Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 Cardiac Diseases modeling using Human Induced Pluripotent Stem Cells-derived cardiomyocytes + 6006 word(s) 6006 2020-06-23 09:54:58 |
2 Cardiac Diseases modeling using Human Induced Pluripotent Stem Cells Meta information modification 6006 2020-06-24 18:27:45 | |
3 Cardiac Diseases modeling using Human Induced Pluripotent Stem Cells-derived cardiomyocytes Meta information modification 6006 2020-06-24 18:30:04 | |
4 Cardiac Diseases modeling using Human Induced Pluripotent Stem Cells-derived cardiomyocytes -2968 word(s) 3038 2020-06-24 18:38:05 | |
5 update layout and reference Meta information modification 3038 2020-06-30 11:29:24 | |
6 Increase the exposure -9 word(s) 3029 2020-10-26 10:23:00 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Parrotta, E.I.; Lucchino, V.; Scaramuzzino, L.; Scalise, S.; Cuda, G. Cardiovascular Diseases and Stem Cells. Encyclopedia. Available online: https://encyclopedia.pub/entry/1188 (accessed on 29 March 2024).
Parrotta EI, Lucchino V, Scaramuzzino L, Scalise S, Cuda G. Cardiovascular Diseases and Stem Cells. Encyclopedia. Available at: https://encyclopedia.pub/entry/1188. Accessed March 29, 2024.
Parrotta, Elvira Immacolata, Valeria Lucchino, Luana Scaramuzzino, Stefania Scalise, Giovanni Cuda. "Cardiovascular Diseases and Stem Cells" Encyclopedia, https://encyclopedia.pub/entry/1188 (accessed March 29, 2024).
Parrotta, E.I., Lucchino, V., Scaramuzzino, L., Scalise, S., & Cuda, G. (2020, June 24). Cardiovascular Diseases and Stem Cells. In Encyclopedia. https://encyclopedia.pub/entry/1188
Parrotta, Elvira Immacolata, et al. "Cardiovascular Diseases and Stem Cells." Encyclopedia. Web. 24 June, 2020.
Cardiovascular Diseases and Stem Cells
Edit

This entry provides an update on previous and current research in the field of Cardiovascular diseases (CVDs), a class of disorders affecting the heart or blood vessels. Despite progress in clinical research and therapy, CVDs still represent the leading cause of mortality and morbidity worldwide. The hallmarks of cardiac diseases include inflammation, fibrosis, scar tissue, hyperplasia, hypertrophy, abnormal ventricular remodeling, and cardiomyocyte death, which is an irreversible process that induces heart failure with progressive and dramatic consequences. Both genetic and environmental factors pathologically contribute to the development of CVDs, but the precise causes that trigger cardiac diseases and their progression are still largely unknown. In this scenario, the possibility to generate patient-specific cardiac cells from induced pluripotent stem cells (iPSCs) represents a powerful platform for the investigation of these life-threatening disorders.

cardiovascular diseases human induced pluripotent stem cells cardiac differentiation iPSC-derived cardiomyocytes cardiac disease modeling

1. Discussion

Cardiovascular diseases (CVDs) include a number of disorders affecting the heart functionality many of which entail a massive loss of cardiomyocytes, leading to heart attack, stroke or even death. Thus, the investigation of CVDs pathophysiology is crucial for the discovery of new curative therapies alternative to heart transplantation. Stem cell-based therapy for CVDs is one of the most promising approaches as it provides important molecular understanding of CVD’s mechanisms and bears many advantages for the identification of personalized, cell-based, therapies. Cardiomyocytes derived from patient-specific human induced pluripotent stem cells (hiPSCs-CMs) have the quality to carry the same mutations of the patient’s cardiac cells, representing the best platform for the creation of cardiac “disease-in-a-dish” models which are advantageous for in vitro high throughput drug screening, tissue engineering and regenerative medicine.

2. Introduction

Cardiovascular diseases (CVDs) are the leading cause of mortality and morbidity worldwide and the development of novel therapeutic treatments still remains a major research goal. The contribution of risk factors, such as cigarette smoking, diabetes, hypertension, and hyperlipidaemia, are well recognized as important players for the initiation of cardiac diseases, for which atherosclerosis is commonly shared by all CVDs [1]. Many cardiac disorders, such as heart failure (HF), ischemic/reperfusion (I/R) damage, and myocardial infarction (MI), are characterized by massive cardiac myocytes death [2]. On the other hand, adult human heart has limited capacity to replenish the loss of cardiomyocytes, having an extremely low regenerative ability after cardiac injury, despite some studies having suggested that human heart owns a certain degree of regenerative capacity [3]. Therefore, the loss of cardiomyocytes irreversibly damages the heart with a progressive decrease of its functionality and eventually develops into heart failure. In the context of such a scenario, the identification of the molecular mechanisms underlying cardiac diseases becomes mandatory and the possibility to capture the early events of disease development is crucial for the unraveling of mechanisms and/or markers that can act as potential targets against which to develop new therapeutic strategies. For decades animal models, especially rodents, have represented the model of election for studying human biology, development, and disease, based on the genetic and physiological similarities between the two species. Nonetheless, their phylogenetic relatedness has developed differently for humans and mice, making the experimental results obtained from animal models strikingly different from what is instead true for humans. Thus, although animal models have offered important contributions in understanding human biology and disease, they do not fully mirror the complexity of diseases as they are present in human systems and fail in the intent to translate the results of mice research to humans [4]. Moreover, many promising chemical compounds and drugs that perform well in preclinical animal studies fail in humans due to lack of safety and/or efficacy. Therefore, the use of human tissues would be the most reliable way for a precise understanding of the molecular underpinnings of human biology and pathology, resulting in more accurate and targeted therapies for many human disorders. The use of primary cells from living affected individuals is limited by the amount of tissue and its lifespan in culture, as well as the technical feasibility of accessing particular cell types such as neurons and cardiomyocytes. Human pluripotent stem cells (hPSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have the capability to self-renew indefinitely and to differentiate into derivatives of the three germ layers (ectoderm, mesoderm, and endoderm). These features make them very promising in a variety of basic research and clinical applications such as developmental biology [5], drug screening, disease modelling, and regenerative medicine. Unlike non-human animal models, tissue specific-derived PSCs offer an unprecedented platform for a comprehensive understanding of the molecular basis of CVDs [6]. The revolutionary discovery that somatic cells can be reprogrammed, via overexpression of a set of specific transcription factors, to induced pluripotent stem cells (iPSCs) [7][8][9], paved the way for the generation of patient-specific iPSCs [8][10]. iPSCs derived from healthy individuals or diseased patients carry the genome of their cell of origin and can be differentiated into any cell type, including cells not otherwise accessible, representing a powerful cell-based model system for human diseases, genetic investigations, drug screening, and personalized therapy [11]. Their human origin, pluripotency and ultimately their capability to differentiate into any disease-relevant cell type, and their epigenetic and genetic matching with the patient they are derived from are all features that make iPSCs the most reliable candidate for studying human disorders at cellular level. Moreover, genome-editing approaches can be used to repair and thus to rescue the disease phenotype in patient-derived iPSCs or to introduce pathologically relevant mutations in wild-type lines. To date, a wide number of different monogenic and complex human cardiac disorders have been modeled in vitro using iPSC technology providing new insights into disease mechanisms. The aim of this work is to address the most relevant scientific advances with respect to the use of iPSCs for cardiac disease modeling and to summarize the revolutionary potential of iPSC-derived cardiomyocytes (iPSC-CMs) for cardiac regenerative medicine.

3. Differentiation of Cardiac Cells from iPSCs

For iPSCs to dominate a wide spectrum of biomedical fields, their effective differentiation into specific cell types is of extreme importance. In vitro CM differentiation from iPSCs is achieved by modulation of signaling pathways known to be involved in cardiac development during normal embryogenesis [12]. To date, there have been three main strategies developed to obtain functional CMs from iPSCs: (1) co-culture with visceral endoderm-like cells (END-2); (2) embryoid body (EB)-based differentiation, and (3) two-dimensional culture. During embryonic development in vivo, visceral END-2 releases factors that lead to cardiac differentiation of the nearby mesoderm [13]; this discovery was the basis of the co-culture strategy in which PSCs cultured either in the presence of END-2 cells or in END-2-conditioned medium enter cardiac fate. Although this protocol was successfully applied to both ESCs [14] and iPSCs [15], the CM yield is very low (less than 10%) [16]. EB-based differentiation is a serum-mediated three-dimensional method relying on the capability of PSCs to form floating cell aggregates when cultivated as single cells in low attachment substrate. These aggregates, known as embryoid bodies (EBs), spontaneously produce derivative cells of all the three germ layers [17]. However, the EB-differentiation procedure, due to the presence of serum, suffers from low reproducibility and inter-line variability [18]. Serum was later replaced by cytokines and growth factors known to be involved in heart development such as Wnt proteins [19], bone morphogenetic proteins (BMPs) and activin A [12][20], and Notch signals [21], together with their corresponding inhibitors [22][23]. Different small molecules have been tested for their ability to promote in vitro cardiac differentiation; activators (CHIR99021) and inhibitors (IWR, XAV, IWP2) of the Wnt pathway have been proved to increase cardiac differentiation [24]. However, this strategy requires a high number of starting cells and has a low efficiency. To overcome the limitations of methods based on EB formation, differentiation protocols have been developed based on cell monolayers but with the use of the same molecular factors described for EB differentiation [25][26]. A monolayer-based strategy allowed significant improvement of the yield of cardiac differentiation and the phenotype of derived cardiac cells exhibiting typical features of ventricular, atrial, or nodal cardiomyocytes [27]. The “matrix sandwich” method is a modification of monolayer assay, consisting of covering confluent iPSCs, previously treated with specific growth factors and cytokines to induce cardiac differentiation, with an overlay composed of matrix (i.e., Matrigel) and culture medium. This method relies on the pivotal role that the extracellular matrix plays in the differentiation process leading to high CM purity and yield [28]. Other differentiation methods require two steps: during the first step iPSCs are induced to differentiate into cardiac progenitor cells (CPCs), which in turn can be further differentiated into different cellular fates including CMs, smooth muscle cells (SMCs), and endothelial cells [29][30]. Moreover, it was recently demonstrated that induced CPCs can be directly generated using mouse fibroblasts, skipping the intermediate stage of iPSCs [31][32]. Although there are currently available protocols providing a differentiation efficiency of up to 80% or more in terms of CM purity [27][30][33], all the so far reported strategies show major limitations such as heterogeneity and immaturity of the cardiac population [34]. The low purity and high heterogeneity of the differentiated CM population constitutes an important obstacle for their use in cell-based therapy that requires efficient purification methods to enrich the cardiac population. So far, several studies have developed efficient isolation methods based on the identification of specific cardiac markers such as SIRPA and VCAM1 [35][36][37]. Other studies have instead developed protocols to differentiate human iPSCs into specific subtypes of functional cardiac cells, such as atrial-, ventricular- [38][39][40], nodal-like [41] and pacemaker cells [42]. Although the cardiac cells obtained from iPSCs can start beating very early during differentiation, they resemble, morphologically and functionally, fetal cardiomyocytes. iPSC-CMs display a disorganized morphology, reduced contractile capacity, alteration of glycolytic metabolism, abnormal electrophysiological properties, and reduced automaticity [43]. This immaturity renders adult-onset heart disease modeling very challenging, owing to the uncertainty regarding the ability of relatively immature iPSC-CMs to fully recapitulate adult disease phenotypes or as a function of aging, while the understanding of early-stage pathological events is not affected by low iPSC-CM maturity. Moreover, the incomplete maturity of iPSC-derived CMs could narrow the effectiveness of these cells in mimicking the pathology, e.g., if this is caused by a gene mutated postnatally, with negative impact on their usefulness for studies on drug effect/toxicity. In order to improve the differentiation strategy of iPSCs toward the generation of a high mature and homogeneous cardiomyocyte population, new differentiation methodologies and technical modifications have been proposed. A long culture period (80–120 days) results in multinucleated iPSC-CMs exhibiting mature sarcomeres and increased electrophysiological properties compared to 20–40-day-old CMs [44]. This higher grade of maturity of long-term culture CMs is strictly related to mitochondrial metabolism regulation, which is necessary for energy production and increased cell contractility [45]. Other methods aimed to improve CM maturation include addition of T3 hormone [46] or dexamethasone [47] in culture medium and stressing CMs with mechanical and electrical stimuli [48]. Among the methods developed to increase the maturation of iPSC-CMs, in vivo environments provided the most mature phenotype of iPSC-CMs [49][50]. Currently, nanotechnology-based approaches offer new perspectives in many fields of biomedical research, including cardiovascular research [51]. Three-dimensional scaffolds produced starting from natural or synthetic materials and functionalized to reach specific mechanical and chemical features can be used for direct iPSC differentiation into cardiomyocytes. Scaffolds designed for cardiac differentiation should possess specific properties such as good elasticity to allow cardiac cell contraction and properties to allow cardiomyocytes to arrange in a polarized and organized structure typical of native myocardium. These properties are retained for example by poly (vinyl alcohol) [52], polyethylene oxide [53], poly(lactic-co-glycolic acid) [54], and poly(caprolactone) [55]. CMs cultured on 3D structures show an enhanced calcium signaling respect to monolayer culture [56]; however, it is mandatory to combine this method with electrical and physiological stimulation to obtain cardiac cells with a complete degree of maturity [57]. The number of differentiated cells obtained using classical culture methods represents an additional shortcoming for the application of iPSCs in cell therapy, given that up to one billion CMs need to be transplanted within the infarcted myocardium to replace damaged tissue [58]. Large-scale production of CMs from iPSCs can be achieved using bioreactors that make the process scalable and reproducible via the continuous control and stabilization of culture parameters [59]. Bioreactors create a dynamic suspension culture in which there is a constant flow of nutrients and homeostasis of pH and oxygen levels [60]. iPSCs cultured in a spinner flask form aggregates that can be used as starting materials for CM production after treatments with molecules acting on the Wnt pathway. The spinner flask methods allow critical variables to be tightly supervised, such as aggregate size and cytokine release, augmenting differentiation efficiency [61]. The culture in the suspension of cells that are adhesion-dependent for survival and proliferation can be obtained through the use of supporting matrices known as microcarriers [62]. Laco et al. (2020) developed a microcarrier culture system in a tank bioreactor that allowed scalable iPSC expansion and CM differentiation and purification, reaching a yield of ~40 CMs per iPSC seeded after 22 days in culture [63]. A future intension of bioreactor use will be the production of large amounts of high quality CMs in GMP manufacturing, to improve their use in clinical practice.

4. iPSCs in Cardiac Disease Modeling

In vitro disease modeling is one of the most speculated about fields using iPSC technology. Modeling human cardiac disorders enables definition of the functional and molecular mechanisms underlying a disease and creates the possibility to develop new therapies. The first lines of iPSCs from patients harboring monogenic and complex genetic diseases were established in 2008; one year later, iPSCs were generated from a human specimen [10]. This pioneering publication has rapidly been followed by a growing body of scientific literature. iPSC-based disease modeling has dramatically influenced cardiovascular medicine, offering the opportunity to understand the pathological mechanisms of cardiac diseases and to develop novel effective therapies. This has greatly attracted the scientific community, providing an unprecedented opportunity to recapitulate human monogenic and complex cardiac diseases in vitro. Before the advent of iPSC technology, the severity of CVDs together with the lack of efficient treatments rather than transplantation pushed researchers to develop model systems of cardiac diseases comprising animal models for in vivo studies, in vitro cellular models based on the use of stem cells, primary cells, and various cell lines, and computational studies [64]. The potential of iPSCs and their capability to differentiate into cardiac relevant cell types, including cardiomyocytes, smooth muscle cells, and vascular endothelial cells, as well as and their genetic match to the patient they are derived from, offers a large spectrum of possibilities for the establishment of a robust in vitro model of the disease. iPSC-derived CMs share the same genetic and molecular blueprints as primary human CMs, along with mechanical and electrophysiological properties. To date, a wide range of cardiac diseases including long QT syndrome [65], Leopard syndrome [66], Brugada syndrome [67], catecholaminergic polymorphic ventricular tachycardia [68], arrhythmogenic right ventricular cardiomyopathy/dysplasia [69], dilated cardiomyopathy [70], left ventricular non-compaction [71], hypertrophic cardiomyopathy [72], Andersen-Tawil syndrome [73], and Timothy syndrome [74] have been modeled using iPSC technology. Cardiac diseases are traditionally divided into three main groups: channelopathies, structural cardiomyopathies, and others disorders that do not fit in as channelopathies or structural cardiomyopathies. Among cardiac diseases that are not recognized as channelopathies and/or structural cardiomyopathies, there are several metabolic disorders with cardiac phenotypes. Some of them, such as Friedreich’s ataxia [75], Barth syndrome [76][77], fatty acid oxidation disorders, and Pompe diseases [78] have successfully been translated to iPSC-CM-based models. Ion channelopathies are perhaps the form of cardiac disease with the most well-established iPSC-based disease models.

5. Conclusions

Cardiovascular diseases represent the leading cause of morbidity and death worldwide. Current therapies are mostly focused on relieving symptoms and preventing complications. Despite the progress in clinical research, many HF patients become refractory to standard and/or palliative medical therapies. Therefore, invasive cardiac transplantation remains the only choice for end-stage HF. Nevertheless, the procedure is highly risky and strikingly dependent on access to suitable donors. A comprehensive understanding of the molecular mechanisms underlying human cardiac diseases has been hampered by the lack of reliable model systems that mirror the human disease phenotype. ESCs have been considered the milestone for studying cardiac diseases since they can proliferate indefinitely and can give rise to any cell type, including cardiomyocytes. However, the destruction of the human embryo necessary for the derivation of ESCs has raised important ethical issues preventing and limiting their use. In addition, an ESC-based cardiac disease model cannot be considered totally reliable because of its misleading outcomes due to the individual-specific genetic and epigenetic background. The extraordinary advance accomplished in stem cell biology with the discovery of human iPSCs has completely reshaped our approach to studying human diseases. iPSC technology, through the generation of specific cellular models carrying pathogenetic mutations responsible for the disease phenotype, has allowed novel molecular targets and signaling pathways to be uncovered for the development of new therapeutic strategies. Outstanding advances in differentiation methods, in combination with new impressive genome editing tools like CRISPR-Cas9, have allowed the generation of patient-specific CMs models and their respective isogenic controls. Although iPSCs have generated great enthusiasm within the scientific community, concerns have been raised regarding their real equivalence to ESCs, but little conclusive evidence has been reported regarding iPSC and ESC cardiac derivatives, strengthening the applications of iPSCs in basic and clinical cardiac research. Concerns around iPSC technology are not just about their similarity to ESCs, but also concern the intrinsic properties of iPSC-derived cardiomyocytes. (1) iPSC-CMs typically exhibit immature structural and functional properties resembling phenotypically and functionally fetal cardiomyocytes: while this feature may even be advantageous to model early disease stages, particularly relevant for diseases showing early onset, the low maturity may create problems with regard to the use of iPSC-CMs in clinically relevant settings such as novel drug testing or evaluating their efficacy/or toxicity; moreover, the relatively immature phenotype of iPSC-CMs may mask important pathological mechanisms typical of adult-onset cardiac diseases; (2) iPSC-CMs present batch wise variations in differentiation; (3) major limitations of iPSC technology are associated with the reprogramming process: reprogrammed cells might retain the epigenetic signature of the somatic cell from which they were derived, chromosomal aberrations and/or accumulation of mutations, and genomic instability. Therefore, international standard processes are required to characterize and validate these cells at every stage, with special attention to their use in clinics to ensure safety. Despite the mentioned limitations, the power of iPSC technology for clinical and basic cardiovascular research remains undisputed.

References

  1. Flora, G.D.; Nayak, M.K. A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr. Pharm. Des. 2019, 25, 4063–4084.
  2. Chiong, M.; Wang, Z.V.; Pedrozo, Z.; Cao, D.J.; Troncoso, R.; Ibacache, M.; Criollo, A.; Nemchenko, A.; Hill, J.A.; Lavandero, S. Cardiomyocyte death: Mechanisms and translational implications. Cell Death Dis. 2011, 2, e244.
  3. Bergmann, O.; Bhardwaj, R.D.; Bernard, S.; Zdunek, S.; Barnabé-Heider, F.; Walsh, S.; Zupicich, J.; Alkass, K.; Buchholz, B.A.; Druid, H.; et al. Evidence for Cardiomyocyte Renewal in Humans. Science 2009, 324, 98–102
  4. Justice, M.J.; Dhillon, P. Using the mouse to model human disease: Increasing validity and reproducibility. Dis. Model. Mech. 2016, 9, 101–103.
  5. Dvash, T.; Sharon, N.; Yanuka, O.; Benvenisty, N. Molecular Analysis of LEFTY-Expressing Cells in Early Human. Stem Cells 2007, 25, 465–472.
  6. Parrotta, E.I.; Scalise, S.; Scaramuzzino, L.; Cuda, G. Stem Cells: The Game Changers of Human Cardiac Disease Modelling and Regenerative Medicine. Int. J. Mol. Sci. 2019, 20, 5760.
  7. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676.
  8. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 2007, 131, 861–872.
  9. Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920.
  10. Park, I.H.; Arora, N.; Huo, H.; Maherali, N.; Ahfeldt, T.; Shimamura, A.; Lensch, M.W.; Cowan, C.; Hochedlinger, K.; Daley, G.Q. Disease-Specific Induced Pluripotent Stem (iPS) Cells. Cell 2008, 134, 877–886.
  11. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130.
  12. Yang, L.; Soonpaa, M.H.; Adler, E.D.; Roepke, T.K.; Kattman, S.J.; Kennedy, M.; Henckaerts, E.; Bonham, K.; Abbott, G.W.; Linden, R.M.; et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 2008, 453, 524–528.
  13. Arai, A.; Yamamoto, K.; Toyama, J. Murine Cardiac Progenitor Cells Require Visceral Embryonic Endoderm and Primitive Streak. Dev. Dyn. 1997, 210, 344–353.
  14. Mummery, C.; Oostwaard, D.W.; Doevendans, P.; Spijker, R.; Van Den Brink, S.; Hassink, R.; Van Der Heyden, M.; Opthof, T.; Pera, M.; Brutel, A.; et al. Differentiation of Human Embryonic Stem Cells to Cardiomyocytes. Role of Coculture With Visceral Endoderm-Like Cells. Circulation 2002, 107, 2733–2740.
  15. Freund, C.; Davis, R.P.; Gkatzis, K.; Oostwaard, D.W.; Mummery, C.L. The first reported generation of human induced pluripotent stem cells (iPS cells) and iPS cell-derived cardiomyocytes in the Netherlands. Netherlands Hear. J. 2010, 18, 51–54.
  16. Passier, R.; Oostwaard, D.W.; Snapper, J.; Kloots, J.; Hassink, R.J.; Kuijk, E.; Roelen, B.; De, B. Increased Cardiomyocyte Differentiation from Human Embryonic Stem Cells in Serum-Free Cultures. Stem Cells 2005, 23, 772–780.
  17. Doetschman, T.C.; Eistetter, H.; Katz, M.; Schmidt, W.; Kemler, R. The in vitro development of blastocyst-derived embryonic stem cell lines: Formation of visceral yolk sac, blood islands and myocardium. Development 1985, 87, 27–45.
  18. Osafune, K.; Caron, L.; Borowiak, M.; Martinez, R.J.; Fitz-gerald, C.S.; Sato, Y.; Cowan, C.A.; Chien, K.R.; Melton, D.A. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat. Biotechnol. 2008, 26, 313–315.
  19. Wang, H.; Hao, J.; Hong, C.C. Cardiac Induction of Embryonic Stem Cells by a Small Molecule Inhibitor of Wnt/β-Catenin Signaling. ACS Chem. Biol. 2011, 6, 192–197.
  20. Laflamme, M.A.; Chen, K.Y.; Naumova, A.V.; Muskheli, V.; Fugate, J.A.; Dupras, S.K.; Reinecke, H.; Xu, C.; Hassanipour, M.; Police, S.; et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 2007, 25, 1015–1024.
  21. Chau, M.D.L.; Tuft, R.; Fogarty, K.; Bao, Z. Notch signaling plays a key role in cardiac cell differentiation. Mech. Dev. 123 2006, 123, 626–640.
  22. Bastakoty, D.; Saraswati, S.; Joshi, P.; Atkinson, J.; Liu, J.; Harris, J.L.; Young, P.P. Temporary, Systemic Inhibition of the WNT/β-Catenin Pathway promotes Regenerative Cardiac Repair following Myocardial Infarct. Cell Stem Cell 2016, 2.
  23. Abad, M.; Hashimoto, H.; Zhou, H.; Morales, M.G.; Chen, B.; Bassel-duby, R.; Olson, E.N. Notch Inhibition Enhances Cardiac Reprogramming by Increasing MEF2C Transcriptional Activity. Stem Cell Rep. 2017, 8, 548–560.
  24. Bhattacharya, S.; Burridge, P.W.; Kropp, E.M.; Chuppa, S.L.; Kwok, W.-M.; Wu, J.C.; Boheler, K.R.; Gundry, R.L. High Efficiency Differentiation of Human Pluripotent Stem Cells to Cardiomyocytes and Characterization by Flow Cytometry. J. Vis. Exp. 2014, 23, 52010.
  25. Paige, S.L.; Osugi, T.; Afanasiev, O.K.; Pabon, L.; Reinecke, H.; Murry, C.E. Endogenous Wnt/b-Catenin Signaling Is Required for Cardiac Differentiation in Human Embryonic Stem Cells. PLoS ONE 2010, 5, e11134.
  26. Kadari, A.; Mekala, S.; Wagner, N.; Malan, D.; Köth, J.; Sasse, P.; Herzig, S.; Brüstle, O.; Ergün, S.; Edenhofer, F. Robust Generation of Cardiomyocytes from Human iPS Cells Requires Precise Modulation of BMP and WNT Signaling. Stem Cell Rev. Rep. 2015, 11, 560–569.
  27. Burridge, P.W.; Matsa, E.; Shukla, P.; Lin, Z.C.; Churko, J.M.; Ebert, A.D.; Lan, F.; Diecke, S.; Huber, B.; Mordwinkin, N.M.; et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 2014, 11, 855–860.
  28. Zhang, J.; Klos, M.; Wilson, G.F.; Herman, A.M.; Lian, X.; Raval, K.K.; Barron, M.R.; Hou, L.; Soerens, A.G.; Yu, J.; et al. Extracellular Matrix Promotes Highly Efficient Cardiac Differentiation of Human Pluripotent Stem Cells: The Matrix Sandwich Method. Circ. Res. 2013, 111, 1125–1136.
  29. Kattman, S.J.; Witty, A.D.; Gagliardi, M.; Dubois, N.C.; Niapour, M.; Hotta, A.; Ellis, J.; Keller, G. 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, 228–240.
  30. Cao, N.; Liang, H.; Huang, J.; Wang, J.; Chen, Y.; Chen, Z.; Yang, H. Highly efficient induction and long-term maintenance of multipotent cardiovascular progenitors from human pluripotent stem cells under defined conditions. Cell Res. 2013, 23, 1119–1132.
  31. Lalit, P.A.; Salick, M.R.; Nelson, D.O.; Squirrell, J.M.; Christina, M.; Patel, N.G.; Saeed, I.; Schmuck, E.G.; Markandeya, Y.S.; Wong, R.; et al. Lineage Reprogramming of Fibroblasts to Proliferative Induced Cardiac Progenitor Cells by Defined Factors. Cell Stem Cell 2017, 18, 354–367.
  32. Zhang, Y.; Cao, N.; Huang, Y.; Spencer, C.I.; Fu, J.; Yu, C.; Liu, K.; Nie, B.; Xu, T.; Li, K.; et al. Expandable Cardiovascular Progenitor Cells Reprogrammed from Fibroblasts. Cell Stem Cell 2018, 18, 368–381.
  33. Lian, X.; Hsiao, C.; Wilson, G.; Zhu, K.; Hazeltine, L.B.; Azarin, S.M.; Raval, K.K. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl. Acad. Sci. USA 2012, 109, E1848–E1857.
  34. Veerman, C.C.; Kosmidis, G.; Mummery, C.L.; Casini, S.; Verkerk, A.O.; Bellin, M. Immaturity of Human Stem-Cell-Derived Cardiomyocytes in Culture: Fatal Flaw or Soluble Problem? Stem Cells Dev. 2015, 24, 1035–1052.
  35. Dubois, N.C.; Craft, A.M.; Sharma, P.; Elliott, D.A.; Stanley, E.G.; Elefanty, A.G.; Gramolini, A.; Keller, G. SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells. Nat. Biotechnol. 2011, 29, 1011–1019.
  36. Elliot, D.A.; Braam, S.R.; Koutsis, K.; Ng, E.S.; Jenny, R.; Lagerqvist, E.L.; Biben, C.; Hatzistavrou, T.; Hirst, C.E.; Yu, Q.C.; et al. NKX2-5eGFP/w hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nat. Methods 2011, 8, 1037–1040.
  37. Uosaki, H.; Fukushima, H.; Takeuchi, A.; Matsuoka, S.; Nakatsuji, N.; Yamanaka, S.; Yamashita, J.K. Efficient and Scalable Purification of Cardiomyocytes from Human Embryonic and Induced Pluripotent Stem Cells by VCAM1 Surface Expression. PLoS ONE 2011, 6, e23657.
  38. Weng, Z.; He, J.; Zi, M.; Chow, Y.; Mok, C.F.; Keung, W.; Chow, H.; Leung, A.Y.H.; Hajjar, R.J.; Li, R.A.; et al. A Simple, Cost-Effective but Highly Efficient System for Deriving Ventricular Cardiomyocytes from Human Pluripotent Stem Cells. Stem Cells Dev. 2014, 23, 1704–1716.
  39. Cyganek, L.; Tiburcy, M.; Sekeres, K.; Gerstenberg, K.; Bohnenberger, H.; Lenz, C.; Henze, S.; Stauske, M.; Salinas, G.; Zimmermann, W.; et al. Deep phenotyping of human induced pluripotent stem cell—Derived atrial and ventricular cardiomyocytes. JCI Insight 2018, 3, e99941.
  40. Argenziano, M.; Lambers, E.; Hong, L.; Sridhar, A.; Zhang, M.; Chalazan, B.; Menon, A.; Savio-galimberti, E.; Wu, J.C.; Rehman, J. Electrophysiologic Characterization of Calcium Handling in Human Induced Pluripotent Stem Cell-Derived Atrial Cardiomyocytes. Stem Cell Rep. 2018, 10, 1867–1878.
  41. Zhang, J.; Wilson, G.F.; Soerens, A.G.; Koonce, C.H.; Yu, J.; Sean, P.; Thomson, J.A.; Kamp, T.J. Functional Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells. Circ. Res. 2009, 104, e30–e41.
  42. Schweizer, P.A.; Darche, F.F.; Ullrich, N.D.; Geschwill, P.; Greber, B.; Rivinius, R.; Seyler, C.; Müller-decker, K.; Draguhn, A.; Utikal, J.; et al. Subtype-specific differentiation of cardiac pacemaker cell clusters from human induced pluripotent stem cells. Stem Cell Res. Ther. 2017, 8, 229.
  43. Machiraju, P.; Greenway, S.C. Current methods for the maturation of induced pluripotent stem cell derived cardiomyocytes. World J. Stem Cells 2019, 11, 33–43.
  44. Lundy, S.D.; Zhu, W.-Z.; Regnier, M.; Laflamme, M.A. Structural and Functional Maturation of Cardiomyocytes Derived From Human Pluripotent Stem Cells. Stem Cells Dev. 2013, 22, 1991–2002.
  45. Ebert, A.; Joshi, A.U.; Andorf, S.; Dai, Y.; Sampathkumar, S.; Chen, H.; Li, Y.; Garg, P.; Toischer, K.; Hasenfuss, G.; et al. Proteasome-Dependent Regulation of Distinct Metabolic States During Long-Term Culture of Human. Circ. Res. 2019, 125, 90–103.
  46. Yang, X.; Rodriguez, M.; Pabon, L.; Fischer, K.A.; Reinecke, H.; Regnier, M.; Sniadecki, N.J.; Ruohola-baker, H.; Murry, C.E. Tri-iodo-L-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 2014, 72, 296–304.
  47. Parikh, S.S.; Blackwell, D.J.; Gomez-hurtado, N.; Frisk, M.; Wang, L.; Kim, K.; Dahl, C.P.; Fiane, A.; Tønnessen, T.; Kryshtal, D.O.; et al. Thyroid and Glucocorticoid Hormones Promote Functional T-Tubule Development in Human-Induced Pluripotent Stem Cell–Derived Cardiomyocytes. Circ. Res. 2017, 121, 1323–1330.
  48. Ruan, J.-L.; Tulloch, N.L.; Razumova, M.V.; Saiget, M.; Muskheli, V.; Pabon, L.; Reinecke, H.; Regnier, M.; Murry, C.E. Mechanical stress conditioning and electrical stimulation Promote contractility and Force Maturation of induced Pluripotent stem cell-Derived human cardiac tissue. Circulation 2016, 134, 1557–1567.
  49. Kadota, S.; Pabon, L.; Reinecke, H.; Murry, C.E. In Vivo Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes in Neonatal and Adult Rat Hearts Shin. Stem Cell Rep. 2017, 8, 278–289.
  50. Cho, G.-S.; Lee, D.I.; Tampakakis, E.; Murphy, S.; Andersen, P.; Uosaki, H.; Chelko, S.; Chakir, K.; Hong, I.; Seo, K.; et al. Neonatal Transplantation Confers Maturation of PSC-Derived Cardiomyocytes Conducive to Modeling Cardiomyopathy. Cell Rep. 2017, 18, 571–582.
  51. Chandarana, M.; Curtis, A.; Hoskins, C. The use of nanotechnology in cardiovascular disease. Appl. Nanosci. 2018, 8, 1607–1619.
  52. Dattola, E.; Parrotta, I.; Scalise, S.; Perozziello, G.; Limongi, T.; Candeloro, P.; Coluccio, L.; Maletta, C.; Bruno, L.; De Angelis, T.; et al. Development of 3D PVA scaffolds for cardiac tissue engineering and cell screening applications. RSC Adv. 2019, 9, 4246–4257.
  53. Khorshidi, S.; Solouk, A.; Mirzadeh, H.; Mazinani, S.; Lagaron, J.M.; Shari, S.; Ramakrishna, S. A review of key challenges of electrospun scaffolds for tissue-engineering applications. J. Tissue Eng. Regen. Med. 2016, 10, 715–738.
  54. Stout, D.A.; Basu, B.; Webster, T.J. Poly (lactic—co-glycolic acid): Carbon nanofiber composites for myocardial tissue engineering applications. Acta Biomater. 2011, 7, 3101–3112.
  55. Chen, Y.; Zeng, D.; Ding, L.; Li, X.; Liu, X.; Li, W.; Wei, T.; Yan, S.; Xie, J.; Wei, L.; et al. Three-dimensional poly-(ε-caprolactone) nanofibrous scaffolds directly promote the cardiomyocyte differentiation of murine- induced pluripotent stem cells through Wnt/β-catenin signaling. BMC Cell Biol. 2015, 16, 22.
  56. Fong, A.H.; Romero-Lopez, M.; Heylman, C.M.; Keating, M.; Tran, D.; Sobrino, A.; Tran, A.Q.; Pham, H.H.; Fimbres, C.; Gershon, P.D.; et al. Three-Dimensional Adult Cardiac Extracellular Matrix Promotes Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Tissue Eng. 2016, 22, 1016–1025.
  57. Kroll, K.; Chabria, M.; Wang, K.; Hausermann, F.; Schuler, F.; Polonchuk, L. Electro-mechanical conditioning of human iPSC-derived cardiomyocytes for translational research. Prog. Biophys. Mol. Biol. 2017, 130, 212–222.
  58. Gerbin, K.A.; Murry, C.E. The winding road to regenerating the human heart. Cardiovasc. Pathol. 2016, 24, 133–140.
  59. Zweigerdt, R.; Olmer, R.; Singh, H.; Haverich, A.; Martin, U. Scalable expansion of human pluripotent stem cells in suspension culture. Nat. Protoc. 2011, 6, 689–700.
  60. Ismadi, M.; Gupta, P.; Fouras, A.; Verma, P.; Jadhav, S. Flow Characterization of a Spinner Flask for Induced Pluripotent Stem Cell Culture Application. PLoS ONE 2014, 9, e106493.
  61. Chen, V.C.; Ye, J.; Shukla, P.; Hua, G.; Chen, D.; Lin, Z.; Liu, J.; Chai, J.; Gold, J.; Wu, J.; et al. Development of a scalable suspension culture for cardiac differentiation from human pluripotent stem cells. Stem Cell Rep. 2015, 15, 365–375.
  62. Badenes, S.M.; Fernandes, T.G.; Cordeiro, C.S.M.; Boucher, S.; Kuninger, D.; Vemuri, M.C.; Diogo, M.M.; Cabral, J.M.S. Defined Essential 8 TM Medium and Vitronectin Efficiently Support Scalable Xeno-Free Expansion of Human Induced Pluripotent Stem Cells in Stirred Microcarrier Culture Systems. PLoS ONE 2016, 11, e0151264.
  63. Laco, F.; Lam, A.T.; Woo, T.; Tong, G.; Ho, V.; Soong, P.; Grishina, E.; Lin, K.; Reuveny, S.; Oh, S.K. Selection of human induced pluripotent stem cells lines optimization of cardiomyocytes differentiation in an integrated suspension microcarrier bioreactor. Stem Cell Res. Ther. 2020, 11, 18.
  64. Savoji, H.; Mohammadi, M.H.; Rafatian, N.; Toroghi, M.K.; Wang, E.Y.; Zhao, Y.; Korolj, A.; Ahadian, S.; Radisic, M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019, 198, 3–26.
  65. Sala, L.; Gnecchi, M.; Schwartz, P.J. Long QT Syndrome Modelling with Cardiomyocytes Derived from Human-induced Pluripotent Stem Cells. Arrhythmia Electrophysiol. Rev. 2019, 8, 105–110.
  66. Carvajal-Vergara, X.; Sevilla, A.; Dsouza, S.L.; Ang, Y.S.; Schaniel, C.; Lee, D.F.; Yang, L.; Kaplan, A.D.; Adler, E.D.; Rozov, R.; et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 2010, 465, 808–812.
  67. Liang, P.; Sallam, K.; Wu, H.; Li, Y.; Itzhaki, I.; Garg, P.; Zhang, Y.; Termglichan, V.; Lan, F.; Gu, M.; et al. Patient-Specific and Genome-Edited Induced Pluripotent Stem Cell–Derived Cardiomyocytes Elucidate Single-Cell Phenotype of Brugada Syndrome. J. Am. Coll. Cardiol. 2016, 68, 2086–2096.
  68. Itzhaki, I.; Maizels, L.; Huber, I.; Gepstein, A.; Arbel, G.; Caspi, O.; Miller, L.; Belhassen, B.; Nof, E.; Glikson, M.; et al. Modeling of Catecholaminergic Polymorphic Ventricular Tachycardia with Patient-Specific Human-Induced Pluripotent Stem Cells. J. Am. Coll. Cardiol. 2012, 60, 990–1000.
  69. Caspi, O.; Huber, I.; Gepstein, A.; Arbel, G.; Maizels, L.; Boulos, M.; Gepstein, L. Modeling of arrhythmogenic right ventricular cardiomyopathy with human induced pluripotent stem cells. Circ. Cardiovasc. Genet. 2013, 6, 557–568.
  70. Shah, D.; Virtanen, L.; Prajapati, C.; Kiamehr, M.; Kallio, P.; Taimen, P.; Aalto-Setälä, K. Modeling of LMNA-Related Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Cells 2019, 8, 594.
  71. Kodo, K.; Ong, S.G.; Jahanbani, F.; Termglinchan, V.; Hirono, K.; InanlooRahatloo, K.; Ebert, A.D.; Shukla, P.; Abilez, O.J.; Churko, J.M.; et al. iPSC-derived cardiomyocytes reveal abnormal TGF-β signalling in left ventricular non-compaction cardiomyopathy. Nat. Cell Biol. 2016, 18, 1031–1042.
  72. Li, S.; Pan, H.; Tan, C.; Sun, Y.; Song, Y.; Zhang, X.; Yang, W.; Wang, X.; Li, D.; Dai, Y.; et al. Mitochondrial Dysfunctions Contribute to Hypertrophic Cardiomyopathy in Patient iPSC-Derived Cardiomyocytes with MT-RNR2 Mutation. Stem Cell Rep. 2018, 10, 808–821.
  73. Kuroda, Y.; Yuasa, S.; Watanabe, Y.; Ito, S.; Egashira, T.; Seki, T.; Hattori, T.; Ohno, S.; Kodaira, M.; Suzuki, T.; et al. Flecainide ameliorates arrhythmogenicity through NCX flux in Andersen-Tawil syndrome-iPS cell-derived cardiomyocytes. Biochem. Biophys. Rep. 2017, 9, 245–256.
  74. Yazawa, M.; Hsueh, B.; Jia, X.; Pasca, A.M.; Jonathan, A.; Bernstein, J.H.; Dolmetsch, R.E. Using iPS cells to investigate cardiac phenotypes in patients with Timothy Syndrome. Nature 2011, 471, 230–234.
  75. Hick, A.; Wattenhofer-Donzé, M.; Chintawar, S.; Tropel, P.; Simard, J.P.; Vaucamps, N.; Gall, D.; Lambot, L.; André, C.; Reutenauer, L.; et al. Neurons and cardiomyocytes derived from induced pluripotent stem cells as a model for mitochondrial defects in Friedreich’s ataxia. Dis. Model. Mech. 2013, 6, 608–621.
  76. Wang, G.; McCain, M.L.; Yang, L.; He, A.; Pasqualini, F.S.; Agarwal, A.; Yuan, H.; Jiang, D.; Zhang, D.; Zangi, L.; et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with iPSC and heart-on-chip technologies. J. Musculoskelet. Neuronal Interact. 2014, 15, 1–9.
  77. Fatica, E.M.; DeLeonibus, G.A.; House, A.; Kodger, J.V.; Pearce, R.W.; Shah, R.R.; Levi, L.; Sandlers, Y. Barth Syndrome: Exploring Cardiac Metabolism with Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Metabolites 2019, 9, 306.
  78. Sato, Y.; Kobayashi, H.; Higuchi, T.; Shimada, Y.; Era, T.; Kimura, S.; Ohashi, T.; Eto, Y.; Ida, H. Disease modeling and lentiviral gene transfer in patient-specific induced pluripotent stem cells from late-onset Pompe disease patient. Mol. Ther. Methods Clin. Dev. 2015, 2, 15023.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 846
Revisions: 6 times (View History)
Update Date: 26 Oct 2020
1000/1000