51
|
Engineering the niche to differentiate and deploy cardiovascular cells. Curr Opin Biotechnol 2021; 74:122-128. [PMID: 34861477 DOI: 10.1016/j.copbio.2021.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/23/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022]
Abstract
Applications for stem cells have ranged from therapeutic interventions to more conventional screening and in vitro modeling, but significant limitations to each is due to the lack of maturity from decades old monolayer protocols. While those methods remain the 'gold standard,' newer three-dimensional methods, when combined with engineered niche, stand to significantly improve cell maturity and enable new applications. Here in three parts, we first discuss past methods, and where and why we believe those methods produced suboptimal myocytes. Second, we note how newer methods are moving the field into an era of cell mechanical, electrical, and biological maturity. Finally, we highlight how these improvements will solve issues of scale and engraftment to yield clinical success. It is our conclusion that only through a combination of diverse cell populations and engineered niche will we create an engineered heart tissue with the maturity and vasculature to integrate successfully into a host.
Collapse
|
52
|
Gilbert G, Kadur Nagaraju C, Duelen R, Amoni M, Bobin P, Eschenhagen T, Roderick HL, Sampaolesi M, Sipido KR. Incomplete Assembly of the Dystrophin-Associated Protein Complex in 2D and 3D-Cultured Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cell Dev Biol 2021; 9:737840. [PMID: 34805146 PMCID: PMC8599983 DOI: 10.3389/fcell.2021.737840] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/22/2021] [Indexed: 11/29/2022] Open
Abstract
Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CM) are increasingly used to study genetic diseases on a human background. However, the lack of a fully mature adult cardiomyocyte phenotype of hiPSC-CM may be limiting the scope of these studies. Muscular dystrophies and concomitant cardiomyopathies result from mutations in genes encoding proteins of the dystrophin-associated protein complex (DAPC), which is a multi-protein membrane-spanning complex. We examined the expression of DAPC components in hiPSC-CM, which underwent maturation in 2D and 3D culture protocols. The results were compared with human adult cardiac tissue and isolated cardiomyocytes. We found that similarly to adult cardiomyocytes, hiPSC-CM express dystrophin, in line with previous studies on Duchenne’s disease. β-dystroglycan was also expressed, but, contrary to findings in adult cardiomyocytes, none of the sarcoglycans nor α-dystroglycan were, despite the presence of their mRNA. In conclusion, despite the robust expression of dystrophin, the absence of several other DAPC protein components cautions for reliance on commonly used protocols for hiPSC-CM maturation for functional assessment of the complete DAPC.
Collapse
Affiliation(s)
- Guillaume Gilbert
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Chandan Kadur Nagaraju
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Robin Duelen
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Matthew Amoni
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Pierre Bobin
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Karin R Sipido
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
53
|
Wiesinger A, Boink GJJ, Christoffels VM, Devalla HD. Retinoic acid signaling in heart development: Application in the differentiation of cardiovascular lineages from human pluripotent stem cells. Stem Cell Reports 2021; 16:2589-2606. [PMID: 34653403 PMCID: PMC8581056 DOI: 10.1016/j.stemcr.2021.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022] Open
Abstract
Retinoic acid (RA) signaling plays an important role during heart development in establishing anteroposterior polarity, formation of inflow and outflow tract progenitors, and growth of the ventricular compact wall. RA is also utilized as a key ingredient in protocols designed for generating cardiac cell types from pluripotent stem cells (PSCs). This review discusses the role of RA in cardiogenesis, currently available protocols that employ RA for differentiation of various cardiovascular lineages, and plausible transcriptional mechanisms underlying this fate specification. These insights will inform further development of desired cardiac cell types from human PSCs and their application in preclinical and clinical research.
Collapse
Affiliation(s)
- Alexandra Wiesinger
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Cardiology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
54
|
Approaches to Optimize Stem Cell-Derived Cardiomyocyte Maturation and Function. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
55
|
Setterberg IE, Le C, Frisk M, Li J, Louch WE. The Physiology and Pathophysiology of T-Tubules in the Heart. Front Physiol 2021; 12:718404. [PMID: 34566684 PMCID: PMC8458775 DOI: 10.3389/fphys.2021.718404] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
In cardiomyocytes, invaginations of the sarcolemmal membrane called t-tubules are critically important for triggering contraction by excitation-contraction (EC) coupling. These structures form functional junctions with the sarcoplasmic reticulum (SR), and thereby enable close contact between L-type Ca2+ channels (LTCCs) and Ryanodine Receptors (RyRs). This arrangement in turn ensures efficient triggering of Ca2+ release, and contraction. While new data indicate that t-tubules are capable of exhibiting compensatory remodeling, they are also widely reported to be structurally and functionally compromised during disease, resulting in disrupted Ca2+ homeostasis, impaired systolic and/or diastolic function, and arrhythmogenesis. This review summarizes these findings, while highlighting an emerging appreciation of the distinct roles of t-tubules in the pathophysiology of heart failure with reduced and preserved ejection fraction (HFrEF and HFpEF). In this context, we review current understanding of the processes underlying t-tubule growth, maintenance, and degradation, underscoring the involvement of a variety of regulatory proteins, including junctophilin-2 (JPH2), amphiphysin-2 (BIN1), caveolin-3 (Cav3), and newer candidate proteins. Upstream regulation of t-tubule structure/function by cardiac workload and specifically ventricular wall stress is also discussed, alongside perspectives for novel strategies which may therapeutically target these mechanisms.
Collapse
Affiliation(s)
- Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Christopher Le
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
56
|
Hawthorne RN, Blazeski A, Lowenthal J, Kannan S, Teuben R, DiSilvestre D, Morrissette-McAlmon J, Saffitz JE, Boheler KR, James CA, Chelko SP, Tomaselli G, Tung L. Altered Electrical, Biomolecular, and Immunologic Phenotypes in a Novel Patient-Derived Stem Cell Model of Desmoglein-2 Mutant ARVC. J Clin Med 2021; 10:jcm10143061. [PMID: 34300226 PMCID: PMC8306340 DOI: 10.3390/jcm10143061] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive heart condition which causes fibro-fatty myocardial scarring, ventricular arrhythmias, and sudden cardiac death. Most cases of ARVC can be linked to pathogenic mutations in the cardiac desmosome, but the pathophysiology is not well understood, particularly in early phases when arrhythmias can develop prior to structural changes. Here, we created a novel human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) model of ARVC from a patient with a c.2358delA variant in desmoglein-2 (DSG2). These DSG2-mutant (DSG2Mut) hiPSC-CMs were compared against two wildtype hiPSC-CM lines via immunostaining, RT-qPCR, Western blot, RNA-Seq, cytokine expression and optical mapping. Mutant cells expressed reduced DSG2 mRNA and had altered localization of desmoglein-2 protein alongside thinner, more disorganized myofibrils. No major changes in other desmosomal proteins were noted. There was increased pro-inflammatory cytokine expression that may be linked to canonical and non-canonical NFκB signaling. Action potentials in DSG2Mut CMs were shorter with increased upstroke heterogeneity, while time-to-peak calcium and calcium decay rate were reduced. These were accompanied by changes in ion channel and calcium handling gene expression. Lastly, suppressing DSG2 in control lines via siRNA allowed partial recapitulation of electrical anomalies noted in DSG2Mut cells. In conclusion, the aberrant cytoskeletal organization, cytokine expression, and electrophysiology found DSG2Mut hiPSC-CMs could underlie early mechanisms of disease manifestation in ARVC patients.
Collapse
Affiliation(s)
- Robert N. Hawthorne
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Adriana Blazeski
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Justin Lowenthal
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Suraj Kannan
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Roald Teuben
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Deborah DiSilvestre
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Jeffrey E. Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Cynthia A. James
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Stephen P. Chelko
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| | - Gordon Tomaselli
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| | - Leslie Tung
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| |
Collapse
|
57
|
Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci Rep 2021; 41:226678. [PMID: 33057659 PMCID: PMC8209171 DOI: 10.1042/bsr20200833] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have the ability to differentiate into cardiomyocytes (CMs). They are not only widely used in cardiac pharmacology screening, human heart disease modeling, and cell transplantation-based treatments, but also the most promising source of CMs for experimental and clinical applications. However, their use is largely restricted by the immature phenotype of structure and function, which is similar to embryonic or fetal CMs and has certain differences from adult CMs. In order to overcome this critical issue, many studies have explored and revealed new strategies to induce the maturity of iPSC-CMs. Therefore, this article aims to review recent induction methods of mature iPSC-CMs, related mechanisms, and limitations.
Collapse
|
58
|
Pushp P, Nogueira DES, Rodrigues CAV, Ferreira FC, Cabral JMS, Gupta MK. A Concise Review on Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Personalized Regenerative Medicine. Stem Cell Rev Rep 2021; 17:748-776. [PMID: 33098306 DOI: 10.1007/s12015-020-10061-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
The induced pluripotent stem cells (iPSCs) are derived from somatic cells by using reprogramming factors such as Oct4, Sox2, Klf4, and c-Myc (OSKM) or Oct4, Sox2, Nanog and Lin28 (OSNL). They resemble embryonic stem cells (ESCs) and have the ability to differentiate into cell lineage of all three germ-layer, including cardiomyocytes (CMs). The CMs can be generated from iPSCs by inducing embryoid bodies (EBs) formation and treatment with activin A, bone morphogenic protein 4 (BMP4), and inhibitors of Wnt signaling. However, these iPSC-derived CMs are a heterogeneous population of cells and require purification and maturation to mimic the in vivo CMs. The matured CMs can be used for various therapeutic purposes in regenerative medicine by cardiomyoplasty or through the development of tissue-engineered cardiac patches. In recent years, significant advancements have been made in the isolation of iPSC and their differentiation, purification, and maturation into clinically usable CMs. Newer small molecules have also been identified to substitute the reprogramming factors for iPSC generation as well as for direct differentiation of somatic cells into CMs without an intermediary pluripotent state. This review provides a concise update on the generation of iPSC-derived CMs and their application in personalized cardiac regenerative medicine. It also discusses the current limitations and challenges in the application of iPSC-derived CMs. Graphical abstract.
Collapse
Affiliation(s)
- Pallavi Pushp
- Department of Biotechnology, Institute of Engineering and Technology (IET), Bundelkhand University, Jhansi, Uttar Pradesh, 284128, India
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769 008, India
| | - Diogo E S Nogueira
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Frederico C Ferreira
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Mukesh Kumar Gupta
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769 008, India.
| |
Collapse
|
59
|
Garry MG, Kren S, Wenger JB, Garry DJ. Biologically Derived, Three-Dimensional, Embryonic Scaffolds for Long-Term Cardiomyocyte Culture. Stem Cells Dev 2021; 30:697-704. [PMID: 33843275 DOI: 10.1089/scd.2021.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The ability to maintain viable cultures of mature, primary cardiomyocytes is challenging. The lack of viable cardiomyocyte cultures severely limits in vitro biochemical assays, toxicology assays, drug screening assays, and other analyses. Here, we describe a novel three-dimensional (3D) embryonic scaffold, which supports the culture of postnatal day 7 murine cardiomyocytes within the embryonic heart for, at least, 28 days. We have observed that these cardiomyocytes display normal differentiation, protein expression, and function after extended culture. This novel culture system will allow for prolonged treatment of cardiomyocytes in a natural 3D orientation and has the potential for providing a superior tool for the screening of therapeutic compounds.
Collapse
Affiliation(s)
- Mary G Garry
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine (IEM), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stefan Kren
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph B Wenger
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel J Garry
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine (IEM), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
60
|
Tampakakis E, Mahmoud AI. The role of hormones and neurons in cardiomyocyte maturation. Semin Cell Dev Biol 2021; 118:136-143. [PMID: 33931308 DOI: 10.1016/j.semcdb.2021.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/29/2021] [Indexed: 12/28/2022]
Abstract
The heart undergoes profound morphological and functional changes as it continues to mature postnatally. However, this phase of cardiac development remains understudied. More recently, cardiac maturation research has attracted a lot of interest due to the need for more mature stem cell-derived cardiomyocytes for disease modeling, drug screening and heart regeneration. Additionally, neonatal heart injury models have been utilized to study heart regeneration, and factors regulating postnatal heart development have been associated with adult cardiac disease. Critical components of cardiac maturation are systemic and local biochemical cues. Specifically, cardiac innervation and the concentration of various metabolic hormones appear to increase perinatally and they have striking effects on cardiomyocytes. Here, we first report some of the key parameters of mature cardiomyocytes and then discuss the specific effects of neurons and hormonal cues on cardiomyocyte maturation. We focus primarily on the structural, electrophysiologic, metabolic, hypertrophic and hyperplastic effects of each factor. This review highlights the significance of underappreciated regulators of cardiac maturation and underscores the need for further research in this exciting field.
Collapse
Affiliation(s)
- Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
61
|
Lu K, Seidel T, Cao-Ehlker X, Dorn T, Batcha AMN, Schneider CM, Semmler M, Volk T, Moretti A, Dendorfer A, Tomasi R. Progressive stretch enhances growth and maturation of 3D stem-cell-derived myocardium. Am J Cancer Res 2021; 11:6138-6153. [PMID: 33995650 PMCID: PMC8120210 DOI: 10.7150/thno.54999] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
Bio-engineered myocardium has great potential to substitute damaged myocardium and for studies of myocardial physiology and disease, but structural and functional immaturity still implies limitations. Current protocols of engineered heart tissue (EHT) generation fall short of simulating the conditions of postnatal myocardial growth, which are characterized by tissue expansion and increased mechanical load. To investigate whether these two parameters can improve EHT maturation, we developed a new approach for the generation of cardiac tissues based on biomimetic stimulation under application of continuously increasing stretch. Methods: EHTs were generated by assembling cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CM) at high cell density in a low collagen hydrogel. Maturation and growth of the EHTs were induced in a custom-made biomimetic tissue culture system that provided continuous electrical stimulation and medium agitation along with progressive stretch at four different increments. Tissues were characterized after a three week conditioning period. Results: The highest rate of stretch (S3 = 0.32 mm/day) increased force development by 5.1-fold compared to tissue with a fixed length, reaching contractility of 11.28 mN/mm². Importantly, intensely stretched EHTs developed physiological length-dependencies of active and passive forces (systolic/diastolic ratio = 9.47 ± 0.84), and a positive force-frequency relationship (1.25-fold contractility at 180 min-1). Functional markers of stretch-dependent maturation included enhanced and more rapid Ca2+ transients, higher amplitude and upstroke velocity of action potentials, and pronounced adrenergic responses. Stretch conditioned hiPSC-CMs displayed structural improvements in cellular volume, linear alignment, and sarcomere length (2.19 ± 0.1 µm), and an overall upregulation of genes that are specifically expressed in adult cardiomyocytes. Conclusions: With the intention to simulate postnatal heart development, we have established techniques of tissue assembly and biomimetic culture that avoid tissue shrinkage and yield muscle fibers with contractility and compliance approaching the properties of adult myocardium. This study demonstrates that cultivation under progressive stretch is a feasible way to induce growth and maturation of stem cell-derived myocardium. The novel tissue-engineering approach fulfills important requirements of disease modelling and therapeutic tissue replacement.
Collapse
|
62
|
Santos ARMP, Jang Y, Son I, Kim J, Park Y. Recapitulating Cardiac Structure and Function In Vitro from Simple to Complex Engineering. MICROMACHINES 2021; 12:mi12040386. [PMID: 33916254 PMCID: PMC8067203 DOI: 10.3390/mi12040386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiac tissue engineering aims to generate in vivo-like functional tissue for the study of cardiac development, homeostasis, and regeneration. Since the heart is composed of various types of cells and extracellular matrix with a specific microenvironment, the fabrication of cardiac tissue in vitro requires integrating technologies of cardiac cells, biomaterials, fabrication, and computational modeling to model the complexity of heart tissue. Here, we review the recent progress of engineering techniques from simple to complex for fabricating matured cardiac tissue in vitro. Advancements in cardiomyocytes, extracellular matrix, geometry, and computational modeling will be discussed based on a technology perspective and their use for preparation of functional cardiac tissue. Since the heart is a very complex system at multiscale levels, an understanding of each technique and their interactions would be highly beneficial to the development of a fully functional heart in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Jongseong Kim
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| | - Yongdoo Park
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| |
Collapse
|
63
|
Disease Modeling and Disease Gene Discovery in Cardiomyopathies: A Molecular Study of Induced Pluripotent Stem Cell Generated Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22073311. [PMID: 33805011 PMCID: PMC8037452 DOI: 10.3390/ijms22073311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/04/2023] Open
Abstract
The in vitro modeling of cardiac development and cardiomyopathies in human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) provides opportunities to aid the discovery of genetic, molecular, and developmental changes that are causal to, or influence, cardiomyopathies and related diseases. To better understand the functional and disease modeling potential of iPSC-differentiated CMs and to provide a proof of principle for large, epidemiological-scale disease gene discovery approaches into cardiomyopathies, well-characterized CMs, generated from validated iPSCs of 12 individuals who belong to four sibships, and one of whom reported a major adverse cardiac event (MACE), were analyzed by genome-wide mRNA sequencing. The generated CMs expressed CM-specific genes and were highly concordant in their total expressed transcriptome across the 12 samples (correlation coefficient at 95% CI =0.92 ± 0.02). The functional annotation and enrichment analysis of the 2116 genes that were significantly upregulated in CMs suggest that generated CMs have a transcriptomic and functional profile of immature atrial-like CMs; however, the CMs-upregulated transcriptome also showed high overlap and significant enrichment in primary cardiomyocyte (p-value = 4.36 × 10−9), primary heart tissue (p-value = 1.37 × 10−41) and cardiomyopathy (p-value = 1.13 × 10−21) associated gene sets. Modeling the effect of MACE in the generated CMs-upregulated transcriptome identified gene expression phenotypes consistent with the predisposition of the MACE-affected sibship to arrhythmia, prothrombotic, and atherosclerosis risk.
Collapse
|
64
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
65
|
Garbern JC, Lee RT. Mitochondria and metabolic transitions in cardiomyocytes: lessons from development for stem cell-derived cardiomyocytes. Stem Cell Res Ther 2021; 12:177. [PMID: 33712058 PMCID: PMC7953594 DOI: 10.1186/s13287-021-02252-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022] Open
Abstract
Current methods to differentiate cardiomyocytes from human pluripotent stem cells (PSCs) inadequately recapitulate complete development and result in PSC-derived cardiomyocytes (PSC-CMs) with an immature or fetal-like phenotype. Embryonic and fetal development are highly dynamic periods during which the developing embryo or fetus is exposed to changing nutrient, oxygen, and hormone levels until birth. It is becoming increasingly apparent that these metabolic changes initiate developmental processes to mature cardiomyocytes. Mitochondria are central to these changes, responding to these metabolic changes and transitioning from small, fragmented mitochondria to large organelles capable of producing enough ATP to support the contractile function of the heart. These changes in mitochondria may not simply be a response to cardiomyocyte maturation; the metabolic signals that occur throughout development may actually be central to the maturation process in cardiomyocytes. Here, we review methods to enhance maturation of PSC-CMs and highlight evidence from development indicating the key roles that mitochondria play during cardiomyocyte maturation. We evaluate metabolic transitions that occur during development and how these affect molecular nutrient sensors, discuss how regulation of nutrient sensing pathways affect mitochondrial dynamics and function, and explore how changes in mitochondrial function can affect metabolite production, the cell cycle, and epigenetics to influence maturation of cardiomyocytes.
Collapse
Affiliation(s)
- Jessica C Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA, 02138, USA
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA, 02138, USA.
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| |
Collapse
|
66
|
iPSC-Cardiomyocyte Models of Brugada Syndrome-Achievements, Challenges and Future Perspectives. Int J Mol Sci 2021; 22:ijms22062825. [PMID: 33802229 PMCID: PMC8001521 DOI: 10.3390/ijms22062825] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
Brugada syndrome (BrS) is an inherited cardiac arrhythmia that predisposes to ventricular fibrillation and sudden cardiac death. It originates from oligogenic alterations that affect cardiac ion channels or their accessory proteins. The main hurdle for the study of the functional effects of those variants is the need for a specific model that mimics the complex environment of human cardiomyocytes. Traditionally, animal models or transient heterologous expression systems are applied for electrophysiological investigations, each of these models having their limitations. The ability to create induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), providing a source of human patient-specific cells, offers new opportunities in the field of cardiac disease modelling. Contemporary iPSC-CMs constitute the best possible in vitro model to study complex cardiac arrhythmia syndromes such as BrS. To date, thirteen reports on iPSC-CM models for BrS have been published and with this review we provide an overview of the current findings, with a focus on the electrophysiological parameters. We also discuss the methods that are used for cell derivation and data acquisition. In the end, we critically evaluate the knowledge gained by the use of these iPSC-CM models and discuss challenges and future perspectives for iPSC-CMs in the study of BrS and other arrhythmias.
Collapse
|
67
|
Kay M, Soltani BM. LncRNAs in Cardiomyocyte Maturation: New Window for Cardiac Regenerative Medicine. Noncoding RNA 2021; 7:ncrna7010020. [PMID: 33802186 PMCID: PMC8005985 DOI: 10.3390/ncrna7010020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiomyocyte (CM) maturation, which is characterized by structural, functional, and metabolic specializations, is the last phase of CM development that prepares the cells for efficient and forceful contraction throughout life. Over the past decades, CM maturation has gained increased attention due to the fact that pluripotent stem cell-derived CMs are structurally, transcriptionally, and functionally immature and embryonic-like, which causes a defect in cell replacement therapy. The current challenge is to discover and understand the molecular mechanisms, which control the CM maturation process. Currently, emerging shreds of evidence emphasize the role of long noncoding RNAs (lncRNAs) in regulating different aspects of CM maturation, including myofibril maturation, electrophysiology, and Ca2+ handling maturation, metabolic maturation and proliferation to hypertrophy transition. Here, we describe the structural and functional characteristics of mature CMs. Furthermore, this review highlights the lncRNAs as crucial regulators of different aspects in CM maturation, which have the potential to be used for mature CM production. With the current advances in oligonucleotide delivery; lncRNAs may serve as putative therapeutic targets to produce highly mature CMs for research and regenerative medicine.
Collapse
|
68
|
Burnett SD, Blanchette AD, Chiu WA, Rusyn I. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes as an in vitro model in toxicology: strengths and weaknesses for hazard identification and risk characterization. Expert Opin Drug Metab Toxicol 2021; 17:887-902. [PMID: 33612039 DOI: 10.1080/17425255.2021.1894122] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes is one of the most widely used cell-based models that resulted from the discovery of how non-embryonic stem cells can be differentiated into multiple cell types. In just one decade, iPSC-derived cardiomyocytes went from a research lab to widespread use in biomedical research and preclinical safety evaluation for drugs and other chemicals. AREAS COVERED This manuscript reviews data on toxicology applications of human iPSC-derived cardiomyocytes. We detail the outcome of a systematic literature search on their use (i) in hazard assessment for cardiotoxicity liabilities, (ii) for risk characterization, (iii) as models for population variability, and (iv) in studies of personalized medicine and disease. EXPERT OPINION iPSC-derived cardiomyocytes are useful to increase the accuracy, precision, and efficiency of cardiotoxicity hazard identification for both drugs and non-pharmaceuticals, with recent efforts beginning to demonstrate their utility for risk characterization. Notable limitations include the needs to improve the maturation of cells in culture, to better understand their potential use identifying structural cardiotoxicity, and for additional case studies involving population-wide and disease-specific risk characterization. Ultimately, the greatest future benefits are likely for non-pharmaceutical chemicals, filling a critical gap where no routine testing for cardiotoxicity is currently performed.
Collapse
Affiliation(s)
- Sarah D Burnett
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Alexander D Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
69
|
Zhao Y, Iyer S, Tavanaei M, Nguyen NT, Lin A, Nguyen TP. Proarrhythmic Electrical Remodeling by Noncardiomyocytes at Interfaces With Cardiomyocytes Under Oxidative Stress. Front Physiol 2021; 11:622613. [PMID: 33603677 PMCID: PMC7884825 DOI: 10.3389/fphys.2020.622613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Life-threatening ventricular arrhythmias, typically arising from interfaces between fibrosis and surviving cardiomyocytes, are feared sequelae of structurally remodeled hearts under oxidative stress. Incomplete understanding of the proarrhythmic electrical remodeling by fibrosis limits the development of novel antiarrhythmic strategies. To define the mechanistic determinants of the proarrhythmia in electrical crosstalk between cardiomyocytes and noncardiomyocytes, we developed a novel in vitro model of interface between neonatal rat ventricular cardiomyocytes (NRVMs) and controls [NRVMs or connexin43 (Cx43)-deficient HeLa cells] vs. Cx43+ noncardiomyocytes [aged rat ventricular myofibroblasts (ARVFs) or HeLaCx43 cells]. We performed high-speed voltage-sensitive optical imaging at baseline and following acute H2O2 exposure. In NRVM-NRVM and NRVM-HeLa controls, no arrhythmias occurred under either experimental condition. In the NRVM-ARVF and NRVM-HeLaCx43 groups, Cx43+ noncardiomyocytes enabled passive decremental propagation of electrical impulses and impaired NRVM activation and repolarization, thereby slowing conduction and prolonging action potential duration. Following H2O2 exposure, arrhythmia triggers, automaticity, and non-reentrant and reentrant arrhythmias emerged. This study reveals that myofibroblasts (which generate cardiac fibrosis) and other noncardiomyocytes can induce not only structural remodeling but also electrical remodeling and that electrical remodeling by noncardiomyocytes can be particularly arrhythmogenic in the presence of an oxidative burst. Synergistic electrical remodeling between H2O2 and noncardiomyocytes may account for the clinical arrhythmogenicity of myofibroblasts at fibrotic interfaces with cardiomyocytes in ischemic/non-ischemic cardiomyopathies. Understanding the enhanced arrhythmogenicity of synergistic electrical remodeling by H2O2 and noncardiomyocytes may guide novel safe-by-design antiarrhythmic strategies for next-generation iatrogenic interfaces between surviving native cardiomyocytes and exogenous stem cells or engineered tissues in cardiac regenerative therapies.
Collapse
Affiliation(s)
- Yali Zhao
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Shankar Iyer
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Maryam Tavanaei
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Nicole T Nguyen
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Andrew Lin
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Thao P Nguyen
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
70
|
de Lange WJ, Farrell ET, Kreitzer CR, Jacobs DR, Lang D, Glukhov AV, Ralphe JC. Human iPSC-engineered cardiac tissue platform faithfully models important cardiac physiology. Am J Physiol Heart Circ Physiol 2021; 320:H1670-H1686. [PMID: 33606581 DOI: 10.1152/ajpheart.00941.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CM) may provide an important bridge between animal models and the intact human myocardium. Fulfilling this potential is hampered by their relative immaturity, leading to poor physiological responsiveness. hiPSC-CMs grown in traditional two-dimensional (2D) culture lack a t-tubular system, have only rudimentary intracellular calcium-handling systems, express predominantly embryonic sarcomeric protein isoforms, and preferentially use glucose as an energy substrate. Culturing hiPSC-CM in a variety of three-dimensional (3D) environments and the addition of nutritional, pharmacological, and electromechanical stimuli have proven, to various degrees, to be beneficial for maturation. We present a detailed assessment of a novel model in which hiPSC-CMs and hiPSC-derived cardiac fibroblasts are cocultured in a 3D fibrin matrix to form engineered cardiac tissue constructs (hiPSC-ECTs). The hiPSC-ECTs are responsive to physiological stimuli, including stretch, frequency, and β-adrenergic stimulation, develop a t-tubular system, and demonstrate calcium-handling and contractile kinetics that compare favorably with ventricular human myocardium. Furthermore, transcript levels of various genes involved in calcium-handling and contraction are increased. These markers of maturation become more robust over a relatively short period of time in culture (6 wk vs. 2 wk in hiPSC-ECTs). A comparison of the hiPSC-ECT molecular and performance variables with those of human cardiac tissue and other available engineered tissue platforms is provided to aid selection of the most appropriate platform for the research question at hand. Important and noteworthy aspects of this human cardiac model system are its reliance on "off-the-shelf" equipment, ability to provide detailed physiological performance data, and the ability to achieve a relatively mature cardiac physiology without additional nutritional, pharmacological, and electromechanical stimuli that may elicit unintended effects on function.NEW & NOTEWORTHY This study seeks to provide an in-depth assessment of contractile performance of human iPSC-derived cardiomyocytes cultured together with fibroblasts in a 3-dimensional-engineered tissue and compares performance both over time as cells mature, and with corresponding measures found in the literature using alternative 3D culture configurations. The suitability of 3D-engineered human cardiac tissues to model cardiac function is emphasized, and data provided to assist in the selection of the most appropriate configuration based on the target application.
Collapse
Affiliation(s)
- Willem J de Lange
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Emily T Farrell
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Caroline R Kreitzer
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Derek R Jacobs
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Di Lang
- Department of Medicine Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Alexey V Glukhov
- Department of Medicine Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
71
|
Zhang R, Guo T, Han Y, Huang H, Shi J, Hu J, Li H, Wang J, Saleem A, Zhou P, Lan F. Design of synthetic microenvironments to promote the maturation of human pluripotent stem cell derived cardiomyocytes. J Biomed Mater Res B Appl Biomater 2020; 109:949-960. [PMID: 33231364 DOI: 10.1002/jbm.b.34759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/08/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022]
Abstract
Cardiomyocyte like cells derived from human pluripotent stem cells (hPSC-CMs) have a good application perspective in many fields such as disease modeling, drug screening and clinical treatment. However, these are severely hampered by the fact that hPSC-CMs are immature compared to adult human cardiomyocytes. Therefore, many approaches such as genetic manipulation, biochemical factors supplement, mechanical stress, electrical stimulation and three-dimensional culture have been developed to promote the maturation of hPSC-CMs. Recently, establishing in vitro synthetic artificial microenvironments based on the in vivo development program of cardiomyocytes has achieved much attention due to their inherent properties such as stiffness, plasticity, nanotopography and chemical functionality. In this review, the achievements and deficiency of reported synthetic microenvironments that mainly discussed comprehensive biological, chemical, and physical factors, as well as three-dimensional culture were mainly discussed, which have significance to improve the microenvironment design and accelerate the maturation of hPSC-CMs.
Collapse
Affiliation(s)
- Rui Zhang
- School and hospital of Stomatology, Lanzhou University, Lanzhou, China.,College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Tianwei Guo
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yu Han
- School and hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Hongxin Huang
- School and hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiamin Shi
- College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jiaxuan Hu
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Hongjiao Li
- School and hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Jianlin Wang
- College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Amina Saleem
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ping Zhou
- School and hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Feng Lan
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
72
|
Sontayananon N, Redwood C, Davies B, Gehmlich K. Fluorescent PSC-Derived Cardiomyocyte Reporter Lines: Generation Approaches and Their Applications in Cardiovascular Medicine. BIOLOGY 2020; 9:biology9110402. [PMID: 33207727 PMCID: PMC7697758 DOI: 10.3390/biology9110402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
Recent advances have made pluripotent stem cell (PSC)-derived cardiomyocytes an attractive option to model both normal and diseased cardiac function at the single-cell level. However, in vitro differentiation yields heterogeneous populations of cardiomyocytes and other cell types, potentially confounding phenotypic analyses. Fluorescent PSC-derived cardiomyocyte reporter systems allow specific cell lineages to be labelled, facilitating cell isolation for downstream applications including drug testing, disease modelling and cardiac regeneration. In this review, the different genetic strategies used to generate such reporter lines are presented with an emphasis on their relative technical advantages and disadvantages. Next, we explore how the fluorescent reporter lines have provided insights into cardiac development and cardiomyocyte physiology. Finally, we discuss how exciting new approaches using PSC-derived cardiomyocyte reporter lines are contributing to progress in cardiac cell therapy with respect to both graft adaptation and clinical safety.
Collapse
Affiliation(s)
- Naeramit Sontayananon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK; (N.S.); (C.R.)
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK; (N.S.); (C.R.)
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
- Correspondence: (B.D.); (K.G.)
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK; (N.S.); (C.R.)
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Correspondence: (B.D.); (K.G.)
| |
Collapse
|
73
|
Jiang L, Liang J, Huang W, Wu Z, Paul C, Wang Y. Strategies and Challenges to Improve Cellular Programming-Based Approaches for Heart Regeneration Therapy. Int J Mol Sci 2020; 21:E7662. [PMID: 33081233 PMCID: PMC7589611 DOI: 10.3390/ijms21207662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used to regenerate damaged heart. Using these approaches, new CMs can be regenerated from differentiation of iPSCs (similar to embryonic stem cells), the direct reprogramming of fibroblasts [induced cardiomyocytes (iCMs)], or induced cardiac progenitors. Although these CMs have been shown to functionally repair infarcted heart, advancements in technology are still in the early stages of development in research laboratories. In this review, reprogramming-based approaches for generating CMs are briefly introduced and reviewed, and the challenges (including low efficiency, functional maturity, and safety issues) that hinder further translation of these approaches into a clinical setting are discussed. The creative and combined optimal methods to address these challenges are also summarized, with optimism that further investigation into tissue engineering, cardiac development signaling, and epigenetic mechanisms will help to establish methods that improve cell-reprogramming approaches for heart regeneration.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
74
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
75
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
76
|
Zhang XH, Morad M. Ca 2+ signaling of human pluripotent stem cells-derived cardiomyocytes as compared to adult mammalian cardiomyocytes. Cell Calcium 2020; 90:102244. [PMID: 32585508 PMCID: PMC7483365 DOI: 10.1016/j.ceca.2020.102244] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) have been extensively used for in vitro modeling of human cardiovascular disease, drug screening and pharmacotherapy, but little rigorous studies have been reported on their biophysical or Ca2+ signaling properties. There is also considerable concern as to the level of their maturity and whether they can serve as reliable models for adult human cardiac myocytes. Ultrastructural difference such as lack of t-tubular network, their polygonal shapes, disorganized sarcomeric myofilament, and their rhythmic automaticity, among others, have been cited as evidence for immaturity of hiPSC-CMs. In this review, we will deal with Ca2+ signaling, its regulation, and its stage of maturity as compared to the mammalian adult cardiomyocytes. We shall summarize the data on functional aspects of Ca2+signaling and its parameters that include: L-type calcium channel (Cav1.2), ICa-induced Ca2+release, CICR, and its parameters, cardiac Na/Ca exchanger (NCX1), the ryanodine receptors (RyR2), sarco-reticular Ca2+pump, SERCA2a/PLB, and the contribution of mitochondrial Ca2+ to hiPSC-CMs excitation-contraction (EC)-coupling as compared with adult mammalian cardiomyocytes. The comparative studies suggest that qualitatively hiPSC-CMs have similar Ca2+signaling properties as those of adult cardiomyocytes, but quantitative differences do exist. This review, we hope, will allow the readers to judge for themselves to what extent Ca2+signaling of hiPSC-CMs represents the adult form of this signaling pathway, and whether these cells can be used as good models of human cardiomyocytes.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States
| | - Martin Morad
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States.
| |
Collapse
|
77
|
Branco MA, Cabral JM, Diogo MM. From Human Pluripotent Stem Cells to 3D Cardiac Microtissues: Progress, Applications and Challenges. Bioengineering (Basel) 2020; 7:E92. [PMID: 32785039 PMCID: PMC7552661 DOI: 10.3390/bioengineering7030092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
The knowledge acquired throughout the years concerning the in vivo regulation of cardiac development has promoted the establishment of directed differentiation protocols to obtain cardiomyocytes (CMs) and other cardiac cells from human pluripotent stem cells (hPSCs), which play a crucial role in the function and homeostasis of the heart. Among other developments in the field, the transition from homogeneous cultures of CMs to more complex multicellular cardiac microtissues (MTs) has increased the potential of these models for studying cardiac disorders in vitro and for clinically relevant applications such as drug screening and cardiotoxicity tests. This review addresses the state of the art of the generation of different cardiac cells from hPSCs and the impact of transitioning CM differentiation from 2D culture to a 3D environment. Additionally, current methods that may be employed to generate 3D cardiac MTs are reviewed and, finally, the adoption of these models for in vitro applications and their adaptation to medium- to high-throughput screening settings are also highlighted.
Collapse
Affiliation(s)
| | | | - Maria Margarida Diogo
- iBB-Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (M.A.B.); (J.M.S.C.)
| |
Collapse
|
78
|
Lemcke H, Skorska A, Lang CI, Johann L, David R. Quantitative Evaluation of the Sarcomere Network of Human hiPSC-Derived Cardiomyocytes Using Single-Molecule Localization Microscopy. Int J Mol Sci 2020; 21:ijms21082819. [PMID: 32316650 PMCID: PMC7216082 DOI: 10.3390/ijms21082819] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
The maturation of iPSC-derived cardiomyocytes is still a critical point for their application in cardiovascular research as well as for their clinical use. Although multiple differentiation protocols have been established, researchers failed to generate fully mature cardiomyocytes in vitro possessing identical phenotype-related and functional properties as their native adult counterparts. Besides electrophysiological and metabolic changes, the establishment of a well structured sarcomere network is important for the development of a mature cardiac phenotype. Here, we present a super resolution-based approach to quantitatively evaluate the structural maturation of iPSC-derived cardiomyocytes. Fluorescence labelling of the α-actinin cytoskeleton and subsequent visualization by photoactivated localization microscopy allows the acquisition of highly resolved images for measuring sarcomere length and z-disc thickness. Our image analysis revealed that iPSC and neonatal cardiomyocyte share high similarity with respect to their sarcomere organization, however, contraction capacity was inferior in iPSC-derived cardiac cells, indicating an early maturation level. Moreover, we demonstrate that this imaging approach can be used as a tool to monitor cardiomyocyte integrity, helping to optimize iPSC differentiation as well as somatic cell direct-reprogramming strategies.
Collapse
Affiliation(s)
- Heiko Lemcke
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Rostock University Medical Center, 18057 Rostock, Germany; (H.L.); (A.S.); (L.J.)
- Faculty of Interdisciplinary Research, Department Life, Light & Matter, University Rostock, 18059 Rostock, Germany
| | - Anna Skorska
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Rostock University Medical Center, 18057 Rostock, Germany; (H.L.); (A.S.); (L.J.)
- Faculty of Interdisciplinary Research, Department Life, Light & Matter, University Rostock, 18059 Rostock, Germany
| | - Cajetan Immanuel Lang
- Department of Cardiology, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Lisa Johann
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Rostock University Medical Center, 18057 Rostock, Germany; (H.L.); (A.S.); (L.J.)
- Faculty of Interdisciplinary Research, Department Life, Light & Matter, University Rostock, 18059 Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Rostock University Medical Center, 18057 Rostock, Germany; (H.L.); (A.S.); (L.J.)
- Faculty of Interdisciplinary Research, Department Life, Light & Matter, University Rostock, 18059 Rostock, Germany
- Correspondence:
| |
Collapse
|
79
|
Abstract
Maturation is the last phase of heart development that prepares the organ for strong, efficient, and persistent pumping throughout the mammal's lifespan. This process is characterized by structural, gene expression, metabolic, and functional specializations in cardiomyocytes as the heart transits from fetal to adult states. Cardiomyocyte maturation gained increased attention recently due to the maturation defects in pluripotent stem cell-derived cardiomyocyte, its antagonistic effect on myocardial regeneration, and its potential contribution to cardiac disease. Here, we review the major hallmarks of ventricular cardiomyocyte maturation and summarize key regulatory mechanisms that promote and coordinate these cellular events. With advances in the technical platforms used for cardiomyocyte maturation research, we expect significant progress in the future that will deepen our understanding of this process and lead to better maturation of pluripotent stem cell-derived cardiomyocyte and novel therapeutic strategies for heart disease.
Collapse
Affiliation(s)
- Yuxuan Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - William Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
80
|
Ahmed RE, Anzai T, Chanthra N, Uosaki H. A Brief Review of Current Maturation Methods for Human Induced Pluripotent Stem Cells-Derived Cardiomyocytes. Front Cell Dev Biol 2020; 8:178. [PMID: 32266260 PMCID: PMC7096382 DOI: 10.3389/fcell.2020.00178] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/03/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Therefore, the discovery of induced pluripotent stem cells (iPSCs) and the subsequent generation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) was a pivotal point in regenerative medicine and cardiovascular research. They constituted an appealing tool for replacing dead and dysfunctional cardiac tissue, screening cardiac drugs and toxins, and studying inherited cardiac diseases. The problem is that these cells remain largely immature, and in order to utilize them, they must reach a functional degree of maturity. To attempt to mimic in vivo environment, various methods including prolonging culture time, co-culture and modulations of chemical, electrical, mechanical culture conditions have been tried. In addition to that, changing the topology of the culture made huge progress with the introduction of the 3D culture that closely resembles the in vivo cardiac topology and overcomes many of the limitations of the conventionally used 2D models. Nonetheless, 3D culture alone is not enough, and using a combination of these methods is being explored. In this review, we summarize the main differences between immature, fetal-like hiPSC-CMs and adult cardiomyocytes, then glance at the current approaches used to promote hiPSC-CMs maturation. In the second part, we focus on the evolving 3D culture model - it's structure, the effect on hiPSC-CMs maturation, incorporation with different maturation methods, limitations and future prospects.
Collapse
Affiliation(s)
- Razan Elfadil Ahmed
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|