johnson A, Jacot JG. Evaluation of endothelial cells differentiated from amniotic fluid-derived stem cells. Tissue Eng Part A. 2012;18:1123–31. doi: 10.1089/ten.tea.2011.0392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Benavides OM, Quinn JP, Pok S, Petsche Connell J, Ruano R, Jacot JG. Capillary- like network formation by human amniotic fluid-derived stem cells within fibrin/poly(ethylene glycol) hydrogels. Tissue Eng Part A. 2015;21:1185–94. doi: 10.1089/ten.tea.2014.0288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bollini S, Smart N, Riley PR. Resident cardiac progenitor cells: at the heart of regeneration. J Mol Cell Cardiol. 2011;50:296–303. doi: 10.1016/j.yjmcc.2010.07.006. [DOI] [PubMed] [Google Scholar]
- 97.Unno K, Jain M, Liao R. Cardiac side population cells: moving toward the center stage in cardiac regeneration. Circ Res. 2012;110:1355–63. doi: 10.1161/CIRCRESAHA.111.243014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Alfakir M, Dawe N, Eyre R, et al. The temporal and spatial expression patterns of ABCG2 in the developing human heart. Int J Cardiol. 2012;156:133–8. doi: 10.1016/j.ijcard.2010.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sandstedt J, Jonsson M, Kajic K, et al. Left atrium of the human adult heart contains a population of side population cells. Basic Res Cardiol. 2012;107:255. doi: 10.1007/s00395-012-0255-7. [DOI] [PubMed] [Google Scholar]
- 100.Cai CL, Liang X, Shi Y, et al. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev Cell. 2003;5:877–89. doi: 10.1016/s1534-5807(03)00363-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Moretti A, Caron L, Nakano A, et al. Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell. 2006;127:1151–65. doi: 10.1016/j.cell.2006.10.029. [DOI] [PubMed] [Google Scholar]
- 102.Kattman SJ, Huber TL, Keller GM. Multipotent Flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. Dev Cell. 2006;11:723–32. doi: 10.1016/j.devcel.2006.10.002. [DOI] [PubMed] [Google Scholar]
- 103.Otaka S, Nagura S, Koike C, et al. Selective isolation of nanog-positive human amniotic mesenchymal cells and differentiation into cardiomyocytes. Cell Reprogram. 2013;15:80–91. doi: 10.1089/cell.2012.0028. [DOI] [PubMed] [Google Scholar]
- 104.van Vliet P, de Boer TP, van der Heyden MA, et al. Hyperpolarization induces differentiation in human cardiomyocyte progenitor cells. Stem Cell Rev Reports. 2010;6:178–85. doi: 10.1007/s12015-010-9142-5. [DOI] [PubMed] [Google Scholar]
- 105.Davis DR, Kizana E, Terrovitis J, et al. Isolation and expansion of functionally-competent cardiac progenitor cells directly from heart biopsies. J Mol Cell Cardiol. 2010;49:312–21. doi: 10.1016/j.yjmcc.2010.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Uchida S, De Gaspari P, Kostin S, et al. Sca1-derived cells are a source of myocardial renewal in the murine adult heart. Stem Cell Reports. 2013;1:397–410. doi: 10.1016/j.stemcr.2013.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ma X, Robin C, Ottersbach K, Dzierzak E. The Ly-6 A (Sca-1) GFP transgene is expressed in all adult mouse hematopoietic stem cells. Stem Cells. 2002;20:514–21. doi: 10.1634/stemcells.20-6-514. [DOI] [PubMed] [Google Scholar]
- 108.Matsuura K, Nagai T, Nishigaki N, et al. Adult cardiac Sca-1-positive cells differentiate into beating cardiomyocytes. J Biol Chem. 2004;279:11384–91. doi: 10.1074/jbc.M310822200. [DOI] [PubMed] [Google Scholar]
- 109.Ott HC, Matthiesen TS, Brechtken J, et al. The adult human heart as a source for stem cells: repair strategies with embryonic-like progenitor cells. Nat Clin Pract Cardiovasc Med. 2007;4(suppl 1):S27–39. doi: 10.1038/ncpcardio0771. [DOI] [PubMed] [Google Scholar]
- 110.Padin-Iruegas ME, Misao Y, Davis ME, et al. Cardiac progenitor cells and biotinylated insulin-like growth factor-1 nanofibers improve endogenous and exogenous myocardial regeneration after infarction. Circulation. 2009;120:876–87. doi: 10.1161/CIRCULATIONAHA.109.852285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Smith RR, Barile L, Cho HC, et al. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007;115:896–908. doi: 10.1161/CIRCULATIONAHA.106.655209. [DOI] [PubMed] [Google Scholar]
- 112.Forte G, Pietronave S, Nardone G, et al. Human cardiac progenitor cell grafts as unrestricted source of supernumerary cardiac cells in healthy murine hearts. Stem Cells. 2011;29:2051–61. doi: 10.1002/stem.763. [DOI] [PubMed] [Google Scholar]
- 113.Laflamme MA, Myerson D, Saffitz JE, Murry CE. Evidence for cardiomyocyte repopulation by extracardiac progenitors in transplanted human hearts. Circ Res. 2002;90:634–40. doi: 10.1161/01.res.0000014822.62629.eb. [DOI] [PubMed] [Google Scholar]
- 114.Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76. doi: 10.1016/s0092-8674(03)00687-1. [DOI] [PubMed] [Google Scholar]
- 115.Frescaline G, Bouderlique T, Mansoor L, et al. Glycosaminoglycan mimetic associated to human mesenchymal stem cell based scaffolds inhibit ectopic bone formation but induce angiogenesis in vivo. Tissue Eng Part A. 2013;19:1–39. doi: 10.1089/ten.tea.2012.0377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wang T, Jiang XJ, Tang QZ, et al. Bone marrow stem cells implantation with α-cyclodextrin/MPEG—PCL—MPEG hydrogel improves cardiac function after myocardial infarction. Acta Biomater. 2009;5:2939–44. doi: 10.1016/j.actbio.2009.04.040. [DOI] [PubMed] [Google Scholar]
- 117.Weber B, Dijkman PE, Scherman J, et al. Off-the-shelf human decellularized tissue-engineered heart valves in a non-human primate model. Biomaterials. 2013;34:7269–80. doi: 10.1016/j.biomaterials.2013.04.059. [DOI] [PubMed] [Google Scholar]
- 118.Dahl SL, Kypson AP, Lawson JH, et al. Readily available tissue-engineered vascular grafts. Sci Transl Med. 2011;3:68ra9. doi: 10.1126/scitranslmed.3001426. [DOI] [PubMed] [Google Scholar]
- 119.Matsumura G, Hibino N, Ikada Y, Kurosawa H, Shin’oka T. Successful application of tissue engineered vascular autografts: clinical experience. Biomaterials. 2003;24:2303–8. doi: 10.1016/s0142-9612(03)00043-7. [DOI] [PubMed] [Google Scholar]
- 120.McAllister TN, Maruszewski M, Garrido SA, et al. Effectiveness of haemodialysis access with an autologous tissue-engineered vascular graft: a multicentre cohort study. Lancet. 2009;373:1440–6. doi: 10.1016/S0140-6736(09)60248-8. [DOI] [PubMed] [Google Scholar]
- 121.Shin SR, Jung SM, Zalabany M, et al. Carbon-nanotube-embedded hydrogel sheets for engineering cardiac constructs and bioactuators. ACS Nano. 2013;7:2369–80. doi: 10.1021/nn305559j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Singelyn JM, Sundaramurthy P, Johnson TD, et al. Catheter-deliverable hydrogel derived from decellularized ventricular extracellular matrix increases endogenous cardiomyocytes and preserves cardiac function post-myocardial infarction. J Am Coll Cardiol. 2012;59:751–63. doi: 10.1016/j.jacc.2011.10.888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Martino S, D’Angelo F, Armentano I, Kenny JM, Orlacchio A. Stem cell-biomaterial interactions for regenerative medicine. Biotechnol Adv. 2012;30:338–51. doi: 10.1016/j.biotechadv.2011.06.015. [DOI] [PubMed] [Google Scholar]
- 124.Lv S, Dudek DM, Cao Y, Balamurali MM, Gosline J, Li H. Designed biomaterials to mimic the mechanical properties of muscles. Nature. 2010;465:69–73. doi: 10.1038/nature09024. [DOI] [PubMed] [Google Scholar]
- 125.Rice JJ, Martino MM, De Laporte L, Tortelli F, Briquez PS, Hubbell JA. Engineering the regenerative microenvironment with biomaterials. Adv Healthc Mater. 2013;2:57–71. doi: 10.1002/adhm.201200197. [DOI] [PubMed] [Google Scholar]
- 126.Stoppato M, Stevens HY, Carletti E, Migliaresi C, Motta A, Guldberg RE. Effects of silk fibroin fiber incorporation on mechanical properties, endothelial cell colonization and vascularization of PDLLA scaffolds. Biomaterials. 2013;34:4573–81. doi: 10.1016/j.biomaterials.2013.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Hubbell JA. Biomaterials in tissue engineering. 0. 1995;13:565–76. doi: 10.1038/nbt0695-565. [DOI] [PubMed] [Google Scholar]
- 128.Syedain ZH, Meier LA, Bjork JW, Lee A, Tranquillo RT. Implantable arterial grafts from human fibroblasts and fibrin using a multi-graft pulsed flow-stretch bioreactor with noninvasive strength monitoring. Biomaterials. 2011;32:714–22. doi: 10.1016/j.biomaterials.2010.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Syedain Z, Bradee A, Kren S, Taylor DA, Tranquillo RT. Decellularized tissue-engineered heart valve leaflets with recellularization potential. Tissue Eng Part A. 2012;19(5–6):759–69. doi: 10.1089/ten.tea.2012.0365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Davis ME, Hsieh PCH, Grodzinsky AJ, Lee RT. Custom design of the cardiac microenvironment with biomaterials. Circ Res. 2005;97:8–15. doi: 10.1161/01.RES.0000173376.39447.01. [DOI] [PMC free article] [PubMed] [Google Scholar]