JARB Journal of Animal Reproduction and Biotehnology

OPEN ACCESS pISSN: 2671-4639
eISSN: 2671-4663

Article Search

Review Article

Article Review Article
Split Viewer

Journal of Animal Reproduction and Biotechnology 2022; 37(2): 67-79

Published online June 30, 2022

https://doi.org/10.12750/JARB.37.2.67

Copyright © The Korean Society of Animal Reproduction and Biotechnology.

In vitro maturation of human pluripotent stem cell-derived cardiomyocyte: A promising approach for cell therapy

Yun-Gwi Park1,# , Yeo-Jin Son1,# , Sung-Hwan Moon1,2,* and Soon-Jung Park1,*

1Stem Cell Research Institute, T&R Biofab Co. Ltd., Seongnam 13487, Korea
2Department of Animal Science, Sangji University, Wonju 26339, Korea

Correspondence to: Sung-Hwan Moon
E-mail: moonsh22@sangji.ac.kr

Soon-Jung Park
E-mail: parksoonjung@gmail.com

#These authors contributed to this work as first authors.

Received: June 6, 2022; Revised: June 17, 2022; Accepted: June 20, 2022

Currently, there is no treatment to reverse or cure heart failure caused by ischemic heart disease and myocardial infarction despite the remarkable advances in modern medicine. In addition, there is a lack of evidence regarding the existence of stem cells involved in the proliferation and regeneration of cardiomyocytes in adult hearts. As an alternative solution to overcome this problem, protocols for differentiating human pluripotent stem cell (hPSC) into cardiomyocyte have been established, which further led to the development of cell therapy in major leading countries in this field. Recently, clinical studies have confirmed the safety of hPSC-derived cardiac progenitor cells (CPCs). Although several institutions have shown progress in their research on cell therapy using hPSC-derived cardiomyocytes, the functions of cardiomyocytes used for transplantation remain to be those of immature cardiomyocytes, which poses a risk of graft-induced arrhythmias in the early stage of transplantation. Over the last decade, research aimed at achieving maturation of immature cardiomyocytes, showing same characteristics as those of mature cardiomyocytes, has been actively conducted using various approaches at leading research institutes worldwide. However, challenges remain in technological development for effective generation of mature cardiomyocytes with the same properties as those present in the adult hearts. Therefore, in this review, we provide an overview of the technological development status for maturation methods of hPSC-derived cardiomyocytes and present a direction for future development of maturation techniques.

Keywords: cardiomyocyte, cell therapy, human pluripotent stem cell, maturation, safety

According to a report by the World Health Organization, cardiovascular diseases (CVDs) are the leading cause of death worldwide, with 8.9 million estimated deaths in 2019. In addition, it has been reported that those who had SARS-CoV-2 infection, which had a sweeping impact worldwide, had sequelae with long-term cardiovascular consequences (Abbasi, 2022). Since human cardiomyocytes cease to proliferate and the cardiac regenerative potential is lost shortly after birth (Hirose et al., 2019), treatments for any damage to the heart require medical interventions such as mechanical devices (e.g., a left ventricular assist device) or a cardiac transplant, allowing only limited available options. To overcome these limitations, the use of adult stem cells and pluripotent stem cells (PSCs) as cell therapy has garnered attention. In particular, differentiation of cardiomyocytes using PSCs capable of differentiating into various somatic cell types has drawn increased interest as alternative approach for cell therapy (Yu et al., 2019; de Lange et al., 2021). Consequently, various methods for the differentiation of cardiomyocytes have been developed and actively utilized to explore cell therapy, offering therapeutic strategies against cardiac repair or drug-induced cardiotoxicity (Burridge et al., 2015; Park et al., 2019; Choi et al., 2020).

Recently, a research team in France reported the completion of a phase 1 clinical trial involving human embryonic stem cells (hESCs)-derived CPCs to demonstrate the safety and effect of cardiac function improvement (Menasché et al., 2018). In Japan, based on the results of preclinical studies with heart failure (HF) animal models (Kashiyama et al., 2019; Kawaguchi et al., 2021), an ongoing clinical trial has been utilizing the transplantation of human induced pluripotent stem cells (hiPSCs)-derived cardiomyocyte sheets or spheroids. As can be seen from the above examples, research on the investigation of cell therapy using hPSC-derived cardiomyocytes for the treatment of heart diseases with limited alternative treatment options has developed into an area of global competition among world-class research groups. However, regarding the applications of hPSC-derived cardiomyocytes (CMs) in clinical settings, it is necessary to overcome the issue of graft-related arrhythmias. In several papers, as a result of transplanting hPSC-derived cardiomyocytes into heart disease model animals, arrhythmias (e.g., ventricular tachycardia) occurred in the early stage of transplantation. This arrhythmia showed time-dependent decreasing (Shiba et al., 2016; Romagnuolo et al., 2019). The induction of arrhythmia occurs due to the fact that the transplanted CMs in the early postoperative period are still immature compared to in vivo CMs. After transplantation, over the course of in vivo maturation of CMs, the incidence of arrhythmia gradually dissipated. Based on this finding, it is expected that graft-related arrhythmias occurring in the early postoperative period can be overcome if maturation of hPSC-derived CMs can be achieved prior to transplantation. Therefore, in this review, 1) an overview of the current progress of clinical studies using human PSC-derived CPCs or CMs is presented, 2) characteristics of immature CMs and mature CMs are comparatively analyzed, and 3) indicators for enhancement of maturation for key techniques of CM maturation are identified. Based on the findings of this review, we aim to present an effective strategy for the development of an optimal maturation technology for CMs.

An overview of clinical trials for cell therapy using hPSC-derived CPCs or CMs

Following the pioneering step of Mesoblast Ltd in 2005 involving a clinical trial using mesenchymal stem cells (MSCs), active research has been ongoing for the development of cell therapy for the treatment of HF and other heart conditions (Hare et al., 2009). However, after Mesoblast Ltd.’s reported failure in meeting the primary endpoint in phase 3 clinical trial, cell therapies using hPSC-derived CMs have drawn increasing interest for research and clinical applications. Unlike MSCs, CMs can directly participate in actual heart functions following engraftment at the transplant site, indicating a higher applicability and significance. Based on this perspective, the present review aims to investigate the current status of clinical trials using PSC-derived CMs (Table 1).

Table 1 . Clinical trials with hPSC-derived CPCs or cardiomyocytes

Institution (Nation)Assistance Publique (France)Osaka University (Japan)Heartseed (Japan)
Status (Term)Phase 1 completed (2013.05.27-2018.03.22)Phase 1 undergo (2019.12.02-2023.05.30)Phase 1 undergo (2022.04.19-2024.03.31)
Target diseaseIschemic Heart DiseaseMyocardial IschemiaHeart Failure
Patient10 patients for 5 years10 patients for 3 years10 patients for 5 years
Cell sourceESCAllogeneic iPSCAllogeneic iPSC
Cell typeCPC (SSEA1+ IsI-1+)CardiomyocyteCardiomyocyte
Transplantation20 cm2 Fibrin patch embedding
hESC-derived CPCs
hiPSC-derived cardiomyocyte sheetshiPSC-derived cardiomyocyte spheroids
Clinical mechanismImproves heart function by releasing factors that promote myocardial and/or vascular regeneration1) Helps heart function through self-beating
2) Improves heart function by releasing factors that promote myocardial and/or vascular regeneration
1) Helps heart function through self-beating
2) Improves heart function by releasing factors that promote myocardial and/or vascular regeneration
Outcome measures1 year follow-up after surgery:
1) Feasibility of patch’s generation and its efficacy on cardiac functions
2) Evidence for new clinical/biological abnormalities, occurrence of arrhythmias or development of a cardiac or extra-cardiac tumor
1 year follow-up after surgery:
1) Left ventricular (LV) ejection fraction, contractile and remodeling of the LV, New York Heart Association functional classification
2) Serious adverse events, abnormal blood biochemical or tumor marker tests and cardiac function clinical events
3) Minnesota Living with Heart Failure Questionnaire, 36-Item Short Form Survey etc.
1 year follow-up after surgery:
1) LV ejection fraction, myocardial wall motion evaluation, myocardial blood flow, myocardial viability
2) Safety and Tolerability
3) Kansas City Cardiomyopathy Questionnaire etc.
Result1. Cardiac function improvement effect
2. No arrhythmia, teratoma and immune-suppressant complications
OngoingOngoing
Pre/Clinical publicationMenasché et al., 2018Kashiyama et al., 2019Kawaguchi et al., 2021

1) A clinical study with hESC-derived CPCs

Regarding cell therapy using hESC-derived cardiovascular progenitors, a phase 1 trial conducted by a research team in France has been reported (Menasché et al., 2018). In the trial, from 2013 to 2018, ten patients with ischemic heart disease received hESC-derived CPCs by epicardial delivery of the cell-laden fibrin patch, and the clinical outcomes of the treatment were monitored. A 20 cm2 fibrin patch with a dose of 5 × 106 to 1 × 107 (Median: 8.2 × 106) hESC-derived CPCs was delivered to the epicardium of the infarct area. A year of follow-up monitoring revealed that one patient died because of treatment-unrelated comorbidities, but all others had uneventful recoveries. In all patients apart from the deceased patient, symptomatic improvement with an increased systolic motion of the cell-treated segments was confirmed, and there were no arrhythmias, complications of the immunosuppressive treatment, or intracardiac or off-target tumors. From these results, it can be interpreted that various factors released from the grafted CPCs promote myocardial and vascular regeneration, and such regeneration contributed to improving the function of infarcted hearts. However, considering that one patient died of heart failure 22 months post-transplantation, it is speculated that there is a limitation to achieving cardiac repair to the level of healthy adults by transplantation of CPCs.

2) A clinical study with hiPSC-derived cardiomyocyte sheets

Clinical trials on cell therapy using hiPSC-derived CMs have been conducted by two Japanese research teams. One of the research teams at Osaka university generated iPSC-CM sheets using cynomolgus macaque to induce cardiomyogenic differentiation in a preclinical study. The generated CMs were in the form of cell sheets (3.6 × 106 cells/sheet), and four pieces of the iPSC-cardiac sheets were transplanted in a cynomolgus macaque MI model. The effect of transplantation was evaluated through a 6-month follow-up monitoring. As a result, it was confirmed that cardiac function improved regardless of the matching/mismatching of the major histocompatibility complex (Kashiyama et al., 2019). Based on this preceding study, the research team is conducting a phase 1 trial to investigate the therapeutic effect of CM sheets derived from allogeneic iPSCs for 10 patients with myocardial ischemia for 3 years since 2019. Through 1-year follow-up post-transplantation, the treatment effect including functional and structural recoveries of the left ventricle, as well as adverse events including abnormal findings in the blood biochemical tests or cardiac function events and tumor development, will be examined for evaluation.

3) A clinical study with hiPSC-derived cardiomyocyte aggregates

Another ongoing clinical trial using hiPSC-derived CMs is being conducted by Tokyo-based Heartseed. First, a preclinical study was conducted using a rat model (i.e. a small animal model) and a micro-miniature pig model (i.e. a large animal model). Cardiac spheroids (200 μm) consisting of approximately 1,000 hiPSC-derived CMs were prepared, and 3,000 spheroids (total cell number: 3 × 106) and 1 × 105 spheroids (total cell number: 1 × 108) were transplanted in the rat heart failure model and the pig heart failure model, respectively, to evaluate the effect of transplantation. As a result, the cardiac spheroid transplantation group maintained the recovery of cardiac function for a longer period of time in the large animal model (Kawaguchi et al., 2021). Based on this preceding study, a phase 1 trial is underway to determine the effect of iPSC-derived cardiomyocyte spheroids on 10 heart failure (HF) patients for 5 years starting from 2022. Each patient will undergo transplantation of cardiac spheroids, and through 1-year post transplantation follow-up, adverse events in terms of safety, tolerability, and cardiac function recovery with outcome measures such as myocardial viability and myocardial blood flow will be evaluated.

Comparison of the characteristics between immature cardiomyocytes and mature cardiomyocytes

There is a general consensus among stem cell researchers on the immature state of hPSC-derived CMs, but methods for evaluating the degree of maturation remain to be determined. Therefore, in addition to developing methods for enhancing the maturation of hPSC-derived CMs, methods for evaluating the degree of maturation also need to be established. As a first step, in this review, we comparatively analyzed the characteristics of mature CMs in adult heart and immature (fetal and hPSC-derived) CMs (Table 2).

Table 2 . Clinical trials with hPSC-derived CPCs or cardiomyocytes

CharacteristicsCategoryCardiomyocytesReferences

Immature (Fetal or PSC-derived)Mature
MorphologyShapeCircular and oblongAnisotropic rod-likeLundy et al., 2013
SizeDiameters: 5-10 μm
Heights: ~5 μm
Length: 30 μm
Width: 10 μm
Volume: 2,000 μm3
Heights: 15 μm
Length: 150 μm
Width: 20 μm
Volume: 40,000 μm3
Bulatovic et al., 2016
Lundy et al., 2013
NucleusMononucleated diploidMultinucleated tetraploidDenning et al., 2016
Structure (Cytoskeleton)Sarcomere: disorganized, short
SR and T-tubule: poorly developed
Sarcomere: highly organized, long
SR and T-tubule: well-developed
Denning et al., 2016
Yang et al., 2014a
ElectrophysiologyBeatingSpontaneously beat
Force: 0.08-4 mN/mm2
mixed action potential (nodal, atrial ventricular-like)
Start: Sinoatrial node
No beat until triggered by the depolarization form adjacent cells
Force: 40-80 mN/mm2
Lin et al., 2017
Resting membrane potential-60 mV, nodal like-90 mVKoivumäki et al., 2018
Conduction velocity10-20 cm/sAround 60 cm/sDenning et al., 2016
Upstroke velocitySlow, 10-50 V/sFast, 150-350 V/sDenning et al., 2016
Veerman et al., 2017
RepolarizationFastSlow, after plateau phaseJeevaratnam et al., 2018
Zhao et al., 2018
Channel proteinINa, ICa-L, Ito
Low IK1,
INa, ICa-L, Ito, IKr, IKsGoversen et al., 2018
Calcium handling1) Similar expression of Na+/Ca2+ exchanger with adult CMs
2) Low Expression of other Ca2+ releasing related protein
3) Increasing of calcium stores in the SR
4) Slower calcium dynamics with delayed time to peak and slower decay of the calcium signal
Depolarization → opening of LTCC → calcium influx → calcium-induced calcium releasing → sliding of myofilament and contraction of muscle
Calcium releasing:
1) SERCA2a
2) Sarcolemmal Na+/Ca2+ exchanger
3) Sarcolemmal Ca2+-ATPase
4) Mitochondrial Ca2+ uniport
Hwang et al., 2015
Karakikes et al., 2015
Youm, 2016
Eisner et al., 2017
MetabolismMitochondriaLocation: perinuclear space
Low number and small size (<5% of total cell volume)
Round shape and low cristae density
In mouse, opened mitochondrial permeability transition (MPT) not induce cytochrome c leakage or apoptosis
Location: between myofibrils and under the sarcolemma
increase in both size and number
(~30% of total cell volume)
Oval shape and dense cristae → sufficient surface area
Feric and Radisic, 2016
Energy sourceEnergy was generated mainly by glycolysis(Low fatty acid/ High glucose or lactate)
Approximately 15% of total energy consumption in fetal cardiomyocytes is supplied with β-oxidation of fatty acids
Approximately 80% of total energy consumption in adult cardiomyocytes is supplied with β-oxidation of fatty acids
60-70% ATP for contractile function, followed SERCA and of other ion transporters
Correia et al., 2017
Galdos et al., 2017
Karakikes et al., 2015
Piquereau et al., 2018
Gene expressionSarcomeric gene (MHC)α-MHCβ-MHCKatrukha, 2013
Lundy et al., 2013
Troponin ITNNI1TNNI3Katrukha, 2013
OthersLow expression levels of SERCA2, Caveolin 3, KCNH2 etc.Lundy et al., 2013

1) Morphology and cellular structure

Mature cardiomyocytes (CMs) in adult hearts have rod-like shapes, and the size of a single CM is about 150 μm in length, 20 μm in width, and 15 μm in height, with a volume of 40,000 μm3 (Bulatovic et al., 2016). Regarding the morphology of the nucleus, mature CMs are multinucleated and have tetraploid nuclei. They are characterized by highly organized sarcomere and well-developed sarcoplasmic reticulum (SR) and transverse tubules (T-tubules) (Yang et al., 2014a). In contrast, immature hPSC-derived CMs have different shapes such as a circular or oblong geometry, and they are approximately 5-10 μm in diameter, 30 μm in length, 10 μm in width, and 5 μm in height, with a volume of 2,000 μm3, showing a smaller size compared to that of mature CMs (Lundy et al., 2013, Bulatovic et al., 2016). Regarding the nucleus morphology, the immature CMs are mononucleated or have diploid nuclei as in the case of other somatic cells. Regarding the structure of the cytoskeleton, the sarcomere is disorganized and shows irregular distribution in the cytoplasm. Moreover, following induction of CM differentiation, no or few T-tubules are observed in hPSC-derived CMs in the early stage of beating (Yang et al., 2014a; Denning et al., 2016; Park et al.,2022) (Fig. 1A).

Figure 1. Experimental methods for maturation of hPSC-derived cardiomyocytes. (A) Compared to matured cardiomyocytes, morphology and structure, calcium handling, metabolism, and electrophysiology are different in immature hPSC-CMs. (B) Various methods of maturation hPSC-CMs.
2) Electrophysiological properties

In general, adult CMs maintain a resting membrane potential of -90 mV until electrical impulses are transmitted from adjacent cells. When triggered by the adjacent cells, fast Na+ channels begin to open, and Na+ leaks into the cell. This produces rapid depolarization with the transmembrane potential rising to approximately +20 mV. The upstroke velocity at this time is measured to be approximately 150-350 V/s (Lin et al., 2017). After depolarization, an outflow of K+ occurs due to diffusion through the cell membrane, but with a constant inward current of Ca2+, the plateau phase is maintained for some time (Liu et al., 2016; Sharifi et al., 2017; Zhao et al., 2018). Next, Ca2+ channels are gradually inactivated, and with a persistent outflow of K+, the transmembrane potential is brought back toward a resting potential of -90mV, thereby completing a cycle. Subsequently, depolarization newly starts when triggered by the adjacent cells (Goversen et al., 2018; Koivumäki et al., 2018). In adult CMs, the action potential duration of one cycle is measured to be ~300 ms, and the conduction velocity, i.e. the velocity of transmission of electrical impulses to adjacent cells, is known to be approximately 50 cm/s (Tveito et al., 2012). In contrast, hPSC-derived CMs showed spontaneous beating, with a resting membrane potential of approximately -60 mV, similar to that of nodal CMs. In addition, when depolarization occurs, the subunits of Nav1.5 protein involved in the inflow of sodium ions are fetal isoforms, and the upstroke velocity is measured at approximately 10-50 V/s, slower than that of adult CMs (Veerman et al., 2017). Furthermore, since IKI, a type of channel protein in adult CMs that promotes K+ inflow and inhibits its outflow, is present at a low concentration in hPSC-derived CMs, K+ ion outflow occurs at a faster rate after depolarization than that in adult CMs. This causes rapid repolarization in which the transmembrane potential is brought back to the resting membrane potential after depolarization (Jeevaratnam et al., 2018).

3) Calcium handling

Calcium ions (Ca2+) play an important role in various signal transduction pathways, metabolism, and transcriptional regulation in cells, and in the case of myocytes, the intracellular Ca2+ concentration is also involved in the contraction and relaxation of the cardiac muscle. This indicates that Ca2+ also plays an important role in CMs, which is also a type of myocyte. In mature CMs, the concentration of Ca2+ is precisely controlled through various mechanisms. In the excitation-contraction coupling, when intracellular depolarization occurs, the L-type calcium channel (LTCC) opens, producing an inward current of Ca2+ from outside the cell membrane to the cytoplasm, and the increased cytoplasmic Ca2+ concentration induces the release of calcium-induced Ca2+ through ryanodine receptors (RYR) in the SR, thereby further increasing the cytoplasmic Ca2+ concentration. Ca2+ with increased cytoplasmic concentration binds to the troponin protein to induce muscle contraction. After the contraction is completed, cytoplasmic Ca2+ concentration is lowered again by removing Ca2+ from the cytoplasm through the following process: (1) Ca2+ is pumped back into SR via sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a); (2) Ca2+ is pumped out of the cell by the sodium-calcium exchanger; (3) Ca2+ uptake into mitochondria; (4) additional leak of Ca2+ by sarcolemmal calcium ATPase (Eisner et al., 2017). In the case of immature CMs lacking T-tubules, LTCC and RYR are separated in space, leading to a delay in the release of Ca2+ in the SR after depolarization, and also, the expression of Ca2+ handling protein is also lower than that in mature CMs, exhibiting a slower calcium dynamic overall. However, among the Ca2+ handling proteins, the expression of sodium-calcium exchanger is higher in immature fetal CMs than in mature CMs, and this characteristic can also be observed in the case of hPSC-derived CMs (Hwang et al., 2015).

4) Metabolism

The human heart requires a continuous supply of energy to maintain its ongoing pumping activity with systolic and diastolic movements from birth to death. Accordingly, the cardiac energy metabolism in CMs is highly effective. The typical features of fetal cardiac mitochondria include a low number and a small size, occupying <5% of the total cell volume. With the development of the fetal heart, both the number and size of the cardiac mitochondria show a robust increase, accounting for ~30% of the cell volume (Galdos et al., 2017). The differences can also be observed in terms of the morphology and location of the individual mitochondria with cardiomyocyte maturation. For mature CMs, mitochondria take a long, oval shape and are located between myofibrils and under the sarcolemma with the formation of dense cristae (Feric and Radisic, 2016). For immature CMs, round-shaped mitochondria are mainly located in the perinuclear space, with rudimentary cristae in the inner membrane of mitochondria. In terms of energy production through cellular metabolism with mitochondria having the central role, approximately 80% of the total energy in mature CMs is generated through β-oxidation metabolism of fatty acids (Karakikes et al., 2015; Piquereau et al., 2018). In the case of immature CMs, approximately only 15% of the energy is produced through β-oxidation of fatty acids, and in many cases, energy is generated through the metabolism of glucose or lactate (Correia et al., 2017). Since hPSC-derived CMs also exist in an immature state, their metabolism largely depends on glycolysis rather than β-oxidation of fatty acids.

5) Gene expression

Mature CMs and immature CMs show differences in terms of subunits of specific genes (proteins) in CMs. Myosin heavy chains (MHCs) are motor proteins that convert chemical energy derived from the hydrolysis of ATP into mechanical force that drives diverse motile processes in cardiac and skeletal muscle. MHCs are also expressed in two different, developmentally-regulated subunits: the fetal α-isoform (α-MHC) and the adult β-isoform (β-MHC). In mature CMs, β-MHC synthesized by the expression of the MYH7 gene is predominant, and in immature CMs, α-MHC synthesized by the expression of the MYH6 gene is mainly expressed (Katrukha, 2013; Lundy et al., 2013). Troponin I (TnI) behaves as a molecular switch of sarcomere within the myocytes and typically has three isoforms (TNNI1: Slow skeletal TnI, TNNI2: Fast skeletal TnI, TNNI3: Cardiac TnI). In mature CMs, cardiac TnI3 is predominantly expressed, whereas in immature CMs, slow skeletal TnI2 is predominantly expressed (Katrukha, 2013). In addition, the expression of genes such as SERCA2, Caveolin 3, and KCNH2 was lower in immature CMs (Lundy et al., 2013).

Methods of promoting maturation of cardiomyocytes

As described above, although there have been numerous studies on the generation and utilization of PSC-derived CMs, these cells show characteristics similar to those of immature fetal CMs. Accordingly, various approaches for obtaining mature cardiomyocytes by inducing in vitro maturation of PSC-derived CMs have been actively investigated. The representative maturation methods in the current research trend are outlined as shown below (Fig. 1B)

1) Long-term culture of cardiomyocytes

In the course of human heart development from fetal to adult CMs, the duration of maturation for fetal immature CMs from birth is over 10 years (Bulatovic et al., 2016). Accordingly, a number of studies have been conducted by different research teams to enhance the maturation of CMs through in-vitro long-term culture of hPSC-derived CMs (Shinozawa et al., 2012; Kamakura et al., 2013; Lundy et al. al., 2013; Cyganek et al., 2018). Through the long-term culture process lasting from 60 days to 1 year, the number of cells expressing the cTnT gene (cTnT+), a representative marker of CMs, and multinucleated cells increase, and CMs undergo morphological changes such as the increase in the sarcomere length and cell size with an increase in the expression of associated genes. Thus, it was confirmed that the maturity of hPSC-derived CMs was enhanced with time. In particular, formation of M-bands was confirmed in CMs following a long-term culture of 360 days or longer, but the amount of sarcomeric structures in a single CM was still scarce compared with that of adult CMs (Kamakura et al., 2013). The findings that hPSC-derived CMs did not reach the full level of maturation as that of adult CMs require additional investigation on other factors such as in vivo paracrine and hormone signals. In a recent study, during the initial process of CM differentiation, platelet-derived growth factor receptor-α (PDGFRα)-positive cells that can respond to CM differentiation cues and are thus known as one of the markers of CM differentiation were first collected, and subsequently, long-term culture of immature CMs was conducted. Through this method, the formation of a T-tubule-like structure, which was not confirmed in previous studies, was exhibited on day 231 of the long-term culture (Fukushima et al., 2020).

2) Addition of maturation factor

From the viewpoint of evolution, the closer the relation to mammals in the phylogenetic tree, the serum thyroxine level increases and the cardiac regenerative potential decreases, showing the inverse correlation between the serum thyroxine level and cardiac regenerative potential. Inactivation of thyroid hormone signaling reduced mouse cardiomyocyte polyploidization, and the mouse CMs remain in the immature state for longer (Hirose et al., 2019). This implies that regulation of thyroid hormones may serve to limit the CM regenerative capacity and induce CM polyploidization, thereby promoting the maturation of CMs. Accordingly, recent studies have investigated the method of adding Tri-iodo-L-thyronine (T3), a thyroid hormone, during the culture of CMs to induce in vitro maturation of CMs.

In a number of studies, the results showed that treatment with T3-induced gene expression of hPSC-derived CMs from the fetal-like level to the adult-like pattern increased the force per contractile along with the rate of calcium release and reuptake with SERCA2α to promote further maturation of CMs (Ivashchenko et al., 2013; Yang et al., 2014b; Birket et al., 2015). In addition, T3 treatment in combination with IGF-1, Dexamethasone increased the expression of PGC-1a and PGC-1b, which are important regulators of fatty acid oxidation (FAO) and mitochondrial function in CMs, and contributed to the formation of a uniform sarcomeric structure, thereby inducing the maturation of immature CMs.

3) 3-Dimensional (3D) cell culture methods

All cells exist in 3D space in vivo, but standard cell culture is performed in 2D, typically in a flat petri dish. This indicates that cells are cultured under conditions different from those of in vivo environments, which may lead to differences in cellular responses and functions compared to those observed in vivo. Therefore, research has been conducted on culturing hPSC-CMs in 3D systems and not in the conventional 2D systems. On day 6 of hPSC-CM differentiation, the cardiac progenitors were recovered, and the 3D aggregation of the hPSC-derived cardiac progenitors was induced. After a 29-day culture, the maturation of 3D-cultured CMs was compared with that of 2D-cultured CMs. In the case of CMs in aggregates, contractility was more than three times faster, and improved action potential kinetics were achieved. Moreover, in terms of gene expression with 3D culture, the expression of atrial-like CM markers decreased, and the expression of ventricular-like CM markers increased (Correia et al, 2018).

Among the 3D culture methods, improved maturation of CMs was reported for a culture of engineered heart tissues (EHTs). The hPSCs were differentiated into CMs, and after 30 days, hPSC-derived CMs were cultured in the form of engineered cardiac tissue constructs, a type of EHTs. Using the EHT culture method for 3 weeks without other methods to promote maturation apart from the 3D culture, T-tubule systems similar to the ventricular CMs of the adult heart were developed, and within a relatively short time of 2 weeks, an increase in expression of various genes related to calcium-handling and contraction was confirmed (de Lange et al, 2021). In terms of cell therapy, 3D culture has advantages over 2D culture. In both the results of a preclinical study (Kawaguchi et al., 2021) published by Heartseed, based on which a phase 1 trial is underway, and a recent report published by the authors’ research team, a single CM or CM aggregates were grafted on the infarcted area of an animal model with MI to evaluate the effect of transplantation. As a result, when CMs were transplanted in the form of 3D aggregates, the in vivo survival rate was higher than that of single CM transplantation, and the results also showed that the engraftment of mature CMs was induced, and the method was more effective in improving the symptoms of MI (Park et al., 2022).

4) Various other cell co-culture methods

As with all other organs in the human body, the heart does not consist of CMs alone but includes various other cells such as endothelial cells (Ecs), MSCs, and fibroblasts. The coexistence of different types of cells means that each cell can influence each other through intracellular interactions, and such interactions are also involved in the maturation of CMs (Zhang et al., 2012). Therefore, research has been conducted to investigate the effect of inducing CM maturation using a co-culture system in which hPSC-derived CMs are cultured with other types of cells. On comparing three different culturing methods of CMs: (1) hiPSC-CMs cultured alone, (2) hiPSC-CM co-cultured with MSC, and (3) addition of MSC-derived soluble factors, both the co-culture group and the soluble factor addition group showed higher structural and functional maturity of CMs (e.g. cell size, sarcomere length, contractility, electrophysiology) compared to the CMs cultured alone (Yoshida et al., 2018). The results confirmed that among CM culture methods, co-culture with MSCs promoted the maturation of CMs by releasing various bioactive factors favorable for cardiac cells. In addition to MSCs, endothelial cells (Ecs), whose number is approximately three times higher than that of CMs in the human heart, are also known to play a significant role in the early-stage heart development or cardiac repair (Talman et al., 2018). Based on this, during hPSC-CM differentiation, CMs were co-cultured with Ecs and after 2 weeks of additional culture, the maturity of cultured CMs was evaluated based on the cell size, sarcomere organization, and gene expression related to mature CMs (Dunn et al., 2019). The results confirmed that the maturity of CMs was higher when co-cultured with CPCs in the early or mid-stage of CM differentiation than in the case of CPC monoculture or co-culture with EC of CMs after the completion of differentiation. The results indicate that Ecs induce maturation of CMs by delivering microRNA through paracrine factors and gap junctions in immature CMs. In addition, when microtissues were prepared using 3D co-culture with CPC, EC, and MSC in a ratio of 2:1:1, respectively, and cultured for 1 week, CMs maturation was accelerated compared to that of microtissues with CPC alone. (Varzideh et al, 2019). For transplantation of hiPSC-derived CMs to an animal MI model, 3D printing was used to prepare hMSC-loaded patch, and the patch was epicardially implanted. As a result, the hMSC-secreted factors improved the survival of hiPSC-CMs compared to the transplantation of CMs alone, and the method was also effective in enhancing vascular regeneration and promoting the maturation of CMs (Park et al., 2019).

5) Electric stimulation of cardiomyocytes

In this maturation method, cardiac cells were isolated from rats to investigate whether the morphology and functionalities of CMs changed (developed) similar to those of in vivo CMs of rats with or without electric stimulation under in vitro culture environment (Lasher et al., 2012). To compare differences in the maturation of CMs with/without electric stimulation, CMs with electric stimulation of 2 ms symmetric biphasic square pulses, 4 V/cm, and 1 Hz for 9 days using a carbon-rod of scm-spacing were compared with CMs without electric stimulation on days 3, 6, and 9. In terms of morphology, CMs applied with electric stimulation showed more similar values of length, width, and height with the age-matched myocardium compared to those of CMs without electric stimulation. In terms of functionalities, the decreased excitation threshold and increased maximum capture rate were observed in CMs with electric stimulation, which are the indicators of maturation. In a study where electric stimulation of 5 ms pulse, 5 V/cm, and 2 Hz was applied to iPSC-derived CMs to induce maturation of CMs, the result confirmed that maturation of CMs can be induced by electric stimulation in terms of cell alignment, contractility and passive stiffness, and the increase in expression of calcium-handling protein (Ruan et al., 2016).

With CVDs reported as the leading cause of death worldwide, development of cell therapy that enables the fundamental treatment of CVDs has gained increasing attention. In particular, there is an ever-increasing importance of cell therapy based on PSC-derived CMs because the method can be applied for various diseases in the heart, a representative organ of the cardiovascular system, and the method allows unlimited (mass) generation of CMs with extremely low regenerative capacity. Therefore, in this review, the current status of clinical trials using PSC-derived CPCs and CMs as cell therapy was outlined, and the different characteristics of mature adult CMs and immature CMs were comparatively analyzed. Furthermore, since PSC-derived CMs are in an immature state, various culture methods that promote maturation of CMs were discussed. A number of studies have shown that immature PSC-derived CMs can mature into having properties similar to those of adult CMs by implementing various maturation methods. Therefore, the authors’ research team aims to develop a more effective method of promoting the maturation of CMs by analysis and incorporating the results of the recent publications and applying an optimal combination of these different methods. Furthermore, with additional implementation of a co-culture system including MSC and EC using 3D printing techniques, the establishment of a future direction for CM engineering by forming surface modification using bioinks, and creation of a 3D-printed cardiac model, it is expected that a model with the same morphology and functionality as in vivo cardiomyocytes can be developed with more efficient intercellular signal transduction and synchronization of the maturation level of individual CMs (Fig. 2). Although challenges still remain to be overcome, continuous efforts on the technological development of maturation methods are expected to present a new opportunity for cardiovascular research, and considering the track record of the emergence of new technologies and analytical tools up until the techniques developed to date, the new technologies and analytical tools are expected to create synergistic effects. If the technology for maturation of hPSC-derived CMs into cells with the same functional and structural characteristics as those of mature CMs in vivo can be developed through various approaches as described above, it is expected that the effects of CM-based cell therapy can be clearly established along with extensive utilization of the developed maturation technology in areas of basic research, including the analysis of cardiac functions, and for various clinical applications, including cardiotoxicity evaluation of drug candidates.

Figure 2. The new strategy to mature hPSC-derived cardiomyocytes into the identical characteristic cardiomyocytes in adult heart through a combination of cardiomyocyte maturation methods.

We thanks to Prof. Seong-Woo Choi of Dongguk University College of Medicine, Na-Kyeong Park of Seoul National University College of Medicine, Dr. Seok-Yun Jung and Mi-Jeong Kim of Stem Cell Research Institute, T&R Biofab Co. Ltd, Republic of Korea for their insightful comments on this manuscript.

This research was supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI20C0184), and the Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korea government (the Ministry of Science and ICT, the Ministry of Health & Welfare) (21A0403L1).

  1. Abbasi J. 2022. The COVID heart-one year after SARS-CoV-2 infection, patients have an array of increased cardiovascular risks. JAMA 327:1113-1114.
    Pubmed CrossRef
  2. Bedada FB, Chan SS, Metzger SK, Zhang L, Zhang J, Garry DJ, Kamp TJ, Metzger JM. 2014. Acquisition of a quantitative, stoichiometrically conserved ratiometric marker of maturation status in stem cell-derived cardiac myocytes. Stem Cell Reports 3:594-605.
    Pubmed KoreaMed CrossRef
  3. Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR, van de Pol V, Dambrot C, Devalla HD, Davis RP, Mastroberardino PG, Atsma DE, Mummery CL. 2015. Contractile defect caused by mutation in MYBPC3 revealed under conditions optimized for human PSC-cardiomyocyte function. Cell Rep. 13:733-745.
    Pubmed KoreaMed CrossRef
  4. Bulatovic I, Månsson-Broberg A, Grinnemo KH. 2016. Human fetal cardiac progenitors: the role of stem cells and progenitors in the fetal and adult heart. Best Pract. Res. Clin. Obstet. Gynaecol. 31:58-68.
    Pubmed CrossRef
  5. Burridge PW, Wu JC. 2015. Chemically defined culture and cardiomyocyte differentiation of human pluripotent stem cells. Curr. Protoc. Hum. Genet. 87:21.3.1-21.3. 15.
    Pubmed KoreaMed CrossRef
  6. Choi SW, Shin JS, Park SJ, Jung E, Park YG, Lee J, Kim SJ, Park HJ, Lee JH, Park SM, Moon SH, Go YY. 2020. Antiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytes. Antiviral Res. 184:104955.
    Pubmed KoreaMed CrossRef
  7. Correia C, Koshkin A, Duarte P, Hu D, Carido M, Sebastião MJ, Gomes-Alves P, Elliott DA, Domian IJ, Teixeira AP, Serra M. 2018. 3D aggregate culture improves metabolic maturation of human pluripotent stem cell derived cardiomyocytes. Biotechnol. Bioeng. 115:630-644.
    Pubmed CrossRef
  8. Correia C, Koshkin A, Duarte P, Hu D, Teixeira A, Domian I, Alves PM. 2017. Distinct carbon sources affect structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Sci. Rep. 7:8590.
    Pubmed KoreaMed CrossRef
  9. Cyganek L, Tiburcy M, Sekeres K, Gerstenberg K, Bohnenberger H, Lenz C, Henze S, Stauske M, Salinas G, Zimmermann WH, Guan K. 2018. Deep phenotyping of human induced pluripotent stem cell-derived atrial and ventricular cardiomyocytes. JCI Insight 3:e99941.
    Pubmed KoreaMed CrossRef
  10. de Lange WJ, Farrell ET, Kreitzer CR, Jacobs DR, Lang D, Ralphe JC. 2021. Human iPSC-engineered cardiac tissue platform faithfully models important cardiac physiology. Am. J. Physiol. Heart Circ. Physiol. 320:H1670-H1686.
    Pubmed KoreaMed CrossRef
  11. Denning C, Borgdorff V, Crutchley J, Firth KS, George V, Kalra S, Kondrashov A, Hoang MD, Mosqueira D, Patel A, Prodanov L, Rajamohan D, Skarnes WC, Young LE. 2016. Cardiomyocytes from human pluripotent stem cells: from laboratory curiosity to industrial biomedical platform. Biochim. Biophys Acta 1863(7 Pt B):1728-1748.
    Pubmed KoreaMed CrossRef
  12. Dunn KK, Reichardt IM, Simmons AD, Jin G, Floy ME, Palecek SP. 2019. Coculture of endothelial cells with human pluripotent stem cell-derived cardiac progenitors reveals a differentiation stage-specific enhancement of cardiomyocyte maturation. Biotechnol. J. 14:e1800725.
    Pubmed KoreaMed CrossRef
  13. Eisner DA, Caldwell JL, Trafford AW. 2017. Calcium and excitation-contraction coupling in the heart. Circ. Res. 121:181-195.
    Pubmed KoreaMed CrossRef
  14. Feric NT and Radisic M. 2016. Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Adv. Drug Deliv. Rev. 96:110-134.
    Pubmed KoreaMed CrossRef
  15. Fukushima H, Yoshioka M, Kawatou M, López-Dávila V, Takeda M, Kanda Y, Sekino Y, Yamashita JK. 2020. Specific induction and long-term maintenance of high purity ventricular cardiomyocytes from human induced pluripotent stem cells. PLoS One 15:e0241287.
    Pubmed KoreaMed CrossRef
  16. Galdos FX, Guo Y, Paige SL, VanDusen NJ, Pu WT. 2017. Cardiac regeneration: lessons from development. Circ. Res. 120:941-959.
    Pubmed KoreaMed CrossRef
  17. Goversen B, van der Heyden MAG, de Boer TP. 2018. The immature electrophysiological phenotype of iPSC-CMs still hampers in vitro drug screening: special focus on IK1. Pharmacol. Ther. 183:127-136.
    Pubmed CrossRef
  18. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB Jr, Reisman MA, Sherman W. 2009. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J. Am. Coll. Cardiol. 54:2277-2286.
    Pubmed KoreaMed CrossRef
  19. Hirose K, Payumo AY, Cutie S, Hoang A, Zhang H, Guyot R, Lunn D, Bigley RB, Yu H, Wang J, Smith M, Gillett E, Muroy SE, Schmid T, Wilson E, Field KA, Reeder DM, Maden M, Yartsev MM, Wolfgang MJ, Grützner F, Scanlan TS, Szweda LI, Buffenstein R, Hu G, Flamant F, Huang GN. 2019. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 364:184-188.
    Pubmed KoreaMed CrossRef
  20. Hwang HS, Kryshtal DO, Feaster TK, Sánchez-Freire V, Zhang J, Kamp TJ, Hong CC, Knollmann BC. 2015. Comparable calcium handling of human iPSC-derived cardiomyocytes generated by multiple laboratories. J. Mol. Cell. Cardiol. 85:79-88.
    Pubmed KoreaMed CrossRef
  21. Ivashchenko CY, Pipes GC, Lozinskaya IM, Lin Z, Xiaoping X, Needle S, Grygielko ET, Hu E, Toomey JR, Willette RN. 2013. Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype. Am. J. Physiol. Heart Circ. Physiol. 305:H913-H922.
    Pubmed CrossRef
  22. Jeevaratnam K, Chadda KR, Camm AJ. 2018. Cardiac potassium channels: physiological insights for targeted therapy. J. Cardiovasc. Pharmacol. Ther. 23:119-129.
    Pubmed KoreaMed CrossRef
  23. Kamakura T, Makiyama T, Sasaki K, Yoshida Y, Wuriyanghai Y, Chen J, Hattori T, Ohno S, Kita T, Horie M, Kimura T. 2013. Ultrastructural maturation of human-induced pluripotent stem cell-derived cardiomyocytes in a long-term culture. Circ. J. 77:1307-1314.
    Pubmed CrossRef
  24. Karakikes I, Ameen M, Wu JC. 2015. Human induced pluripotent stem cell-derived cardiomyocytes: insights into molecular, cellular, and functional phenotypes. Circ. Res. 117:80-88.
    Pubmed KoreaMed CrossRef
  25. Kashiyama N, Miyagawa S, Fukushima S, Kawamura T, Kawamura A, Yoshida S, Eiraku S, Harada A, Matsunaga K, Watabe T, Toda K, Sawa Y. 2019. MHC-mismatched allotransplantation of induced pluripotent stem cell-derived cardiomyocyte sheets to improve cardiac function in a primate ischemic cardiomyopathy model. Transplantation 103:1582-1590.
    Pubmed CrossRef
  26. Katrukha IA. 2013. Human cardiac troponin complex. Structure and functions. Biochemistry (Mosc.) 78:1447-1465.
    Pubmed CrossRef
  27. Kawaguchi S, Soma Y, Nakajima K, Kanazawa H, Tohyama S, Tabei R, Hirano A, Handa N, Yamada Y, Okuda S, Hishikawa S, Teratani T, Kunita S, Kishino Y, Okada M, Tanosaki S, Someya S, Morita Y, Tani H, Kawai Y, Yamazaki M, Ito A, Shibata R, Murohara T, Tabata Y, Kobayashi E, Shimizu H, Fujita J. 2021. Intramyocardial transplantation of human iPS cell-derived cardiac spheroids improves cardiac function in heart failure animals. JACC Basic Transl. Sci. 6:239-254.
    Pubmed KoreaMed CrossRef
  28. Koivumäki JT, Naumenko N, Tuomainen T, Takalo J, Oksanen M, Puttonen KA, Lehtonen Š, Kuusisto J, Laakso M, Tavi P. 2018. Structural immaturity of human iPSC-derived cardiomyocytes: in silico investigation of effects on function and disease modeling. Front. Physiol. 9:80.
    Pubmed KoreaMed CrossRef
  29. Lasher RA, Pahnke AQ, Johnson JM, Hitchcock RW. 2012. Electrical stimulation directs engineered cardiac tissue to an age-matched native phenotype. J. Tissue Eng. 3:2041731412455354.
    Pubmed KoreaMed CrossRef
  30. Lin ZC, McGuire AF, Burridge PW, Matsa E, Lou HY, Cui B. 2017. Accurate nanoelectrode recording of human pluripotent stem cell-derived cardiomyocytes for assaying drugs and modeling disease. Microsyst. Nanoeng. 3:16080.
    Pubmed KoreaMed CrossRef
  31. Liu J, Backx PH. 2016. The electrophysiological development of cardiomyocytes. Adv. Drug Deliv. Rev. 96:253-273.
    Pubmed CrossRef
  32. Lundy SD, Zhu WZ, Laflamme MA. 2013. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 22:1991-2002.
    Pubmed KoreaMed CrossRef
  33. Menasché P, Vanneaux V, Hagège A, Bel A, Cholley B, Parouchev A, Cacciapuoti I, Al-Daccak R, Benhamouda N, Blons H, Agbulut O, Tosca L, Trouvin JH, Fabreguettes JR, Bellamy V, Charron D, Tartour E, Tachdjian G, Larghero J. 2018. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J. Am. Coll. Cardiol. 71:429-438.
    Pubmed CrossRef
  34. Park SJ, Kim H, Lee S, Kim J, Jung TH, Choi SW, Park BW, Kang SW, Elliott DA, Stanley EG, Elefanty AG, Ban K, Moon SH. 2022. Effect and application of cryopreserved three-dimensional microcardiac spheroids in myocardial infarction therapy. Clin. Transl. Med. 12:e721.
    Pubmed KoreaMed CrossRef
  35. Park SJ, Kim RY, Park BW, Lee S, Choi SW, Park JH, Choi JJ, Kim SW, Jang J, Cho DW, Chung HM, Moon SH, Park HJ. 2019. Dual stem cell therapy synergistically improves cardiac function and vascular regeneration following myocardial infarction. Nat. Commun. 10:3123.
    Pubmed KoreaMed CrossRef
  36. Piquereau J and Ventura-Clapier R. 2018. Maturation of cardiac energy metabolism during perinatal development. Front. Physiol. 9:959.
    Pubmed KoreaMed CrossRef
  37. Romagnuolo R, Masoudpour H, Porta-Sánchez A, Qiang B, Barry J, Laskary A, Qi X, Massé S, Magtibay K, Kawajiri H, Wu J, Valdman Sadikov T, Rothberg J, Panchalingam KM, Titus E, Li RK, Zandstra PW, Wright GA, Nanthakumar K, Ghugre NR, Laflamme MA. 2019. Human embryonic stem cell-derived cardiomyocytes regenerate the infarcted pig heart but induce ventricular tachyarrhythmias. Stem Cell Reports 12:967-981.
    Pubmed KoreaMed CrossRef
  38. Ruan JL, Tulloch NL, Razumova MV, Saiget M, Muskheli V, Pabon L, Reinecke H, Murry CE. 2016. Mechanical stress conditioning and electrical stimulation promote contractility and force maturation of induced pluripotent stem cell-derived human cardiac tissue. Circulation 134:1557-1567.
    Pubmed KoreaMed CrossRef
  39. Sharifi M, Buzatu D, Wilkes J. 2017. Development of models for predicting Torsade de Pointes cardiac arrhythmias using perceptron neural networks. BMC Bioinformatics 18(Suppl 14:497.
    Pubmed KoreaMed CrossRef
  40. Shiba Y, Gomibuchi T, Seto T, Wada Y, Ichimura H, Tanaka Y, Ogasawara T, Okada K, Shiba N, Sakamoto K, Ido D, Shiina T, Ohkura M, Nakai J, Uno N, Kazuki Y, Oshimura M, Ikeda U. 2016. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 538:388-391.
    Pubmed CrossRef
  41. Shinozawa T, Imahashi K, Sawada H, Takami K. 2012. Determination of appropriate stage of human-induced pluripotent stem cell-derived cardiomyocytes for drug screening and pharmacological evaluation in vitro. J. Biomol. Screen. 17:1192-1203.
    Pubmed CrossRef
  42. Talman V and Kivelä R. 2018. Cardiomyocyte-endothelial cell interactions in cardiac remodeling and regeneration. Front. Cardiovasc. Med. 5:101.
    Pubmed KoreaMed CrossRef
  43. Tveito A, Lines GT, Edwards AG, Maleckar MM, Michailova A, McCulloch A. 2012. Slow Calcium-Depolarization-Calcium waves may initiate fast local depolarization waves in ventricular tissue. Prog. Biophys. Mol. Biol. 110:295-304.
    Pubmed KoreaMed CrossRef
  44. Varzideh F, Pahlavan S. 2019. Coculture with noncardiac cells promoted maturation of human stem cell-derived cardiomyocyte microtissues. J. Cell. Biochem. 120:16681-16691.
    Pubmed CrossRef
  45. Veerman CC, Mengarelli I, Lodder EM, Kosmidis G, Bellin M, Zhang M, Dittmann S, Guan K, Wilde AAM, Schulze-Bahr E, Greber B, Verkerk AO. 2017. Switch from fetal to adult SCN5A isoform in human induced pluripotent stem cell-derived cardiomyocytes unmasks the cellular phenotype of a conduction disease-causing mutation. J. Am. Heart Assoc. 6:e005135.
    Pubmed KoreaMed CrossRef
  46. Yang X, Murry CE. 2014a. Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ. Res. 114:511-523.
    Pubmed KoreaMed CrossRef
  47. Yang X, Rodriguez M, Pabon L, Fischer KA, Reinecke H, Regnier M, Sniadecki NJ, Murry CE. 2014b. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 72:296-304.
    Pubmed KoreaMed CrossRef
  48. Yoshida S, Miyagawa S, Fukushima S, Kawamura T, Kashiyama N, Ohashi F, Toyofuku T, Sawa Y. 2018. Maturation of human induced pluripotent stem cell-derived cardiomyocytes by soluble factors from human mesenchymal stem cells. Mol. Ther. 26:2681-2695.
    Pubmed KoreaMed CrossRef
  49. Youm JB. 2016. Electrophysiological properties and calcium handling of embryonic stem cell-derived cardiomyocytes. Integr. Med. Res. 5:3-10.
    Pubmed KoreaMed CrossRef
  50. Yu Y, Qin N, Lu XA, Li J, Han X, Ni X, Ye L, Shen Z, Chen W, Zhao ZA, Hu S. 2019. Human embryonic stem cell-derived cardiomyocyte therapy in mouse permanent ischemia and ischemia-reperfusion models. Stem Cell Res. Ther. 10:167.
    Pubmed KoreaMed CrossRef
  51. Zhang P, Mende U. 2012. Cross talk between cardiac myocytes and fibroblasts: from multiscale investigative approaches to mechanisms and functional consequences. Am. J. Physiol. Heart Circ. Physiol. 303:H1385-H1396.
    Pubmed KoreaMed CrossRef
  52. Zhao Z, Lan H, El-Battrawy I, Li X, Buljubasic F, Sattler K, Yücel G, Lang S, Tiburcy M, Zimmermann WH, Cyganek L, Utikal J, Wieland T, Borggrefe M, Akin I. 2018. Ion channel expression and characterization in human induced pluripotent stem cell-derived cardiomyocytes. Stem Cells Int. 2018:6067096.
    Pubmed KoreaMed CrossRef

Article

Review Article

Journal of Animal Reproduction and Biotechnology 2022; 37(2): 67-79

Published online June 30, 2022 https://doi.org/10.12750/JARB.37.2.67

Copyright © The Korean Society of Animal Reproduction and Biotechnology.

In vitro maturation of human pluripotent stem cell-derived cardiomyocyte: A promising approach for cell therapy

Yun-Gwi Park1,# , Yeo-Jin Son1,# , Sung-Hwan Moon1,2,* and Soon-Jung Park1,*

1Stem Cell Research Institute, T&R Biofab Co. Ltd., Seongnam 13487, Korea
2Department of Animal Science, Sangji University, Wonju 26339, Korea

Correspondence to:Sung-Hwan Moon
E-mail: moonsh22@sangji.ac.kr

Soon-Jung Park
E-mail: parksoonjung@gmail.com

#These authors contributed to this work as first authors.

Received: June 6, 2022; Revised: June 17, 2022; Accepted: June 20, 2022

Abstract

Currently, there is no treatment to reverse or cure heart failure caused by ischemic heart disease and myocardial infarction despite the remarkable advances in modern medicine. In addition, there is a lack of evidence regarding the existence of stem cells involved in the proliferation and regeneration of cardiomyocytes in adult hearts. As an alternative solution to overcome this problem, protocols for differentiating human pluripotent stem cell (hPSC) into cardiomyocyte have been established, which further led to the development of cell therapy in major leading countries in this field. Recently, clinical studies have confirmed the safety of hPSC-derived cardiac progenitor cells (CPCs). Although several institutions have shown progress in their research on cell therapy using hPSC-derived cardiomyocytes, the functions of cardiomyocytes used for transplantation remain to be those of immature cardiomyocytes, which poses a risk of graft-induced arrhythmias in the early stage of transplantation. Over the last decade, research aimed at achieving maturation of immature cardiomyocytes, showing same characteristics as those of mature cardiomyocytes, has been actively conducted using various approaches at leading research institutes worldwide. However, challenges remain in technological development for effective generation of mature cardiomyocytes with the same properties as those present in the adult hearts. Therefore, in this review, we provide an overview of the technological development status for maturation methods of hPSC-derived cardiomyocytes and present a direction for future development of maturation techniques.

Keywords: cardiomyocyte, cell therapy, human pluripotent stem cell, maturation, safety

INTRODUCTION

According to a report by the World Health Organization, cardiovascular diseases (CVDs) are the leading cause of death worldwide, with 8.9 million estimated deaths in 2019. In addition, it has been reported that those who had SARS-CoV-2 infection, which had a sweeping impact worldwide, had sequelae with long-term cardiovascular consequences (Abbasi, 2022). Since human cardiomyocytes cease to proliferate and the cardiac regenerative potential is lost shortly after birth (Hirose et al., 2019), treatments for any damage to the heart require medical interventions such as mechanical devices (e.g., a left ventricular assist device) or a cardiac transplant, allowing only limited available options. To overcome these limitations, the use of adult stem cells and pluripotent stem cells (PSCs) as cell therapy has garnered attention. In particular, differentiation of cardiomyocytes using PSCs capable of differentiating into various somatic cell types has drawn increased interest as alternative approach for cell therapy (Yu et al., 2019; de Lange et al., 2021). Consequently, various methods for the differentiation of cardiomyocytes have been developed and actively utilized to explore cell therapy, offering therapeutic strategies against cardiac repair or drug-induced cardiotoxicity (Burridge et al., 2015; Park et al., 2019; Choi et al., 2020).

Recently, a research team in France reported the completion of a phase 1 clinical trial involving human embryonic stem cells (hESCs)-derived CPCs to demonstrate the safety and effect of cardiac function improvement (Menasché et al., 2018). In Japan, based on the results of preclinical studies with heart failure (HF) animal models (Kashiyama et al., 2019; Kawaguchi et al., 2021), an ongoing clinical trial has been utilizing the transplantation of human induced pluripotent stem cells (hiPSCs)-derived cardiomyocyte sheets or spheroids. As can be seen from the above examples, research on the investigation of cell therapy using hPSC-derived cardiomyocytes for the treatment of heart diseases with limited alternative treatment options has developed into an area of global competition among world-class research groups. However, regarding the applications of hPSC-derived cardiomyocytes (CMs) in clinical settings, it is necessary to overcome the issue of graft-related arrhythmias. In several papers, as a result of transplanting hPSC-derived cardiomyocytes into heart disease model animals, arrhythmias (e.g., ventricular tachycardia) occurred in the early stage of transplantation. This arrhythmia showed time-dependent decreasing (Shiba et al., 2016; Romagnuolo et al., 2019). The induction of arrhythmia occurs due to the fact that the transplanted CMs in the early postoperative period are still immature compared to in vivo CMs. After transplantation, over the course of in vivo maturation of CMs, the incidence of arrhythmia gradually dissipated. Based on this finding, it is expected that graft-related arrhythmias occurring in the early postoperative period can be overcome if maturation of hPSC-derived CMs can be achieved prior to transplantation. Therefore, in this review, 1) an overview of the current progress of clinical studies using human PSC-derived CPCs or CMs is presented, 2) characteristics of immature CMs and mature CMs are comparatively analyzed, and 3) indicators for enhancement of maturation for key techniques of CM maturation are identified. Based on the findings of this review, we aim to present an effective strategy for the development of an optimal maturation technology for CMs.

An overview of clinical trials for cell therapy using hPSC-derived CPCs or CMs

Following the pioneering step of Mesoblast Ltd in 2005 involving a clinical trial using mesenchymal stem cells (MSCs), active research has been ongoing for the development of cell therapy for the treatment of HF and other heart conditions (Hare et al., 2009). However, after Mesoblast Ltd.’s reported failure in meeting the primary endpoint in phase 3 clinical trial, cell therapies using hPSC-derived CMs have drawn increasing interest for research and clinical applications. Unlike MSCs, CMs can directly participate in actual heart functions following engraftment at the transplant site, indicating a higher applicability and significance. Based on this perspective, the present review aims to investigate the current status of clinical trials using PSC-derived CMs (Table 1).

Table 1. Clinical trials with hPSC-derived CPCs or cardiomyocytes.

Institution (Nation)Assistance Publique (France)Osaka University (Japan)Heartseed (Japan)
Status (Term)Phase 1 completed (2013.05.27-2018.03.22)Phase 1 undergo (2019.12.02-2023.05.30)Phase 1 undergo (2022.04.19-2024.03.31)
Target diseaseIschemic Heart DiseaseMyocardial IschemiaHeart Failure
Patient10 patients for 5 years10 patients for 3 years10 patients for 5 years
Cell sourceESCAllogeneic iPSCAllogeneic iPSC
Cell typeCPC (SSEA1+ IsI-1+)CardiomyocyteCardiomyocyte
Transplantation20 cm2 Fibrin patch embedding
hESC-derived CPCs
hiPSC-derived cardiomyocyte sheetshiPSC-derived cardiomyocyte spheroids
Clinical mechanismImproves heart function by releasing factors that promote myocardial and/or vascular regeneration1) Helps heart function through self-beating
2) Improves heart function by releasing factors that promote myocardial and/or vascular regeneration
1) Helps heart function through self-beating
2) Improves heart function by releasing factors that promote myocardial and/or vascular regeneration
Outcome measures1 year follow-up after surgery:
1) Feasibility of patch’s generation and its efficacy on cardiac functions
2) Evidence for new clinical/biological abnormalities, occurrence of arrhythmias or development of a cardiac or extra-cardiac tumor
1 year follow-up after surgery:
1) Left ventricular (LV) ejection fraction, contractile and remodeling of the LV, New York Heart Association functional classification
2) Serious adverse events, abnormal blood biochemical or tumor marker tests and cardiac function clinical events
3) Minnesota Living with Heart Failure Questionnaire, 36-Item Short Form Survey etc.
1 year follow-up after surgery:
1) LV ejection fraction, myocardial wall motion evaluation, myocardial blood flow, myocardial viability
2) Safety and Tolerability
3) Kansas City Cardiomyopathy Questionnaire etc.
Result1. Cardiac function improvement effect
2. No arrhythmia, teratoma and immune-suppressant complications
OngoingOngoing
Pre/Clinical publicationMenasché et al., 2018Kashiyama et al., 2019Kawaguchi et al., 2021

1) A clinical study with hESC-derived CPCs

Regarding cell therapy using hESC-derived cardiovascular progenitors, a phase 1 trial conducted by a research team in France has been reported (Menasché et al., 2018). In the trial, from 2013 to 2018, ten patients with ischemic heart disease received hESC-derived CPCs by epicardial delivery of the cell-laden fibrin patch, and the clinical outcomes of the treatment were monitored. A 20 cm2 fibrin patch with a dose of 5 × 106 to 1 × 107 (Median: 8.2 × 106) hESC-derived CPCs was delivered to the epicardium of the infarct area. A year of follow-up monitoring revealed that one patient died because of treatment-unrelated comorbidities, but all others had uneventful recoveries. In all patients apart from the deceased patient, symptomatic improvement with an increased systolic motion of the cell-treated segments was confirmed, and there were no arrhythmias, complications of the immunosuppressive treatment, or intracardiac or off-target tumors. From these results, it can be interpreted that various factors released from the grafted CPCs promote myocardial and vascular regeneration, and such regeneration contributed to improving the function of infarcted hearts. However, considering that one patient died of heart failure 22 months post-transplantation, it is speculated that there is a limitation to achieving cardiac repair to the level of healthy adults by transplantation of CPCs.

2) A clinical study with hiPSC-derived cardiomyocyte sheets

Clinical trials on cell therapy using hiPSC-derived CMs have been conducted by two Japanese research teams. One of the research teams at Osaka university generated iPSC-CM sheets using cynomolgus macaque to induce cardiomyogenic differentiation in a preclinical study. The generated CMs were in the form of cell sheets (3.6 × 106 cells/sheet), and four pieces of the iPSC-cardiac sheets were transplanted in a cynomolgus macaque MI model. The effect of transplantation was evaluated through a 6-month follow-up monitoring. As a result, it was confirmed that cardiac function improved regardless of the matching/mismatching of the major histocompatibility complex (Kashiyama et al., 2019). Based on this preceding study, the research team is conducting a phase 1 trial to investigate the therapeutic effect of CM sheets derived from allogeneic iPSCs for 10 patients with myocardial ischemia for 3 years since 2019. Through 1-year follow-up post-transplantation, the treatment effect including functional and structural recoveries of the left ventricle, as well as adverse events including abnormal findings in the blood biochemical tests or cardiac function events and tumor development, will be examined for evaluation.

3) A clinical study with hiPSC-derived cardiomyocyte aggregates

Another ongoing clinical trial using hiPSC-derived CMs is being conducted by Tokyo-based Heartseed. First, a preclinical study was conducted using a rat model (i.e. a small animal model) and a micro-miniature pig model (i.e. a large animal model). Cardiac spheroids (200 μm) consisting of approximately 1,000 hiPSC-derived CMs were prepared, and 3,000 spheroids (total cell number: 3 × 106) and 1 × 105 spheroids (total cell number: 1 × 108) were transplanted in the rat heart failure model and the pig heart failure model, respectively, to evaluate the effect of transplantation. As a result, the cardiac spheroid transplantation group maintained the recovery of cardiac function for a longer period of time in the large animal model (Kawaguchi et al., 2021). Based on this preceding study, a phase 1 trial is underway to determine the effect of iPSC-derived cardiomyocyte spheroids on 10 heart failure (HF) patients for 5 years starting from 2022. Each patient will undergo transplantation of cardiac spheroids, and through 1-year post transplantation follow-up, adverse events in terms of safety, tolerability, and cardiac function recovery with outcome measures such as myocardial viability and myocardial blood flow will be evaluated.

Comparison of the characteristics between immature cardiomyocytes and mature cardiomyocytes

There is a general consensus among stem cell researchers on the immature state of hPSC-derived CMs, but methods for evaluating the degree of maturation remain to be determined. Therefore, in addition to developing methods for enhancing the maturation of hPSC-derived CMs, methods for evaluating the degree of maturation also need to be established. As a first step, in this review, we comparatively analyzed the characteristics of mature CMs in adult heart and immature (fetal and hPSC-derived) CMs (Table 2).

Table 2. Clinical trials with hPSC-derived CPCs or cardiomyocytes.

CharacteristicsCategoryCardiomyocytesReferences

Immature (Fetal or PSC-derived)Mature
MorphologyShapeCircular and oblongAnisotropic rod-likeLundy et al., 2013
SizeDiameters: 5-10 μm
Heights: ~5 μm
Length: 30 μm
Width: 10 μm
Volume: 2,000 μm3
Heights: 15 μm
Length: 150 μm
Width: 20 μm
Volume: 40,000 μm3
Bulatovic et al., 2016
Lundy et al., 2013
NucleusMononucleated diploidMultinucleated tetraploidDenning et al., 2016
Structure (Cytoskeleton)Sarcomere: disorganized, short
SR and T-tubule: poorly developed
Sarcomere: highly organized, long
SR and T-tubule: well-developed
Denning et al., 2016
Yang et al., 2014a
ElectrophysiologyBeatingSpontaneously beat
Force: 0.08-4 mN/mm2
mixed action potential (nodal, atrial ventricular-like)
Start: Sinoatrial node
No beat until triggered by the depolarization form adjacent cells
Force: 40-80 mN/mm2
Lin et al., 2017
Resting membrane potential-60 mV, nodal like-90 mVKoivumäki et al., 2018
Conduction velocity10-20 cm/sAround 60 cm/sDenning et al., 2016
Upstroke velocitySlow, 10-50 V/sFast, 150-350 V/sDenning et al., 2016
Veerman et al., 2017
RepolarizationFastSlow, after plateau phaseJeevaratnam et al., 2018
Zhao et al., 2018
Channel proteinINa, ICa-L, Ito
Low IK1,
INa, ICa-L, Ito, IKr, IKsGoversen et al., 2018
Calcium handling1) Similar expression of Na+/Ca2+ exchanger with adult CMs
2) Low Expression of other Ca2+ releasing related protein
3) Increasing of calcium stores in the SR
4) Slower calcium dynamics with delayed time to peak and slower decay of the calcium signal
Depolarization → opening of LTCC → calcium influx → calcium-induced calcium releasing → sliding of myofilament and contraction of muscle
Calcium releasing:
1) SERCA2a
2) Sarcolemmal Na+/Ca2+ exchanger
3) Sarcolemmal Ca2+-ATPase
4) Mitochondrial Ca2+ uniport
Hwang et al., 2015
Karakikes et al., 2015
Youm, 2016
Eisner et al., 2017
MetabolismMitochondriaLocation: perinuclear space
Low number and small size (<5% of total cell volume)
Round shape and low cristae density
In mouse, opened mitochondrial permeability transition (MPT) not induce cytochrome c leakage or apoptosis
Location: between myofibrils and under the sarcolemma
increase in both size and number
(~30% of total cell volume)
Oval shape and dense cristae → sufficient surface area
Feric and Radisic, 2016
Energy sourceEnergy was generated mainly by glycolysis(Low fatty acid/ High glucose or lactate)
Approximately 15% of total energy consumption in fetal cardiomyocytes is supplied with β-oxidation of fatty acids
Approximately 80% of total energy consumption in adult cardiomyocytes is supplied with β-oxidation of fatty acids
60-70% ATP for contractile function, followed SERCA and of other ion transporters
Correia et al., 2017
Galdos et al., 2017
Karakikes et al., 2015
Piquereau et al., 2018
Gene expressionSarcomeric gene (MHC)α-MHCβ-MHCKatrukha, 2013
Lundy et al., 2013
Troponin ITNNI1TNNI3Katrukha, 2013
OthersLow expression levels of SERCA2, Caveolin 3, KCNH2 etc.Lundy et al., 2013

1) Morphology and cellular structure

Mature cardiomyocytes (CMs) in adult hearts have rod-like shapes, and the size of a single CM is about 150 μm in length, 20 μm in width, and 15 μm in height, with a volume of 40,000 μm3 (Bulatovic et al., 2016). Regarding the morphology of the nucleus, mature CMs are multinucleated and have tetraploid nuclei. They are characterized by highly organized sarcomere and well-developed sarcoplasmic reticulum (SR) and transverse tubules (T-tubules) (Yang et al., 2014a). In contrast, immature hPSC-derived CMs have different shapes such as a circular or oblong geometry, and they are approximately 5-10 μm in diameter, 30 μm in length, 10 μm in width, and 5 μm in height, with a volume of 2,000 μm3, showing a smaller size compared to that of mature CMs (Lundy et al., 2013, Bulatovic et al., 2016). Regarding the nucleus morphology, the immature CMs are mononucleated or have diploid nuclei as in the case of other somatic cells. Regarding the structure of the cytoskeleton, the sarcomere is disorganized and shows irregular distribution in the cytoplasm. Moreover, following induction of CM differentiation, no or few T-tubules are observed in hPSC-derived CMs in the early stage of beating (Yang et al., 2014a; Denning et al., 2016; Park et al.,2022) (Fig. 1A).

Figure 1.Experimental methods for maturation of hPSC-derived cardiomyocytes. (A) Compared to matured cardiomyocytes, morphology and structure, calcium handling, metabolism, and electrophysiology are different in immature hPSC-CMs. (B) Various methods of maturation hPSC-CMs.
2) Electrophysiological properties

In general, adult CMs maintain a resting membrane potential of -90 mV until electrical impulses are transmitted from adjacent cells. When triggered by the adjacent cells, fast Na+ channels begin to open, and Na+ leaks into the cell. This produces rapid depolarization with the transmembrane potential rising to approximately +20 mV. The upstroke velocity at this time is measured to be approximately 150-350 V/s (Lin et al., 2017). After depolarization, an outflow of K+ occurs due to diffusion through the cell membrane, but with a constant inward current of Ca2+, the plateau phase is maintained for some time (Liu et al., 2016; Sharifi et al., 2017; Zhao et al., 2018). Next, Ca2+ channels are gradually inactivated, and with a persistent outflow of K+, the transmembrane potential is brought back toward a resting potential of -90mV, thereby completing a cycle. Subsequently, depolarization newly starts when triggered by the adjacent cells (Goversen et al., 2018; Koivumäki et al., 2018). In adult CMs, the action potential duration of one cycle is measured to be ~300 ms, and the conduction velocity, i.e. the velocity of transmission of electrical impulses to adjacent cells, is known to be approximately 50 cm/s (Tveito et al., 2012). In contrast, hPSC-derived CMs showed spontaneous beating, with a resting membrane potential of approximately -60 mV, similar to that of nodal CMs. In addition, when depolarization occurs, the subunits of Nav1.5 protein involved in the inflow of sodium ions are fetal isoforms, and the upstroke velocity is measured at approximately 10-50 V/s, slower than that of adult CMs (Veerman et al., 2017). Furthermore, since IKI, a type of channel protein in adult CMs that promotes K+ inflow and inhibits its outflow, is present at a low concentration in hPSC-derived CMs, K+ ion outflow occurs at a faster rate after depolarization than that in adult CMs. This causes rapid repolarization in which the transmembrane potential is brought back to the resting membrane potential after depolarization (Jeevaratnam et al., 2018).

3) Calcium handling

Calcium ions (Ca2+) play an important role in various signal transduction pathways, metabolism, and transcriptional regulation in cells, and in the case of myocytes, the intracellular Ca2+ concentration is also involved in the contraction and relaxation of the cardiac muscle. This indicates that Ca2+ also plays an important role in CMs, which is also a type of myocyte. In mature CMs, the concentration of Ca2+ is precisely controlled through various mechanisms. In the excitation-contraction coupling, when intracellular depolarization occurs, the L-type calcium channel (LTCC) opens, producing an inward current of Ca2+ from outside the cell membrane to the cytoplasm, and the increased cytoplasmic Ca2+ concentration induces the release of calcium-induced Ca2+ through ryanodine receptors (RYR) in the SR, thereby further increasing the cytoplasmic Ca2+ concentration. Ca2+ with increased cytoplasmic concentration binds to the troponin protein to induce muscle contraction. After the contraction is completed, cytoplasmic Ca2+ concentration is lowered again by removing Ca2+ from the cytoplasm through the following process: (1) Ca2+ is pumped back into SR via sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a); (2) Ca2+ is pumped out of the cell by the sodium-calcium exchanger; (3) Ca2+ uptake into mitochondria; (4) additional leak of Ca2+ by sarcolemmal calcium ATPase (Eisner et al., 2017). In the case of immature CMs lacking T-tubules, LTCC and RYR are separated in space, leading to a delay in the release of Ca2+ in the SR after depolarization, and also, the expression of Ca2+ handling protein is also lower than that in mature CMs, exhibiting a slower calcium dynamic overall. However, among the Ca2+ handling proteins, the expression of sodium-calcium exchanger is higher in immature fetal CMs than in mature CMs, and this characteristic can also be observed in the case of hPSC-derived CMs (Hwang et al., 2015).

4) Metabolism

The human heart requires a continuous supply of energy to maintain its ongoing pumping activity with systolic and diastolic movements from birth to death. Accordingly, the cardiac energy metabolism in CMs is highly effective. The typical features of fetal cardiac mitochondria include a low number and a small size, occupying <5% of the total cell volume. With the development of the fetal heart, both the number and size of the cardiac mitochondria show a robust increase, accounting for ~30% of the cell volume (Galdos et al., 2017). The differences can also be observed in terms of the morphology and location of the individual mitochondria with cardiomyocyte maturation. For mature CMs, mitochondria take a long, oval shape and are located between myofibrils and under the sarcolemma with the formation of dense cristae (Feric and Radisic, 2016). For immature CMs, round-shaped mitochondria are mainly located in the perinuclear space, with rudimentary cristae in the inner membrane of mitochondria. In terms of energy production through cellular metabolism with mitochondria having the central role, approximately 80% of the total energy in mature CMs is generated through β-oxidation metabolism of fatty acids (Karakikes et al., 2015; Piquereau et al., 2018). In the case of immature CMs, approximately only 15% of the energy is produced through β-oxidation of fatty acids, and in many cases, energy is generated through the metabolism of glucose or lactate (Correia et al., 2017). Since hPSC-derived CMs also exist in an immature state, their metabolism largely depends on glycolysis rather than β-oxidation of fatty acids.

5) Gene expression

Mature CMs and immature CMs show differences in terms of subunits of specific genes (proteins) in CMs. Myosin heavy chains (MHCs) are motor proteins that convert chemical energy derived from the hydrolysis of ATP into mechanical force that drives diverse motile processes in cardiac and skeletal muscle. MHCs are also expressed in two different, developmentally-regulated subunits: the fetal α-isoform (α-MHC) and the adult β-isoform (β-MHC). In mature CMs, β-MHC synthesized by the expression of the MYH7 gene is predominant, and in immature CMs, α-MHC synthesized by the expression of the MYH6 gene is mainly expressed (Katrukha, 2013; Lundy et al., 2013). Troponin I (TnI) behaves as a molecular switch of sarcomere within the myocytes and typically has three isoforms (TNNI1: Slow skeletal TnI, TNNI2: Fast skeletal TnI, TNNI3: Cardiac TnI). In mature CMs, cardiac TnI3 is predominantly expressed, whereas in immature CMs, slow skeletal TnI2 is predominantly expressed (Katrukha, 2013). In addition, the expression of genes such as SERCA2, Caveolin 3, and KCNH2 was lower in immature CMs (Lundy et al., 2013).

Methods of promoting maturation of cardiomyocytes

As described above, although there have been numerous studies on the generation and utilization of PSC-derived CMs, these cells show characteristics similar to those of immature fetal CMs. Accordingly, various approaches for obtaining mature cardiomyocytes by inducing in vitro maturation of PSC-derived CMs have been actively investigated. The representative maturation methods in the current research trend are outlined as shown below (Fig. 1B)

1) Long-term culture of cardiomyocytes

In the course of human heart development from fetal to adult CMs, the duration of maturation for fetal immature CMs from birth is over 10 years (Bulatovic et al., 2016). Accordingly, a number of studies have been conducted by different research teams to enhance the maturation of CMs through in-vitro long-term culture of hPSC-derived CMs (Shinozawa et al., 2012; Kamakura et al., 2013; Lundy et al. al., 2013; Cyganek et al., 2018). Through the long-term culture process lasting from 60 days to 1 year, the number of cells expressing the cTnT gene (cTnT+), a representative marker of CMs, and multinucleated cells increase, and CMs undergo morphological changes such as the increase in the sarcomere length and cell size with an increase in the expression of associated genes. Thus, it was confirmed that the maturity of hPSC-derived CMs was enhanced with time. In particular, formation of M-bands was confirmed in CMs following a long-term culture of 360 days or longer, but the amount of sarcomeric structures in a single CM was still scarce compared with that of adult CMs (Kamakura et al., 2013). The findings that hPSC-derived CMs did not reach the full level of maturation as that of adult CMs require additional investigation on other factors such as in vivo paracrine and hormone signals. In a recent study, during the initial process of CM differentiation, platelet-derived growth factor receptor-α (PDGFRα)-positive cells that can respond to CM differentiation cues and are thus known as one of the markers of CM differentiation were first collected, and subsequently, long-term culture of immature CMs was conducted. Through this method, the formation of a T-tubule-like structure, which was not confirmed in previous studies, was exhibited on day 231 of the long-term culture (Fukushima et al., 2020).

2) Addition of maturation factor

From the viewpoint of evolution, the closer the relation to mammals in the phylogenetic tree, the serum thyroxine level increases and the cardiac regenerative potential decreases, showing the inverse correlation between the serum thyroxine level and cardiac regenerative potential. Inactivation of thyroid hormone signaling reduced mouse cardiomyocyte polyploidization, and the mouse CMs remain in the immature state for longer (Hirose et al., 2019). This implies that regulation of thyroid hormones may serve to limit the CM regenerative capacity and induce CM polyploidization, thereby promoting the maturation of CMs. Accordingly, recent studies have investigated the method of adding Tri-iodo-L-thyronine (T3), a thyroid hormone, during the culture of CMs to induce in vitro maturation of CMs.

In a number of studies, the results showed that treatment with T3-induced gene expression of hPSC-derived CMs from the fetal-like level to the adult-like pattern increased the force per contractile along with the rate of calcium release and reuptake with SERCA2α to promote further maturation of CMs (Ivashchenko et al., 2013; Yang et al., 2014b; Birket et al., 2015). In addition, T3 treatment in combination with IGF-1, Dexamethasone increased the expression of PGC-1a and PGC-1b, which are important regulators of fatty acid oxidation (FAO) and mitochondrial function in CMs, and contributed to the formation of a uniform sarcomeric structure, thereby inducing the maturation of immature CMs.

3) 3-Dimensional (3D) cell culture methods

All cells exist in 3D space in vivo, but standard cell culture is performed in 2D, typically in a flat petri dish. This indicates that cells are cultured under conditions different from those of in vivo environments, which may lead to differences in cellular responses and functions compared to those observed in vivo. Therefore, research has been conducted on culturing hPSC-CMs in 3D systems and not in the conventional 2D systems. On day 6 of hPSC-CM differentiation, the cardiac progenitors were recovered, and the 3D aggregation of the hPSC-derived cardiac progenitors was induced. After a 29-day culture, the maturation of 3D-cultured CMs was compared with that of 2D-cultured CMs. In the case of CMs in aggregates, contractility was more than three times faster, and improved action potential kinetics were achieved. Moreover, in terms of gene expression with 3D culture, the expression of atrial-like CM markers decreased, and the expression of ventricular-like CM markers increased (Correia et al, 2018).

Among the 3D culture methods, improved maturation of CMs was reported for a culture of engineered heart tissues (EHTs). The hPSCs were differentiated into CMs, and after 30 days, hPSC-derived CMs were cultured in the form of engineered cardiac tissue constructs, a type of EHTs. Using the EHT culture method for 3 weeks without other methods to promote maturation apart from the 3D culture, T-tubule systems similar to the ventricular CMs of the adult heart were developed, and within a relatively short time of 2 weeks, an increase in expression of various genes related to calcium-handling and contraction was confirmed (de Lange et al, 2021). In terms of cell therapy, 3D culture has advantages over 2D culture. In both the results of a preclinical study (Kawaguchi et al., 2021) published by Heartseed, based on which a phase 1 trial is underway, and a recent report published by the authors’ research team, a single CM or CM aggregates were grafted on the infarcted area of an animal model with MI to evaluate the effect of transplantation. As a result, when CMs were transplanted in the form of 3D aggregates, the in vivo survival rate was higher than that of single CM transplantation, and the results also showed that the engraftment of mature CMs was induced, and the method was more effective in improving the symptoms of MI (Park et al., 2022).

4) Various other cell co-culture methods

As with all other organs in the human body, the heart does not consist of CMs alone but includes various other cells such as endothelial cells (Ecs), MSCs, and fibroblasts. The coexistence of different types of cells means that each cell can influence each other through intracellular interactions, and such interactions are also involved in the maturation of CMs (Zhang et al., 2012). Therefore, research has been conducted to investigate the effect of inducing CM maturation using a co-culture system in which hPSC-derived CMs are cultured with other types of cells. On comparing three different culturing methods of CMs: (1) hiPSC-CMs cultured alone, (2) hiPSC-CM co-cultured with MSC, and (3) addition of MSC-derived soluble factors, both the co-culture group and the soluble factor addition group showed higher structural and functional maturity of CMs (e.g. cell size, sarcomere length, contractility, electrophysiology) compared to the CMs cultured alone (Yoshida et al., 2018). The results confirmed that among CM culture methods, co-culture with MSCs promoted the maturation of CMs by releasing various bioactive factors favorable for cardiac cells. In addition to MSCs, endothelial cells (Ecs), whose number is approximately three times higher than that of CMs in the human heart, are also known to play a significant role in the early-stage heart development or cardiac repair (Talman et al., 2018). Based on this, during hPSC-CM differentiation, CMs were co-cultured with Ecs and after 2 weeks of additional culture, the maturity of cultured CMs was evaluated based on the cell size, sarcomere organization, and gene expression related to mature CMs (Dunn et al., 2019). The results confirmed that the maturity of CMs was higher when co-cultured with CPCs in the early or mid-stage of CM differentiation than in the case of CPC monoculture or co-culture with EC of CMs after the completion of differentiation. The results indicate that Ecs induce maturation of CMs by delivering microRNA through paracrine factors and gap junctions in immature CMs. In addition, when microtissues were prepared using 3D co-culture with CPC, EC, and MSC in a ratio of 2:1:1, respectively, and cultured for 1 week, CMs maturation was accelerated compared to that of microtissues with CPC alone. (Varzideh et al, 2019). For transplantation of hiPSC-derived CMs to an animal MI model, 3D printing was used to prepare hMSC-loaded patch, and the patch was epicardially implanted. As a result, the hMSC-secreted factors improved the survival of hiPSC-CMs compared to the transplantation of CMs alone, and the method was also effective in enhancing vascular regeneration and promoting the maturation of CMs (Park et al., 2019).

5) Electric stimulation of cardiomyocytes

In this maturation method, cardiac cells were isolated from rats to investigate whether the morphology and functionalities of CMs changed (developed) similar to those of in vivo CMs of rats with or without electric stimulation under in vitro culture environment (Lasher et al., 2012). To compare differences in the maturation of CMs with/without electric stimulation, CMs with electric stimulation of 2 ms symmetric biphasic square pulses, 4 V/cm, and 1 Hz for 9 days using a carbon-rod of scm-spacing were compared with CMs without electric stimulation on days 3, 6, and 9. In terms of morphology, CMs applied with electric stimulation showed more similar values of length, width, and height with the age-matched myocardium compared to those of CMs without electric stimulation. In terms of functionalities, the decreased excitation threshold and increased maximum capture rate were observed in CMs with electric stimulation, which are the indicators of maturation. In a study where electric stimulation of 5 ms pulse, 5 V/cm, and 2 Hz was applied to iPSC-derived CMs to induce maturation of CMs, the result confirmed that maturation of CMs can be induced by electric stimulation in terms of cell alignment, contractility and passive stiffness, and the increase in expression of calcium-handling protein (Ruan et al., 2016).

CONCLUSION REMARKS AND FUTURE PERSPECTIVES

With CVDs reported as the leading cause of death worldwide, development of cell therapy that enables the fundamental treatment of CVDs has gained increasing attention. In particular, there is an ever-increasing importance of cell therapy based on PSC-derived CMs because the method can be applied for various diseases in the heart, a representative organ of the cardiovascular system, and the method allows unlimited (mass) generation of CMs with extremely low regenerative capacity. Therefore, in this review, the current status of clinical trials using PSC-derived CPCs and CMs as cell therapy was outlined, and the different characteristics of mature adult CMs and immature CMs were comparatively analyzed. Furthermore, since PSC-derived CMs are in an immature state, various culture methods that promote maturation of CMs were discussed. A number of studies have shown that immature PSC-derived CMs can mature into having properties similar to those of adult CMs by implementing various maturation methods. Therefore, the authors’ research team aims to develop a more effective method of promoting the maturation of CMs by analysis and incorporating the results of the recent publications and applying an optimal combination of these different methods. Furthermore, with additional implementation of a co-culture system including MSC and EC using 3D printing techniques, the establishment of a future direction for CM engineering by forming surface modification using bioinks, and creation of a 3D-printed cardiac model, it is expected that a model with the same morphology and functionality as in vivo cardiomyocytes can be developed with more efficient intercellular signal transduction and synchronization of the maturation level of individual CMs (Fig. 2). Although challenges still remain to be overcome, continuous efforts on the technological development of maturation methods are expected to present a new opportunity for cardiovascular research, and considering the track record of the emergence of new technologies and analytical tools up until the techniques developed to date, the new technologies and analytical tools are expected to create synergistic effects. If the technology for maturation of hPSC-derived CMs into cells with the same functional and structural characteristics as those of mature CMs in vivo can be developed through various approaches as described above, it is expected that the effects of CM-based cell therapy can be clearly established along with extensive utilization of the developed maturation technology in areas of basic research, including the analysis of cardiac functions, and for various clinical applications, including cardiotoxicity evaluation of drug candidates.

Figure 2.The new strategy to mature hPSC-derived cardiomyocytes into the identical characteristic cardiomyocytes in adult heart through a combination of cardiomyocyte maturation methods.

Acknowledgements

We thanks to Prof. Seong-Woo Choi of Dongguk University College of Medicine, Na-Kyeong Park of Seoul National University College of Medicine, Dr. Seok-Yun Jung and Mi-Jeong Kim of Stem Cell Research Institute, T&R Biofab Co. Ltd, Republic of Korea for their insightful comments on this manuscript.

Author Contributions

Conceptualization, S-J.P. and S-H.M.; writing and editing, Y-J.S, and Y-G.P.

Funding

This research was supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI20C0184), and the Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korea government (the Ministry of Science and ICT, the Ministry of Health & Welfare) (21A0403L1).

Ethical Approval

Not applicable.

Consent to Participate

Not applicable.

Consent to Publish

Yes.

Availability of Data and Materials

Not applicable.

Conflicts of Interest

No potential conflict of interest relevant to this article was reported.

Fig 1.

Figure 1.Experimental methods for maturation of hPSC-derived cardiomyocytes. (A) Compared to matured cardiomyocytes, morphology and structure, calcium handling, metabolism, and electrophysiology are different in immature hPSC-CMs. (B) Various methods of maturation hPSC-CMs.
Journal of Animal Reproduction and Biotechnology 2022; 37: 67-79https://doi.org/10.12750/JARB.37.2.67

Fig 2.

Figure 2.The new strategy to mature hPSC-derived cardiomyocytes into the identical characteristic cardiomyocytes in adult heart through a combination of cardiomyocyte maturation methods.
Journal of Animal Reproduction and Biotechnology 2022; 37: 67-79https://doi.org/10.12750/JARB.37.2.67

Table 1 . Clinical trials with hPSC-derived CPCs or cardiomyocytes.

Institution (Nation)Assistance Publique (France)Osaka University (Japan)Heartseed (Japan)
Status (Term)Phase 1 completed (2013.05.27-2018.03.22)Phase 1 undergo (2019.12.02-2023.05.30)Phase 1 undergo (2022.04.19-2024.03.31)
Target diseaseIschemic Heart DiseaseMyocardial IschemiaHeart Failure
Patient10 patients for 5 years10 patients for 3 years10 patients for 5 years
Cell sourceESCAllogeneic iPSCAllogeneic iPSC
Cell typeCPC (SSEA1+ IsI-1+)CardiomyocyteCardiomyocyte
Transplantation20 cm2 Fibrin patch embedding
hESC-derived CPCs
hiPSC-derived cardiomyocyte sheetshiPSC-derived cardiomyocyte spheroids
Clinical mechanismImproves heart function by releasing factors that promote myocardial and/or vascular regeneration1) Helps heart function through self-beating
2) Improves heart function by releasing factors that promote myocardial and/or vascular regeneration
1) Helps heart function through self-beating
2) Improves heart function by releasing factors that promote myocardial and/or vascular regeneration
Outcome measures1 year follow-up after surgery:
1) Feasibility of patch’s generation and its efficacy on cardiac functions
2) Evidence for new clinical/biological abnormalities, occurrence of arrhythmias or development of a cardiac or extra-cardiac tumor
1 year follow-up after surgery:
1) Left ventricular (LV) ejection fraction, contractile and remodeling of the LV, New York Heart Association functional classification
2) Serious adverse events, abnormal blood biochemical or tumor marker tests and cardiac function clinical events
3) Minnesota Living with Heart Failure Questionnaire, 36-Item Short Form Survey etc.
1 year follow-up after surgery:
1) LV ejection fraction, myocardial wall motion evaluation, myocardial blood flow, myocardial viability
2) Safety and Tolerability
3) Kansas City Cardiomyopathy Questionnaire etc.
Result1. Cardiac function improvement effect
2. No arrhythmia, teratoma and immune-suppressant complications
OngoingOngoing
Pre/Clinical publicationMenasché et al., 2018Kashiyama et al., 2019Kawaguchi et al., 2021

Table 2 . Clinical trials with hPSC-derived CPCs or cardiomyocytes.

CharacteristicsCategoryCardiomyocytesReferences

Immature (Fetal or PSC-derived)Mature
MorphologyShapeCircular and oblongAnisotropic rod-likeLundy et al., 2013
SizeDiameters: 5-10 μm
Heights: ~5 μm
Length: 30 μm
Width: 10 μm
Volume: 2,000 μm3
Heights: 15 μm
Length: 150 μm
Width: 20 μm
Volume: 40,000 μm3
Bulatovic et al., 2016
Lundy et al., 2013
NucleusMononucleated diploidMultinucleated tetraploidDenning et al., 2016
Structure (Cytoskeleton)Sarcomere: disorganized, short
SR and T-tubule: poorly developed
Sarcomere: highly organized, long
SR and T-tubule: well-developed
Denning et al., 2016
Yang et al., 2014a
ElectrophysiologyBeatingSpontaneously beat
Force: 0.08-4 mN/mm2
mixed action potential (nodal, atrial ventricular-like)
Start: Sinoatrial node
No beat until triggered by the depolarization form adjacent cells
Force: 40-80 mN/mm2
Lin et al., 2017
Resting membrane potential-60 mV, nodal like-90 mVKoivumäki et al., 2018
Conduction velocity10-20 cm/sAround 60 cm/sDenning et al., 2016
Upstroke velocitySlow, 10-50 V/sFast, 150-350 V/sDenning et al., 2016
Veerman et al., 2017
RepolarizationFastSlow, after plateau phaseJeevaratnam et al., 2018
Zhao et al., 2018
Channel proteinINa, ICa-L, Ito
Low IK1,
INa, ICa-L, Ito, IKr, IKsGoversen et al., 2018
Calcium handling1) Similar expression of Na+/Ca2+ exchanger with adult CMs
2) Low Expression of other Ca2+ releasing related protein
3) Increasing of calcium stores in the SR
4) Slower calcium dynamics with delayed time to peak and slower decay of the calcium signal
Depolarization → opening of LTCC → calcium influx → calcium-induced calcium releasing → sliding of myofilament and contraction of muscle
Calcium releasing:
1) SERCA2a
2) Sarcolemmal Na+/Ca2+ exchanger
3) Sarcolemmal Ca2+-ATPase
4) Mitochondrial Ca2+ uniport
Hwang et al., 2015
Karakikes et al., 2015
Youm, 2016
Eisner et al., 2017
MetabolismMitochondriaLocation: perinuclear space
Low number and small size (<5% of total cell volume)
Round shape and low cristae density
In mouse, opened mitochondrial permeability transition (MPT) not induce cytochrome c leakage or apoptosis
Location: between myofibrils and under the sarcolemma
increase in both size and number
(~30% of total cell volume)
Oval shape and dense cristae → sufficient surface area
Feric and Radisic, 2016
Energy sourceEnergy was generated mainly by glycolysis(Low fatty acid/ High glucose or lactate)
Approximately 15% of total energy consumption in fetal cardiomyocytes is supplied with β-oxidation of fatty acids
Approximately 80% of total energy consumption in adult cardiomyocytes is supplied with β-oxidation of fatty acids
60-70% ATP for contractile function, followed SERCA and of other ion transporters
Correia et al., 2017
Galdos et al., 2017
Karakikes et al., 2015
Piquereau et al., 2018
Gene expressionSarcomeric gene (MHC)α-MHCβ-MHCKatrukha, 2013
Lundy et al., 2013
Troponin ITNNI1TNNI3Katrukha, 2013
OthersLow expression levels of SERCA2, Caveolin 3, KCNH2 etc.Lundy et al., 2013

References

  1. Abbasi J. 2022. The COVID heart-one year after SARS-CoV-2 infection, patients have an array of increased cardiovascular risks. JAMA 327:1113-1114.
    Pubmed CrossRef
  2. Bedada FB, Chan SS, Metzger SK, Zhang L, Zhang J, Garry DJ, Kamp TJ, Metzger JM. 2014. Acquisition of a quantitative, stoichiometrically conserved ratiometric marker of maturation status in stem cell-derived cardiac myocytes. Stem Cell Reports 3:594-605.
    Pubmed KoreaMed CrossRef
  3. Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR, van de Pol V, Dambrot C, Devalla HD, Davis RP, Mastroberardino PG, Atsma DE, Mummery CL. 2015. Contractile defect caused by mutation in MYBPC3 revealed under conditions optimized for human PSC-cardiomyocyte function. Cell Rep. 13:733-745.
    Pubmed KoreaMed CrossRef
  4. Bulatovic I, Månsson-Broberg A, Grinnemo KH. 2016. Human fetal cardiac progenitors: the role of stem cells and progenitors in the fetal and adult heart. Best Pract. Res. Clin. Obstet. Gynaecol. 31:58-68.
    Pubmed CrossRef
  5. Burridge PW, Wu JC. 2015. Chemically defined culture and cardiomyocyte differentiation of human pluripotent stem cells. Curr. Protoc. Hum. Genet. 87:21.3.1-21.3. 15.
    Pubmed KoreaMed CrossRef
  6. Choi SW, Shin JS, Park SJ, Jung E, Park YG, Lee J, Kim SJ, Park HJ, Lee JH, Park SM, Moon SH, Go YY. 2020. Antiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytes. Antiviral Res. 184:104955.
    Pubmed KoreaMed CrossRef
  7. Correia C, Koshkin A, Duarte P, Hu D, Carido M, Sebastião MJ, Gomes-Alves P, Elliott DA, Domian IJ, Teixeira AP, Serra M. 2018. 3D aggregate culture improves metabolic maturation of human pluripotent stem cell derived cardiomyocytes. Biotechnol. Bioeng. 115:630-644.
    Pubmed CrossRef
  8. Correia C, Koshkin A, Duarte P, Hu D, Teixeira A, Domian I, Alves PM. 2017. Distinct carbon sources affect structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Sci. Rep. 7:8590.
    Pubmed KoreaMed CrossRef
  9. Cyganek L, Tiburcy M, Sekeres K, Gerstenberg K, Bohnenberger H, Lenz C, Henze S, Stauske M, Salinas G, Zimmermann WH, Guan K. 2018. Deep phenotyping of human induced pluripotent stem cell-derived atrial and ventricular cardiomyocytes. JCI Insight 3:e99941.
    Pubmed KoreaMed CrossRef
  10. de Lange WJ, Farrell ET, Kreitzer CR, Jacobs DR, Lang D, Ralphe JC. 2021. Human iPSC-engineered cardiac tissue platform faithfully models important cardiac physiology. Am. J. Physiol. Heart Circ. Physiol. 320:H1670-H1686.
    Pubmed KoreaMed CrossRef
  11. Denning C, Borgdorff V, Crutchley J, Firth KS, George V, Kalra S, Kondrashov A, Hoang MD, Mosqueira D, Patel A, Prodanov L, Rajamohan D, Skarnes WC, Young LE. 2016. Cardiomyocytes from human pluripotent stem cells: from laboratory curiosity to industrial biomedical platform. Biochim. Biophys Acta 1863(7 Pt B):1728-1748.
    Pubmed KoreaMed CrossRef
  12. Dunn KK, Reichardt IM, Simmons AD, Jin G, Floy ME, Palecek SP. 2019. Coculture of endothelial cells with human pluripotent stem cell-derived cardiac progenitors reveals a differentiation stage-specific enhancement of cardiomyocyte maturation. Biotechnol. J. 14:e1800725.
    Pubmed KoreaMed CrossRef
  13. Eisner DA, Caldwell JL, Trafford AW. 2017. Calcium and excitation-contraction coupling in the heart. Circ. Res. 121:181-195.
    Pubmed KoreaMed CrossRef
  14. Feric NT and Radisic M. 2016. Maturing human pluripotent stem cell-derived cardiomyocytes in human engineered cardiac tissues. Adv. Drug Deliv. Rev. 96:110-134.
    Pubmed KoreaMed CrossRef
  15. Fukushima H, Yoshioka M, Kawatou M, López-Dávila V, Takeda M, Kanda Y, Sekino Y, Yamashita JK. 2020. Specific induction and long-term maintenance of high purity ventricular cardiomyocytes from human induced pluripotent stem cells. PLoS One 15:e0241287.
    Pubmed KoreaMed CrossRef
  16. Galdos FX, Guo Y, Paige SL, VanDusen NJ, Pu WT. 2017. Cardiac regeneration: lessons from development. Circ. Res. 120:941-959.
    Pubmed KoreaMed CrossRef
  17. Goversen B, van der Heyden MAG, de Boer TP. 2018. The immature electrophysiological phenotype of iPSC-CMs still hampers in vitro drug screening: special focus on IK1. Pharmacol. Ther. 183:127-136.
    Pubmed CrossRef
  18. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB Jr, Reisman MA, Sherman W. 2009. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J. Am. Coll. Cardiol. 54:2277-2286.
    Pubmed KoreaMed CrossRef
  19. Hirose K, Payumo AY, Cutie S, Hoang A, Zhang H, Guyot R, Lunn D, Bigley RB, Yu H, Wang J, Smith M, Gillett E, Muroy SE, Schmid T, Wilson E, Field KA, Reeder DM, Maden M, Yartsev MM, Wolfgang MJ, Grützner F, Scanlan TS, Szweda LI, Buffenstein R, Hu G, Flamant F, Huang GN. 2019. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 364:184-188.
    Pubmed KoreaMed CrossRef
  20. Hwang HS, Kryshtal DO, Feaster TK, Sánchez-Freire V, Zhang J, Kamp TJ, Hong CC, Knollmann BC. 2015. Comparable calcium handling of human iPSC-derived cardiomyocytes generated by multiple laboratories. J. Mol. Cell. Cardiol. 85:79-88.
    Pubmed KoreaMed CrossRef
  21. Ivashchenko CY, Pipes GC, Lozinskaya IM, Lin Z, Xiaoping X, Needle S, Grygielko ET, Hu E, Toomey JR, Willette RN. 2013. Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype. Am. J. Physiol. Heart Circ. Physiol. 305:H913-H922.
    Pubmed CrossRef
  22. Jeevaratnam K, Chadda KR, Camm AJ. 2018. Cardiac potassium channels: physiological insights for targeted therapy. J. Cardiovasc. Pharmacol. Ther. 23:119-129.
    Pubmed KoreaMed CrossRef
  23. Kamakura T, Makiyama T, Sasaki K, Yoshida Y, Wuriyanghai Y, Chen J, Hattori T, Ohno S, Kita T, Horie M, Kimura T. 2013. Ultrastructural maturation of human-induced pluripotent stem cell-derived cardiomyocytes in a long-term culture. Circ. J. 77:1307-1314.
    Pubmed CrossRef
  24. Karakikes I, Ameen M, Wu JC. 2015. Human induced pluripotent stem cell-derived cardiomyocytes: insights into molecular, cellular, and functional phenotypes. Circ. Res. 117:80-88.
    Pubmed KoreaMed CrossRef
  25. Kashiyama N, Miyagawa S, Fukushima S, Kawamura T, Kawamura A, Yoshida S, Eiraku S, Harada A, Matsunaga K, Watabe T, Toda K, Sawa Y. 2019. MHC-mismatched allotransplantation of induced pluripotent stem cell-derived cardiomyocyte sheets to improve cardiac function in a primate ischemic cardiomyopathy model. Transplantation 103:1582-1590.
    Pubmed CrossRef
  26. Katrukha IA. 2013. Human cardiac troponin complex. Structure and functions. Biochemistry (Mosc.) 78:1447-1465.
    Pubmed CrossRef
  27. Kawaguchi S, Soma Y, Nakajima K, Kanazawa H, Tohyama S, Tabei R, Hirano A, Handa N, Yamada Y, Okuda S, Hishikawa S, Teratani T, Kunita S, Kishino Y, Okada M, Tanosaki S, Someya S, Morita Y, Tani H, Kawai Y, Yamazaki M, Ito A, Shibata R, Murohara T, Tabata Y, Kobayashi E, Shimizu H, Fujita J. 2021. Intramyocardial transplantation of human iPS cell-derived cardiac spheroids improves cardiac function in heart failure animals. JACC Basic Transl. Sci. 6:239-254.
    Pubmed KoreaMed CrossRef
  28. Koivumäki JT, Naumenko N, Tuomainen T, Takalo J, Oksanen M, Puttonen KA, Lehtonen Š, Kuusisto J, Laakso M, Tavi P. 2018. Structural immaturity of human iPSC-derived cardiomyocytes: in silico investigation of effects on function and disease modeling. Front. Physiol. 9:80.
    Pubmed KoreaMed CrossRef
  29. Lasher RA, Pahnke AQ, Johnson JM, Hitchcock RW. 2012. Electrical stimulation directs engineered cardiac tissue to an age-matched native phenotype. J. Tissue Eng. 3:2041731412455354.
    Pubmed KoreaMed CrossRef
  30. Lin ZC, McGuire AF, Burridge PW, Matsa E, Lou HY, Cui B. 2017. Accurate nanoelectrode recording of human pluripotent stem cell-derived cardiomyocytes for assaying drugs and modeling disease. Microsyst. Nanoeng. 3:16080.
    Pubmed KoreaMed CrossRef
  31. Liu J, Backx PH. 2016. The electrophysiological development of cardiomyocytes. Adv. Drug Deliv. Rev. 96:253-273.
    Pubmed CrossRef
  32. Lundy SD, Zhu WZ, Laflamme MA. 2013. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 22:1991-2002.
    Pubmed KoreaMed CrossRef
  33. Menasché P, Vanneaux V, Hagège A, Bel A, Cholley B, Parouchev A, Cacciapuoti I, Al-Daccak R, Benhamouda N, Blons H, Agbulut O, Tosca L, Trouvin JH, Fabreguettes JR, Bellamy V, Charron D, Tartour E, Tachdjian G, Larghero J. 2018. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J. Am. Coll. Cardiol. 71:429-438.
    Pubmed CrossRef
  34. Park SJ, Kim H, Lee S, Kim J, Jung TH, Choi SW, Park BW, Kang SW, Elliott DA, Stanley EG, Elefanty AG, Ban K, Moon SH. 2022. Effect and application of cryopreserved three-dimensional microcardiac spheroids in myocardial infarction therapy. Clin. Transl. Med. 12:e721.
    Pubmed KoreaMed CrossRef
  35. Park SJ, Kim RY, Park BW, Lee S, Choi SW, Park JH, Choi JJ, Kim SW, Jang J, Cho DW, Chung HM, Moon SH, Park HJ. 2019. Dual stem cell therapy synergistically improves cardiac function and vascular regeneration following myocardial infarction. Nat. Commun. 10:3123.
    Pubmed KoreaMed CrossRef
  36. Piquereau J and Ventura-Clapier R. 2018. Maturation of cardiac energy metabolism during perinatal development. Front. Physiol. 9:959.
    Pubmed KoreaMed CrossRef
  37. Romagnuolo R, Masoudpour H, Porta-Sánchez A, Qiang B, Barry J, Laskary A, Qi X, Massé S, Magtibay K, Kawajiri H, Wu J, Valdman Sadikov T, Rothberg J, Panchalingam KM, Titus E, Li RK, Zandstra PW, Wright GA, Nanthakumar K, Ghugre NR, Laflamme MA. 2019. Human embryonic stem cell-derived cardiomyocytes regenerate the infarcted pig heart but induce ventricular tachyarrhythmias. Stem Cell Reports 12:967-981.
    Pubmed KoreaMed CrossRef
  38. Ruan JL, Tulloch NL, Razumova MV, Saiget M, Muskheli V, Pabon L, Reinecke H, Murry CE. 2016. Mechanical stress conditioning and electrical stimulation promote contractility and force maturation of induced pluripotent stem cell-derived human cardiac tissue. Circulation 134:1557-1567.
    Pubmed KoreaMed CrossRef
  39. Sharifi M, Buzatu D, Wilkes J. 2017. Development of models for predicting Torsade de Pointes cardiac arrhythmias using perceptron neural networks. BMC Bioinformatics 18(Suppl 14:497.
    Pubmed KoreaMed CrossRef
  40. Shiba Y, Gomibuchi T, Seto T, Wada Y, Ichimura H, Tanaka Y, Ogasawara T, Okada K, Shiba N, Sakamoto K, Ido D, Shiina T, Ohkura M, Nakai J, Uno N, Kazuki Y, Oshimura M, Ikeda U. 2016. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 538:388-391.
    Pubmed CrossRef
  41. Shinozawa T, Imahashi K, Sawada H, Takami K. 2012. Determination of appropriate stage of human-induced pluripotent stem cell-derived cardiomyocytes for drug screening and pharmacological evaluation in vitro. J. Biomol. Screen. 17:1192-1203.
    Pubmed CrossRef
  42. Talman V and Kivelä R. 2018. Cardiomyocyte-endothelial cell interactions in cardiac remodeling and regeneration. Front. Cardiovasc. Med. 5:101.
    Pubmed KoreaMed CrossRef
  43. Tveito A, Lines GT, Edwards AG, Maleckar MM, Michailova A, McCulloch A. 2012. Slow Calcium-Depolarization-Calcium waves may initiate fast local depolarization waves in ventricular tissue. Prog. Biophys. Mol. Biol. 110:295-304.
    Pubmed KoreaMed CrossRef
  44. Varzideh F, Pahlavan S. 2019. Coculture with noncardiac cells promoted maturation of human stem cell-derived cardiomyocyte microtissues. J. Cell. Biochem. 120:16681-16691.
    Pubmed CrossRef
  45. Veerman CC, Mengarelli I, Lodder EM, Kosmidis G, Bellin M, Zhang M, Dittmann S, Guan K, Wilde AAM, Schulze-Bahr E, Greber B, Verkerk AO. 2017. Switch from fetal to adult SCN5A isoform in human induced pluripotent stem cell-derived cardiomyocytes unmasks the cellular phenotype of a conduction disease-causing mutation. J. Am. Heart Assoc. 6:e005135.
    Pubmed KoreaMed CrossRef
  46. Yang X, Murry CE. 2014a. Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ. Res. 114:511-523.
    Pubmed KoreaMed CrossRef
  47. Yang X, Rodriguez M, Pabon L, Fischer KA, Reinecke H, Regnier M, Sniadecki NJ, Murry CE. 2014b. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 72:296-304.
    Pubmed KoreaMed CrossRef
  48. Yoshida S, Miyagawa S, Fukushima S, Kawamura T, Kashiyama N, Ohashi F, Toyofuku T, Sawa Y. 2018. Maturation of human induced pluripotent stem cell-derived cardiomyocytes by soluble factors from human mesenchymal stem cells. Mol. Ther. 26:2681-2695.
    Pubmed KoreaMed CrossRef
  49. Youm JB. 2016. Electrophysiological properties and calcium handling of embryonic stem cell-derived cardiomyocytes. Integr. Med. Res. 5:3-10.
    Pubmed KoreaMed CrossRef
  50. Yu Y, Qin N, Lu XA, Li J, Han X, Ni X, Ye L, Shen Z, Chen W, Zhao ZA, Hu S. 2019. Human embryonic stem cell-derived cardiomyocyte therapy in mouse permanent ischemia and ischemia-reperfusion models. Stem Cell Res. Ther. 10:167.
    Pubmed KoreaMed CrossRef
  51. Zhang P, Mende U. 2012. Cross talk between cardiac myocytes and fibroblasts: from multiscale investigative approaches to mechanisms and functional consequences. Am. J. Physiol. Heart Circ. Physiol. 303:H1385-H1396.
    Pubmed KoreaMed CrossRef
  52. Zhao Z, Lan H, El-Battrawy I, Li X, Buljubasic F, Sattler K, Yücel G, Lang S, Tiburcy M, Zimmermann WH, Cyganek L, Utikal J, Wieland T, Borggrefe M, Akin I. 2018. Ion channel expression and characterization in human induced pluripotent stem cell-derived cardiomyocytes. Stem Cells Int. 2018:6067096.
    Pubmed KoreaMed CrossRef

JARB Journal of Animal Reproduction and Biotehnology

qr code

OPEN ACCESS pISSN: 2671-4639
eISSN: 2671-4663