BMB Reports 2023; 56(1): 32-42  https://doi.org/10.5483/BMBRep.2022-0185
Engineered human cardiac tissues for modeling heart diseases
Sungjin Min1 & Seung-Woo Cho1,2,*
1Department of Biotechnology, Yonsei University, Seoul 03722, 2Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
Correspondence to: Tel: +82-2-2123-5662; Fax: +82-2-362-7265; E-mail: seungwoocho@yonsei.ac.kr
Received: October 9, 2022; Revised: November 28, 2022; Accepted: November 28, 2022; Published online: November 28, 2022.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-on-a-chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed.
Keywords: Cardiac tissue models, Disease modeling, Organoids, Stem cells, Tissue engineering
INTRODUCTION

The heart is a vital organ that maintains every other organ in the human body through the pumping and circulation of blood. Therefore, heart problems pose a great threat to human morbidity and mortality. In this context, cardiovascular disease, including ischemic heart disease, cardiomyopathy, and hypertensive heart disease, is one of the leading causes of death for mankind, and the global death toll and consequential burdens have continued to increase (1). To overcome these diseases, it is necessary to identify the pathological factors of disease progression and their mechanism, and to further develop effective treatments suitable for each disease. For this purpose, diverse three-dimensional (3D) human cardiac tissue models that mimic the human heart have been actively developed. These tissue models are free from the ethical issues which usually surround conventional animal models and can also eliminate gaps present between different species. Depending on which cardiac cells and engineering platforms are used, cardiac tissue models showing various shapes and characteristics can be developed (Fig. 1). Moreover, some biophysical cues such as mechanical and electrical stimulation have been applied to cardiac tissue models to further generate functionally matured ones.

The developed cardiac tissue models can be utilized for the modeling of various heart diseases. By using patient-derived induced pluripotent stem cells (iPSCs), cardiac tissue models expressing disease-associated genes can be developed (2). This model helps us to understand the differences in characteristics and phenotypes caused by a particular gene. Recently, such models can also be produced by gene editing methods, such as the CRISPR-Cas9 system, which manipulate specific target genes, and the role of genes can be investigated through comparison with normal models (3). Finally, heart disease models can be established by induction of the causative pathway of disease or by exposure to unfavorable environments (e.g., hypoxia, inflammation). In this review, we describe the different approaches to generating human cardiac tissue models using cardiac cells and engineering platforms, and then introduce several heart disease models produced with engineered cardiac tissue models.

CELL SOURCES FOR CARDIAC TISSUE MODELS

Cardiomyocytes for cardiac tissue models

Cardiomyocytes (CMs) are the basic beating units of the heart, and there are several types such as pacemaker cells, ventricular CMs, and atrial CMs. For decades, fully differentiated primary CMs extracted from the heart have been used as in vitro models. However, there are limitations as they are difficult to use due to their low viability and loss of maturity during culture (4). As an alternative, obtaining CMs using cellular reprogramming, which changes the fate of cells, is currently in the spotlight.

There are two methods for acquiring CMs, the differentiation method from human iPSCs and the direct reprogramming technique. The differentiation method from iPSCs first generates mesoderm cells, and then CMs are generated through cardiac progenitor cells. This process mimics normal embryonic development, for which several signaling pathways, such as Wnt/β-catenin and Activin A/BMP4 signaling, are regulated at each stage (5). With this method, CMs can be obtained without burden to the patient and an unlimited supply may be possible if iPSCs are cultured adequately. However, the differentiation method still has limitations such as yield, purity, variability, reproducibility, and cost-effectiveness (6). Direct reprogramming, which enables the acquisition of CMs without going through pluripotent stem cells, is gaining attention as an alternative to produce cardiac cells (7). Directly induced CMs were first produced using three transcription factors (Gata4, Mef2c, and Tbx5) in 2010, and many studies have been conducted to produce induced CMs more simply and efficiently by adding small molecules or microRNAs (8-10). Although the iPSC technology is superior in terms of expandability and efficiency, direct reprogramming can provide cardiac lineage cells with less tumorigenicity via relatively simple process (11).

Non-myocytes for cardiac tissue models

Since cardiac tissue models, composed only of CMs, lack maturity compared to the native heart, the cell composition of cardiac heart tissue models is currently being highlighted (12). An adult human heart contains many types of non-myocytes, including endothelial cells (ECs), cardiac fibroblast (CFs), and leukocytes, and their amount is even greater than that of CMs (13). Each plays a different role in the human heart and affects the maturity of CMs, which will eventually be important in creating disease models (14).

The ECs distributed inside the vessel form the major population of the heart, and serve to transport oxygen and nutrients to maintain the heart. For implementation of vascularized networks in cardiac tissue models, human umbilical vein endothelial cells (HUVECs), human cardiac microvascular endothelial cells (HMVECs), and human adipose-derived stem/stromal cells (hASCs) have widely been used (15-17). Although vascular structures can be produced using these cells, studies on the differentiation of human iPSCs to ECs are also actively underway, due to the strengths of reprogramming techniques, such as the possibility of mass production, development of patient-specific models, and simulation of interaction in developmental stages. ECs can be differentiated simultaneously with CMs from the cardiac mesoderm, and such differentiated ECs are advantageous for reconstituting vascular networks in heart (18). In recent years, more efficient EC differentiation methods have continued to be developed (19, 20). For example, the method of generating functional ECs with high purity from cardiogenic and hemogenic mesoderm was developed (20). For the same reason, the CF differentiation methods from human iPSC have also been developed (21, 22). Until now, CMs, ECs, and CFs have been recognized as the three major cell types for cardiac tissue models, and other non-myocytes, such as smooth muscle cells, have been occasionally added (23). Furthermore, as the immune response is a critical factor for cardiac homeostasis and disease pathophysiology, it is necessary to implement immune cells in 3D cardiac models (24). For example, macrophages in heart are known to interact with CMs via connexin 43 for improved electrical conduction (25).

Cellular interactions for cardiac tissue models

CMs and non-myocytes play important roles in the transmission of signals through interactions with other cells, and these interactions are also involved in disease progression and normal development. The ECs contribute to cardiac development, remodeling, and regeneration by interacting with CMs (26). They not only secrete several paracrine factors, such as nitric oxide (NO), neuregulin-1 (NRG-1), and apelin (APLN), to improve function and contractility of CMs during normal development, as well as to enhance the cardio-protection in a disease environment (26). ECs actively communicate with other non-myocytes. ECs resident in endocardium contribute to CF generation through endothelial-to-mesenchymal transition during normal embryonic development (27). ECs also participate in immune responses, helping immune cells migrate, by secreting cytokines, and even acting as an antigen presenter (28). The main role of CFs, generally located between CMs, is to construct the extracellular matrix (ECM) environment and maintain the homeostasis of ECM in the heart. In normal conditions, they communicate with CMs via gap junctions, membrane nanotubes, and paracrine signaling, and ultimately enhance the structural maturation and electrophysiological function of CMs (22, 29). However, excess activation and accumulation of CFs could be one of the phenotypes in cardiac diseases, such as myocardial infarction, cardiac fibrosis, and hypertensive heart disease. From this point of view, the implementation of CFs is important in establishing both the functionally advanced normal cardiac models and the heart disease models. The interactions between CFs and other non-myocytes have been examined in previous studies. When tested in the co-culture model, CFs assisted the proliferation of ECs and their sprouting (30). IL-1β expression in CFs, under a disease condition of myocarditis, has been shown to recruit leukocytes and induce the inflammatory process (31). Therefore, the incorporation of non-myocytes to cardiac tissue models is essential to replicate their interactions in a human heart tissue and predict the disease environment more precisely. In addition, it is required to develop culture platforms that can realize their interactions, including the secretion and absorption of cytokines, while each cell type can be maintained in optimal conditions.

ENGINEERED CARDIAC TISSUE MODELS

Cardiac spheroids

Various types of cardiac tissue models, possessing different characteristics and merits, can be produced depending on technologies used for fabrication. The simplest form of the model is a cardiac spheroid, which can be mainly prepared using devices that enable the collection of single cells, including a microwell, hanging drop plate, and V-bottom plate (22, 32, 33). The multicellular cardiac spheroids are fabricated by combination of CMs, ECs, and CFs. For instance, multicellular spheroids comprising human iPSC-derived CMs (70%), ECs (15%), and CFs (15%) showed enhanced electrical maturation and contractile phenotypes, compared with multicellular spheroids generated with other fibroblasts (e.g., skin fibroblasts) (22). In another study, spheroid models produced with different cell compositions (50% CMs, 25% ECs, and 25% CFs) were used to investigate the crosstalk between three cell types in doxorubicin-induced cardiotoxicity mediated by nitric oxide synthase (32). Since these methods can easily produce a large amount of uniform-sized 3D tissue models mixed with various cells, it is suitable for high-throughput drug screening (34). Therefore, cardiac spheroids are likely to be used as a screening platform for determining the efficacy and cardiotoxicity of the drug in pharmaceutical industry.

Engineered heart tissues

Engineered heart tissue (EHT) models, first established by Zimmerman et al., can form the desired shape by using a mold and contraction of the hydrogel (35). Depending on the structure of the mold, EHTs of various shapes such as ring, rod, and patch, have been fabricated. The EHT model can recapitulate the environment of in vivo CMs in a simple way. The alignment of CMs can be naturally developed by contraction of hydrogel, which eventually enhances electrical integration and synchronized contraction of the tissue models (36). The use of hydrogel enables the implementation of cell-ECM interaction and the encapsulation of bioactive proteins or functional materials (e.g., carbon nanotubes, gold nanostructure, and graphene oxide). Moreover, incorporation of additional devices, such as solid/elastic pillars with micromanipulator, allows the application of mechanical stimulation or the measurement of contractile force (37, 38).

Heart-on-a-chip

Organ-on-a-chip technology has been applied to develop in vitro models of various organs over a number of years. Cardiac tissue models can be manufactured using a microfluidic platform and cardiac cells, called a heart-on-a-chip (39). Microfluidic heart-on-a-chip shows unique advantages such as supply of well-controlled flow or stimulation, and the ability to analyze physiological functions by integrating biosensors (40-42). Since chips are generally fabricated using elastomer polydimethylsiloxane (PDMS), they have elastic mechanical properties suitable for stretching the entire device (39). Another advantage of the chip system is compartmentalization, which facilitates the co-culture of various cell types including CMs, ECs, and CFs in different chambers and channels. This is particularly helpful in establishing the vascularization of tissue models and, observing cell type-dependent drug-induced responses, such as contractile function of CMs and permeability change of ECs (40). To create a 3D cell structure on a chip, beyond the limitations of the conventional 2D cell layer, the EHT was integrated with microfluidic chips, leading to cardiac tissue models with the merits of each system. For example, the EHT formed in the chip could be electrically stimulated by 3D electrodes integrated in the chip, and endothelialization could also be induced through separate channels (43). Moreover, EHT fabricated using human iPSC-derived CMs (80%) and human iPSC-derived stromal cells (20%) within microfluidic chip showed an increase in cellular alignment and ECM production, and further matured under medium condition enriched with fatty acids (44).

Cardiac organoids

In recent years, the development of cardiac organoids, which mimic early heart development in the human body, has been demonstrated. The first developed cardiac organoids derived from aggregates of human iPSCs recapitulated the cardiomyogenesis of embryonic development and the expression of other surrounding regions and cells, such as mesenchymal cells and ECs (45). As cardiac organoids are generated via intrinsic development program, they can represent the in vivo human heart better than other cardiac tissue models. More recently, self-organizing cardiac organoids have been developed to contain the chamber with vascularization (46, 47). However, current cardiac organoids still remain in the early developmental stage (48). Therefore, efforts are required to produce more complex and mature cardiac organoids through long-term incubation in bioreactors and microfluidic chips or by incorporating additional cell types that do not develop naturally along with organoids (e.g., immune cells). In addition, unlike other cardiac models fabricated with defined cell numbers and controlled stages of cell differentiation, cardiac organoids may encounter the issues on batch variation and heterogeneity, which need to be addressed for standardization.

Cardiac tissues with functional hydrogels

Establishing ECM environments artificially in cardiac tissue models has been conducted using hydrogels prepared from synthetic materials (e.g., poly(ethylene glycol) and natural materials (e.g., fibrin, collagen, and Matrigel) (49). The hydrogels play a role in anchoring cells at certain locations, and influence cell differentiation and tissue formation through cell-matrix interactions. Hydrogels prepared from fibrin and collagen have been used to fabricate EHT models, and Matrigel has been used to embed cardiac organoids. Hydrogels made of synthetic materials have a low variability due to their chemically defined composition, and are easy to control mechanical and physical properties (e.g., modulus, stiffness). On the other hand, natural hydrogels exhibit superb biocompatibility and similarity to the native tissue environment (50). Decellularization technique allows removal of cellular components from native organs and tissues, resulting in fabrication of biological scaffolds containing tissue-specific ECM. Therefore, decellularized tissue-derived matrix can reconstitute the complex extracellular environments of the tissues (51-53). Decellularized tissue matrix has been manufactured as hydrogel and integrated into tissue models (54, 55). For instance, EHT model with heart ECM was fabricated using neonatal rat cardiomyocytes and 3D bioprinting (56). Decellularized heart-derived ECM hydrogel was used as a bioink with other supporting materials (e.g., poly (ethylene/vinyl acetate), which contributed to the increased alignment and differentiation of CMs in the EHT model (56). In another study, EHT model was established using human iPSC-CMs encapsulated in decellularized heart ECM hydrogel with reduced graphene oxide to implement tissue-specific biochemical cues and electrical conductivity (57). This hybrid scaffold enhanced contraction force, conduction velocity, calcium handling, and action potential duration of the EHT model (57).

Cardiac tissues stimulated with electrical/mechanical cues

Cardiac tissue models currently tend to be manufactured using human iPSCs. However, the maturity and functionality of these tissue models are still limited when compared to the in vivo human heart. To solve this problem, many efforts to create advanced models have proceeded in the direction of recapitulating the microenvironment and signaling of the native heart (58). Thus, in addition to incorporation of non-myocytes or a chip system, as previously mentioned, the stimulation of cardiac tissue models with an electrical pulse and cyclic stretch, has been tested for cardiac maturation. Electrical signals, which play a critical role in inducing the conduction of heartbeat, are associated with the functional phenotypes of CMs such as electrical interconnectivity and synchronized contraction (59, 60). Accordingly, the malfunction of electrical signal often leads to heart rhythm problems such as arrhythmia (59). When electrical stimulation was applied to the cardiac tissue models through bioreactors and biowire devices, structural and functional maturity could be substantially enhanced (Fig. 2A) (61, 62). Similarly, application of cyclic stretch using elastic scaffolds and stretchable bioreactors promoted the maturation of EHT models through the formation of aligned structure and increased electrical functionality (Fig. 2B, C) (63, 64). Another study reported that maturation of EHT model fabricated with early-stage human iPSC-CMs was enhanced by gradually increasing the frequency of electrical and mechanical stimulation (from 2 Hz to 6 Hz by 0.33 Hz per day) (65). Maturity of this EHT model was similar to that of the adult heart, which was confirmed through various analyses, such as ultrastructure, oxidative metabolism, frequency-dependent acceleration of relaxation, positive force-frequency relationship, and calcium handling (65).

HEART DISEASE MODELING

Cardiomyopathy

Cardiomyopathy is a heart disease indicated by enlarged, thickened, or stiffened heart muscle, which is associated with several intrinsic and extrinsic factors, but has been mainly recognized as an inherited disorder (66). Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM), characterized by a thickening of the left ventricle and dilated left ventricle, respectively, are representative examples among the several types of cardiomyopathies (67). HCM is caused, in the majority of cases, by mutations of genes that encode the sarcomere proteins, while DCM can be caused by more diverse factors such as mutation of sarcomere and cytoskeletal proteins, systemic pathology, maternal mitochondrial DNA, and X-linked mutations (68).

For cardiomyopathies caused by genetic mutations, disease models can be produced using either patient-derived iPSCs or gene editing systems like CRISPR/Cas9 if the genetic information is known. In 2015, EHT was developed for DCM modeling using patient-derived iPSC-CMs which have mutations in the TTN gene encoding titin, one of the filamentous proteins contained in sarcomeres (69). Interestingly, contractile deficits were not significantly observed in single cells, but they were expressed in the developed DCM EHT model (69). These results may be attributed to the increased CM alignment and enhanced contractile proteins in the EHT models. The TTN gene-deficient EHT models were manufactured via the CRISPR/Cas9 gene editing, which was verified by confirming reduced contraction. Barth syndrome (BTHS), caused by mutations in the TAZ gene encoding tafazzin, is a disease characterized by the combinational symptoms of cardiomyopathies such as HCM and DCM (70). The BTHS heart-on-chip model, developed using patient-derived iPSCs or the CRISPR/Cas9 gene editing system, showed a diminished contractile stress (71). This impaired phenotype was restored by treatment with TAZ nucleoside-modified messenger RNA (modRNA) or linoleic acid (71). Ma et al. constructed a myosin-binding protein C (MYBPC3) deficient human iPSC line using TALEN-mediated gene editing. Cardiac microtissues were then generated using MYBPC3-deficient human iPSC-CMs and synthetic filamentous matrices fabricated via two-photon polymerization (72). They found that the lack of a MYBPC3 (MYBPC3−/−) gene in a mechanically overloaded environment driven by filamentous matrices caused impaired contractile phenotypes and calcium dynamics, as seen in HCM and DCM (72). The effect of heterozygous gene deficiency (MYBPC3+/−) was also investigated using cardiac microtissue fabricated with micro-heart muscle arrays (73). In another study, the association of HCM with mutation in the ACTN2 gene, which encodes α-actinin 2, was investigated using an EHT model (Fig. 3A) (74). The ACTN2 mutant EHT model, fabricated with patient-derived iPSC-CMs and CRISPR/Cas9-mediated gene editing, possessed HCM phenotypes such as a longer action potential and the prolonged relaxation of contraction (74). As such, cardiac tissue models can be used to identify novel genes related to cardiomyopathy and investigate their roles in cardiomyopathy occurrence.

Cardiomyopathies occur often without genetic mutation and family history, such as nonfamilial HCM. These cases can be modeled through the provision of external factors or stimuli. Zhao et al. fabricated an EHT model using patient-derived iPSCs and a biowire chip, and then induced a left ventricular hypertrophy by chronic electrical stimulation for a period of up to 6 months (75). The left ventricular hypertrophy EHT model showed different mRNA profiles and contractile functions, when compared to a normal model (75). In a separate study, non-genetic cardiomyopathy was induced by treating angiotensin II in an EHT model that was constructed using human iPSC-CMs and human ventricular cardiac fibroblasts in a biowire chip (76). The EHT model of angiotensin II-induced progressive cardiomyopathy showed a gradually decreased contractile force and electrical activity, and was used for evaluation of the toxicity and efficacy of the drugs (losartan, relaxin, and saracatinib) (76).

Arrhythmia

Arrhythmia is a common disease in which an irregular heartbeat appears for various reasons. Arrhythmia is caused predominantly by problems in the electrical circuit in the heart (77). One of the major inherited arrhythmic disorders is long QT syndrome (LQTS), which is caused by mutation of cardiac ion channels. LQTS is named due to its clinical feature of a prolonged QT interval, and there are several types depending on which ion channel is affected (78). Thus, LQTS cardiac tissue models could be developed by incorporation of LQTS patient-derived cardiac cells or by the treatment of drugs that block specific cardiac ion channels. For instance, ring-shaped EHT models were generated with LQT2 patient-derived iPSC-CMs (79). These models exhibited a prolonged action potential duration, an abnormal calcium transient, and arrhythmic responses (79). The same research group demonstrated ring-shaped atrial arrhythmia in the EHT models and tested therapeutic strategies of electrical stimulation or anti-arrhythmic drugs (Fig. 3B) (80). QT prolongation in the heart could also occur due to the adverse action of the drugs (81). Previous study demonstrated the development of two types of LQTS EHT models (LQT1 and LQT2) through the treatment of drugs which inhibit each ion channel (HMR-1556 and E-4031 for blocking IKs and IKr, respectively) (82). Torsade de Pointes (TdP) means “twisting of the points” of heartbeat, which is one of the representative characteristics of life-threatening arrhythmia (81). As the occurrence of TdP, along with QT prolongation, contributes to the withdrawal of new drugs, drug safety screening systems have been focused on these features (83, 84). To model TdP in vitro, E-4031, which prolongs the field potential duration, was treated to cardiac tissue models, and consequently, spiral wave re-entry and TdP-like arrhythmic responses were observed (84). In another study, anatomical defects were induced to produce a structural arrhythmia disease model (85). After constructing ventricle models with neonatal rat ventricular myocytes and nanofiber spinning system, an artificial hole injury was generated to induce an arrhythmia with an abnormal flow of contraction (85).

Myocardial ischemia and myocardial infarction

Myocardial ischemia is characterized by a limited oxygen supply due to a blood vessel occlusion, which leads to the deterioration of heart function. Myocardial infarction is a life-threatening disease in which necrosis of the heart occurs due to extended ischemia or an acute blockage of the coronary artery (86). Models to simulate these events can be developed through culture in hypoxic conditions. For example, a previous study cultivated a cardiac tissue model fabricated using human iPSC-CMs in three types of hydrogels with different stiffness (0.8, 8, and 30 kPa) under conditions of a 1% oxygen concentration and investigated the effects of cell age and tissue stiffness on viability and reactive oxygen species (ROS) production (87). In another study, a myocardial infarction model was constructed by gradually decreasing the oxygen concentration towards the inside of the multicellular spheroids comprising 50% human iPSC-CMs and 50% non-myocytes (CFs, HUVECs, and hASCs) under conditions of a 10% oxygen concentration (88). This spherical disease model exhibited several pathological phenotypes, such as a fibrotic response and impaired calcium handling, and was used to test anti-fibrotic drugs and to identify the exacerbation of cardiotoxicity by drugs (88).

Reperfusion therapy is widely used to treat myocardial infarction, however, it can cause additional injury, called ischemia-reperfusion injury (IRI). IRI cardiac tissue models were first developed in the form of EHT in 2019 via a method of restoring oxygenation from ischemic conditions (89). These IRI models can be used for testing several therapeutic approaches targeting ischemic preconditioning, intracellular pH, the opening of the mitochondrial permeability transition pore (MPTP), and oxidative stress (89). Spherical IRI cardiac tissue models were also developed in another study using a similar inducing method, and these models showed some hallmarks of disease phenotypes such as cell death and increased secretion of cytokines associated with inflammation, angiogenesis, and cell migration (90). The angiogenic effect of IRI was confirmed through enhanced tube formation of HUVECs cultured with conditioned medium from spherical IRI tissue models (90). ECs are known to be an important mediator in myocardial dysfunction after IRI (91). To accurately predict pathological responses in IRI, it is necessary to implement the interaction between ECs and CMs. In this context, the cardioprotective roles of endothelial extracellular vesicles and their mechanism were investigated using an IRI heart-on-a-chip model (92). In a separate study, an IRI chip model was constructed through co-culture of human iPSC-CMs and iPSC-ECs in microfluidic chip, and changes in TSG101 and CD63 subunit expression of exosomes secreted in chip models were detected after ischemic injury and IRI, respectively (93).

Cardiac fibrosis

Severe heart defects caused by myocardial infarction and ischemia evoke cardiac fibrosis, which eventually leads to heart failure. CF, known to maintain the homeostasis of ECM, is a key cell type in cardiac fibrosis characterized by excessive ECM production and disrupted balance of ECM homeostasis (94). In this regard, cardiac tissue models based on CFs have been developed for modeling cardiac fibrosis, and various fibrosis induction methods have been used. To simulate a fibrotic response in cardiac tissue models, the application of biomechanical cues could be used. In many organs including the heart, overexpression of transforming growth factor-β (TGF-β) signaling is known to accelerate fibrosis after injury by activating fibroblasts, remodeling ECM, and promoting myofibroblast conversion (95). Accordingly, activation of TGF-β signaling resulted in a fibrotic response of the cardiac tissue model with CMs and CFs encapsulated in gelatin methacryloyl (GelMA) hydrogels (96). Moreover, cyclic mechanical compression was applied to a microdevice containing CFs encapsulated with GelMA hydrogels, and this triggered a phenotypic remodeling of CFs to myofibroblasts (97). In another study, cardiac fibrosis-on-a-chip was designed and fabricated in the form of EHT containing human iPSC-CMs and CFs, and fibrosis was induced by activation of TGF-β signaling (98). This fibrosis EHT model in chip showed hallmarks of fibrosis such as collagen deposition and increased stiffness, and therapeutic efficacy of anti-fibrotic drugs like pirfenidone was examined (98).

The fibrotic cardiac tissue model could be optimized by tuning the ratio of CMs and CFs. Wang et al. used a biowire chip for generating EHT models, and a 3:1 and 1:3 ratio of CMs and CFs was incorporated for a normal EHT model and cardiac fibrosis EHT model, respectively (99). The resultant fibrotic EHT model showed numerous disease phenotypes such as collagen deposition, diminished contractile function, abnormal calcium transient, and impaired electrophysiological properties (99). The drug effect was investigated using p-guanidinomethyl-phenylacetyl-Arg-Val-Arg-4-amidinobenzylamide (PCI) in fibrotic EHT models at early and late time points, and the changes in passive tension, active force, and collagen deposition were compared with those of normal EHT model (99). Early treatment of PCI reduced passive tension, active force, and collagen deposition in the fibrotic EHT model, whereas only passive tension was significantly decreased by the treatment of PCI at late time points (99). In another study, Daly et al. developed a cardiac fibrosis model by adjusting the ratio of CFs in a cardiac tissue model manufactured by assembling spheroids using 3D bioprinting (Fig. 3C) (100). In this study, a total of 20% of CFs were used for the healthy spheroids and 80% of CFs were used for scarred spheroids. The developed fibrotic spheroids had a severely damaged contraction profiles (100). With the advantage of bioprinting, it was possible to create a cardiac tissue model that was locally scarred in a specific area, and the delay of the contraction due to scarred region was spatially demonstrated (100). When the cholesterol modified miR-302b/c was applied for cardiac repair in the fibrotic model, recovery of contractile phenotype and activation delay was observed (100).

CONCLUSION

Owing to the development of stem cell and reprogramming technologies and the inaccuracy and ethical issues in animal experiments, 3D cardiac model production is in the spotlight. Numerous cardiac tissue models, each having their own advantages, have been developed and shown the benefits of modeling heart diseases. The cardiac tissue models that incorporate genetic diseases have been realized by using reprogramming and gene editing techniques. Modeling heart diseases with various risk factors in addition to genetic factors, such as myocardial infarction and cardiac fibrosis, has been achieved via creation of disease-specific microenvironments. Despite significant improvement in heart disease modeling, the complexity and maturity of the cardiac tissue models are still insufficient to simulate the complicated features and pathophysiology of heart diseases. Therefore, application and incorporation of tissue-engineering platforms (e.g., functional biomaterial scaffolds, microfluidic chip, bioprinting, and electroconductive materials, etc.) need to be further expanded for precise recapitulation of the cardiac microenvironments, induction of apparent disease phenotypes, and high-throughput culture and analysis.

ACKNOWLEDGEMENTS

This work was supported by the Bio & Medical Technology Development Program (2022M3A9B6082675) of the National Research Foundation of Korea (NRF) funded by the Korean government, the Ministry of Science and ICT (MSIT). This work was also supported by the Yonsei Signature Research Cluster Program of 2021-22-0014.

CONFLICTS OF INTEREST

The authors have no conflicting interests.

FIGURES
Fig. 1. Cell components and engineering platforms for the development of cardiac tissue models. Several types of cardiac tissue models have been developed, to include various cardiac cells, using engineering platforms such as microwells, microfluidic devices, functional hydrogel, and bioprinting. This figure was created with BioRender.com.
Fig. 2. Biophysical stimulation to improve the cardiac tissue models. (A) Cardiac spheroids cultured in microbioreactors and subjected to electrical stimulation at various frequencies. Electrical signals increased expression of gap junction and sarcomere thickness in cardiac spheroids. Adapted from Eng et al. (61) (CC BY 4.0 license) Copyright 2016, The Authors, published by Springer Nature. (B) EHT models stimulated with uniaxial cyclic stretch (10% strain, 1 Hz) using a bioreactor. The expression of gap junction and sarcomere structure and electrical responses were enhanced by mechanical stimulation. Adapted from Massai et al. (63) (CC BY 4.0 license) Copyright 2020, The Authors, published by Frontiers Editorial Office. (C) Ring-shaped EHT models with electrical pulses and stretch conditioning, which showed improved biomechanical properties and electrical coupling. Adapted from Lu et al. (64) (CC BY 4.0 license) Copyright 2021, The Authors, published by Ivyspring International Publisher.
Fig. 3. Modeling various heart diseases using cardiac tissue models. (A) Cardiomyopathy modeling using EHT with genetic defects of ACTN2. Adapted from Prondzynski et al. (74) (CC BY 4.0 license) Copyright 2019, The Authors, published by John Wiley and Sons. (B) Atrial arrhythmia modeling using ring-shaped EHT. Adapted from Goldfracht et al. (80) (CC BY 4.0 license) Copyright 2020, The Authors, published by Springer Nature. (C) Cardiac fibrosis modeling by assembling spheroids using 3D bioprinting. Adapted from Daly et al. (100) (CC BY 4.0 license) Copyright 2021, The Authors, published by Springer Nature.
REFERENCES
  1. Roth GA, Mensah GA, Johnson CO et al (2020) Global burden of cardiovascular diseases and risk factors, 1990-2019: update from the GBD 2019 study. J Am Coll Cardiol 76, 2982-3021
    Pubmed KoreaMed CrossRef
  2. Tani H and Tohyama S (2022) Human engineered heart tissue models for disease modeling and drug discovery. Front Cell Dev Biol 10, 855763
    Pubmed KoreaMed CrossRef
  3. Liu N and Olson EN (2022) CRISPR modeling and correction of cardiovascular disease. Circ Res 130, 1827-1850
    Pubmed KoreaMed CrossRef
  4. Karbassi E, Fenix A, Marchiano S et al (2020) Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat Rev Cardiol 17, 341-359
    Pubmed KoreaMed CrossRef
  5. Yoshida Y and Yamanaka S (2017) Induced pluripotent stem cells 10 years later: for cardiac applications. Circ Res 120, 1958-1968
    Pubmed CrossRef
  6. Kahn-Krell A, Pretorius D, Ou J et al (2021) Bioreactor suspension culture: differentiation and production of cardiomyocyte spheroids from human induced pluripotent stem cells. Front Bioeng Biotechnol 9, 674260
    Pubmed KoreaMed CrossRef
  7. Sadahiro T, Yamanaka S and Ieda M (2015) Direct cardiac reprogramming: progress and challenges in basic biology and clinical applications. Circ Res 116, 1378-1391
    Pubmed CrossRef
  8. Ieda M, Fu JD, Delgado-Olguin P et al (2010) Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142, 375-386
    Pubmed KoreaMed CrossRef
  9. Wang H, Cao N, Spencer CI et al (2014) Small molecules enable cardiac reprogramming of mouse fibroblasts with a single factor, Oct4. Cell Rep 6, 951-960
    Pubmed KoreaMed CrossRef
  10. Paoletti C, Divieto C, Tarricone G, Di Meglio F, Nurzynska D and Chiono V (2020) MicroRNA-mediated direct reprogramming of human adult fibroblasts toward cardiac phenotype. Front Bioeng Biotechnol 8, 529
    Pubmed KoreaMed CrossRef
  11. Kurotsu S, Suzuki T and Ieda M (2017) Direct reprogramming, epigenetics, and cardiac regeneration. J Card Fail 23, 552-557
    Pubmed CrossRef
  12. Guo Y and Pu WT (2020) Cardiomyocyte maturation: new phase in development. Circ Res 126, 1086-1106
    Pubmed KoreaMed CrossRef
  13. Pinto AR, Ilinykh A, Ivey MJ et al (2016) Revisiting cardiac cellular composition. Circ Res 118, 400-409
    Pubmed KoreaMed CrossRef
  14. Kamo T, Akazawa H and Komuro I (2015) Cardiac nonmyocytes in the hub of cardiac hypertrophy. Circ Res 117, 89-98
    Pubmed CrossRef
  15. Morrissette-McAlmon J, Ginn B, Somers S et al (2020) Biomimetic model of contractile cardiac tissue with endothelial networks stabilized by adipose-derived stromal/stem cells. Sci Rep 10, 8387
    Pubmed KoreaMed CrossRef
  16. Tsukamoto Y, Akagi T and Akashi M (2020) Vascularized cardiac tissue construction with orientation by layer-by-layer method and 3D printer. Sci Rep 10, 5484
    Pubmed KoreaMed CrossRef
  17. Koivisto M, Tolvanen TA, Toimela T et al (2022) Functional human cell-based vascularised cardiac tissue model for biomedical research and testing. Sci Rep 12, 13459
    Pubmed KoreaMed CrossRef
  18. Giacomelli E, Bellin M, Sala L et al (2017) Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development 144, 1008-1017
    Pubmed KoreaMed CrossRef
  19. Hamad S, Derichsweiler D, Gaspar JA et al (2022) High-efficient serum-free differentiation of endothelial cells from human iPS cells. Stem Cell Res Ther 13, 251
    Pubmed KoreaMed CrossRef
  20. Palpant NJ, Pabon L, Friedman CE et al (2017) Generating high-purity cardiac and endothelial derivatives from patterned mesoderm using human pluripotent stem cells. Nat Protoc 12, 15-31
    Pubmed KoreaMed CrossRef
  21. Zhang J, Tao R, Campbell KF et al (2019) Functional cardiac fibroblasts derived from human pluripotent stem cells via second heart field progenitors. Nat Commun 10, 2238
    Pubmed KoreaMed CrossRef
  22. Giacomelli E, Meraviglia V, Campostrini G et al (2020) Human-iPSC-derived cardiac stromal cells enhance maturation in 3D cardiac microtissues and reveal non-cardiomyocyte contributions to heart disease. Cell Stem Cell 26, 862-879
    Pubmed KoreaMed CrossRef
  23. Kahn-Krell A, Pretorius D, Guragain B et al (2022) A three-dimensional culture system for generating cardiac spheroids composed of cardiomyocytes, endothelial cells, smooth-muscle cells, and cardiac fibroblasts derived from human induced-pluripotent stem cells. Front Bioeng Biotechnol 10, 908848
    Pubmed KoreaMed CrossRef
  24. Swirski FK and Nahrendorf M (2018) Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol 18, 733-744
    Pubmed CrossRef
  25. Hulsmans M, Clauss S, Xiao L et al (2017) Macrophages facilitate electrical conduction in the heart. Cell 169, 510-522
    Pubmed KoreaMed CrossRef
  26. Colliva A, Braga L, Giacca M and Zacchigna S (2020) Endothelial cell-cardiomyocyte crosstalk in heart development and disease. J Physiol 598, 2923-2939
    Pubmed KoreaMed CrossRef
  27. Karra R, Walter AO and Wu SM (2017) The relationship between cardiac endothelium and fibroblasts: it's complicated. J Clin Invest 127, 2892-2894
    Pubmed KoreaMed CrossRef
  28. Mai J, Virtue A, Shen J, Wang H and Yang XF (2013) An evolving new paradigm: endothelial cells--conditional innate immune cells. J Hematol Oncol 6, 61
    Pubmed KoreaMed CrossRef
  29. Hall C, Gehmlich K, Denning C and Pavlovic D (2021) Complex relationship between cardiac fibroblasts and cardiomyocytes in health and disease. J Am Heart Assoc 10, e019338
    Pubmed KoreaMed CrossRef
  30. Twardowski RL and Black LD 3rd (2014) Cardiac fibroblasts support endothelial cell proliferation and sprout formation but not the development of multicellular sprouts in a fibrin gel co-culture model. Ann Biomed Eng 42, 1074-1084
    Pubmed KoreaMed CrossRef
  31. Xuan Y, Chen C, Wen Z and Wang DW (2022) The roles of cardiac fibroblasts and endothelial cells in myocarditis. Front Cardiovasc Med 9, 882027
    Pubmed KoreaMed CrossRef
  32. Polonchuk L, Chabria M, Badi L et al (2017) Cardiac spheroids as promising in vitro models to study the human heart microenvironment. Sci Rep 7, 7005
    Pubmed KoreaMed CrossRef
  33. Min S, Lee HJ, Jin Y et al (2020) Biphasic electrical pulse by a micropillar electrode array enhances maturation and drug response of reprogrammed cardiac spheroids. Nano Lett 20, 6947-6956
    Pubmed CrossRef
  34. Lee JM, Park DY, Yang L et al (2018) Generation of uniform-sized multicellular tumor spheroids using hydrogel microwells for advanced drug screening. Sci Rep 8, 17145
    Pubmed KoreaMed CrossRef
  35. Zimmermann WH, Schneiderbanger K, Schubert P et al (2002) Tissue engineering of a differentiated cardiac muscle construct. Circ Res 90, 223-230
    Pubmed CrossRef
  36. Takada T, Sasaki D, Matsuura K et al (2022) Aligned human induced pluripotent stem cell-derived cardiac tissue improves contractile properties through promoting unidirectional and synchronous cardiomyocyte contraction. Biomaterials 281, 121351
    Pubmed CrossRef
  37. Zhang W, Kong CW, Tong MH et al (2017) Maturation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) in 3D collagen matrix: effects of niche cell supplementation and mechanical stimulation. Acta Biomater 49, 204-217
    Pubmed CrossRef
  38. Rivera-Arbelaez JM, Cofino-Fabres C, Schwach V et al (2022) Contractility analysis of human engineered 3D heart tissues by an automatic tracking technique using a standalone application. PLoS One 17, e0266834
    Pubmed KoreaMed CrossRef
  39. Marsano A, Conficconi C, Lemme M et al (2016) Beating heart on a chip: a novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 16, 599-610
    Pubmed CrossRef
  40. Paloschi V, Sabater-Lleal M, Middelkamp H et al (2021) Organ-on-a-chip technology: a novel approach to investigate cardiovascular diseases. Cardiovasc Res 117, 2742-2754
    Pubmed KoreaMed CrossRef
  41. Cho AN, Jin Y, An Y et al (2021) Microfluidic device with brain extracellular matrix promotes structural and functional maturation of human brain organoids. Nat Commun 12, 4730
    Pubmed KoreaMed CrossRef
  42. Kim J, Lee KT, Lee JS et al (2021) Fungal brain infection modelled in a human-neurovascular-unit-on-a-chip with a functional blood-brain barrier. Nat Biomed Eng 5, 830-846
    Pubmed CrossRef
  43. Vivas A, IJspeert C, Pan JY et al (2022) Generation and culture of cardiac microtissues in a microfluidic chip with a reversible open top enables electrical pacing, dynamic drug dosing and endothelial cell co-culture. Adv Mater Technol 7, 2101355
    CrossRef
  44. Huebsch N, Charrez B, Neiman G et al (2022) Metabolically driven maturation of human-induced-pluripotent-stem-cell-derived cardiac microtissues on microfluidic chips. Nat Biomed Eng 6, 372-388
    Pubmed CrossRef
  45. Drakhlis L, Biswanath S, Farr CM et al (2021) Human heart-forming organoids recapitulate early heart and foregut development. Nat Biotechnol 39, 737-746
    Pubmed KoreaMed CrossRef
  46. Hofbauer P, Jahnel SM, Papai N et al (2021) Cardioids reveal self-organizing principles of human cardiogenesis. Cell 184, 3299-3317
    Pubmed CrossRef
  47. Lewis-Israeli YR, Wasserman AH, Gabalski MA et al (2021) Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat Commun 12, 5142
    Pubmed KoreaMed CrossRef
  48. Cho J, Lee H, Rah W, Chang HJ and Yoon YS (2022) From engineered heart tissue to cardiac organoid. Theranostics 12, 2758-2772
    Pubmed KoreaMed CrossRef
  49. Cui H, Liu Y, Cheng Y et al (2014) In vitro study of electroactive tetraaniline-containing thermosensitive hydrogels for cardiac tissue engineering. Biomacromolecules 15, 1115-1123
    Pubmed CrossRef
  50. Elkhoury K, Morsink M, Sanchez-Gonzalez L, Kahn C, Tamayol A and Arab-Tehrany E (2021) Biofabrication of natural hydrogels for cardiac, neural, and bone tissue engineering applications. Bioact Mater 6, 3904-3923
    Pubmed KoreaMed CrossRef
  51. Jin Y, Lee JS, Kim J et al (2018) Three-dimensional brain-like microenvironments facilitate the direct reprogramming of fibroblasts into therapeutic neurons. Nat Biomed Eng 2, 522-539
    Pubmed CrossRef
  52. Lee JS, Roh YH, Choi YS et al (2019) Tissue beads: tissue‐specific extracellular matrix microbeads to potentiate reprogrammed cell‐based therapy. Adv Funct Mater 29, 1807803
    CrossRef
  53. Jin Y, Kim J, Lee JS et al (2018) Vascularized liver organoids generated using induced hepatic tissue and dynamic liver‐specific microenvironment as a drug testing platform. Adv Funct Mater 28, 1801954
    CrossRef
  54. Wang RM and Christman KL (2016) Decellularized myocardial matrix hydrogels: in basic research and preclinical studies. Adv Drug Deliv Rev 96, 77-82
    Pubmed KoreaMed CrossRef
  55. Kim S, Min S, Choi YS et al (2022) Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 13, 1692
    Pubmed KoreaMed CrossRef
  56. Das S, Kim SW, Choi YJ et al (2019) Decellularized extracellular matrix bioinks and the external stimuli to enhance cardiac tissue development in vitro. Acta Biomater 95, 188-200
    Pubmed CrossRef
  57. Tsui JH, Leonard A, Camp ND et al (2021) Tunable electroconductive decellularized extracellular matrix hydrogels for engineering human cardiac microphysiological systems. Biomaterials 272, 120764
    Pubmed KoreaMed CrossRef
  58. Song M, Jang Y, Kim SJ and Park Y (2022) Cyclic stretching induces maturation of human-induced pluripotent stem cell-derived cardiomyocytes through nuclear-mechanotransduction. Tissue Eng Regen Med 19, 781-792
    Pubmed KoreaMed CrossRef
  59. Stoppel WL, Kaplan DL and Black LD 3rd (2016) Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 96, 135-155
    Pubmed KoreaMed CrossRef
  60. Radisic M, Park H, Shing H et al (2004) Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proc Natl Acad Sci U S A 101, 18129-18134
    Pubmed KoreaMed CrossRef
  61. Eng G, Lee BW, Protas L et al (2016) Autonomous beating rate adaptation in human stem cell-derived cardiomyocytes. Nat Commun 7, 10312
    Pubmed KoreaMed CrossRef
  62. Nunes SS, Miklas JW, Liu J et al (2013) Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat Methods 10, 781-787
    Pubmed KoreaMed CrossRef
  63. Massai D, Pisani G, Isu G et al (2020) Bioreactor platform for biomimetic culture and in situ monitoring of the mechanical response of in vitro engineered models of cardiac tissue. Front Bioeng Biotechnol 8, 733
    Pubmed KoreaMed CrossRef
  64. Lu K, Seidel T, Cao-Ehlker X et al (2021) Progressive stretch enhances growth and maturation of 3D stem-cell-derived myocardium. Theranostics 11, 6138-6153
    Pubmed KoreaMed CrossRef
  65. Ronaldson-Bouchard K, Ma SP, Yeager K et al (2018) Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556, 239-243
    Pubmed KoreaMed CrossRef
  66. Elliott PM, Anastasakis A et al; Authors/Task Force m (2014) 2014 ESC guidelines on diagnosis and management of hypertrophic cardiomyopathy: the task force for the diagnosis and management of hypertrophic cardiomyopathy of the European Society of Cardiology (ESC). Eur Heart J 35, 2733-2779
    Pubmed CrossRef
  67. Cassani M, Fernandes S, Vrbsky J, Ergir E, Cavalieri F and Forte G (2020) Combining nanomaterials and developmental pathways to design new treatments for cardiac regeneration: the pulsing heart of advanced therapies. Front Bioeng Biotechnol 8, 323
    Pubmed KoreaMed CrossRef
  68. Harvey PA and Leinwand LA (2011) The cell biology of disease: cellular mechanisms of cardiomyopathy. J Cell Biol 194, 355-365
    Pubmed KoreaMed CrossRef
  69. Hinson JT, Chopra A, Nafissi N et al (2015) HEART DISEASE. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349, 982-986
    Pubmed KoreaMed CrossRef
  70. Clarke SL, Bowron A, Gonzalez IL et al (2013) Barth syndrome. Orphanet J Rare Dis 8, 23
    Pubmed KoreaMed CrossRef
  71. Wang G, McCain ML, Yang L et al (2014) Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat Med 20, 616-623
    Pubmed KoreaMed CrossRef
  72. Ma Z, Huebsch N, Koo S et al (2018) Contractile deficits in engineered cardiac microtissues as a result of MYBPC3 deficiency and mechanical overload. Nat Biomed Eng 2, 955-967
    Pubmed KoreaMed CrossRef
  73. Guo J, Jiang H, Oguntuyo K, Rios B, Boodram Z and Huebsch N (2021) Interplay of genotype and substrate stiffness in driving the hypertrophic cardiomyopathy phenotype in iPSC-micro-heart muscle arrays. Cell Mol Bioeng 14, 409-425
    Pubmed KoreaMed CrossRef
  74. Prondzynski M, Lemoine MD, Zech AT et al (2019) Disease modeling of a mutation in alpha-actinin 2 guides clinical therapy in hypertrophic cardiomyopathy. EMBO Mol Med 11, e11115
    Pubmed KoreaMed CrossRef
  75. Zhao Y, Rafatian N, Feric NT et al (2019) A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 176, 913-927
    Pubmed KoreaMed CrossRef
  76. Wang EY, Kuzmanov U, Smith JB et al (2021) An organ-on-a-chip model for pre-clinical drug evaluation in progressive non-genetic cardiomyopathy. J Mol Cell Cardiol 160, 97-110
    Pubmed CrossRef
  77. Waldmann V, Narayanan K, Combes N, Jost D, Jouven X and Marijon E (2018) Electrical cardiac injuries: current concepts and management. Eur Heart J 39, 1459-1465
    Pubmed CrossRef
  78. Moss AJ and Kass RS (2005) Long QT syndrome: from channels to cardiac arrhythmias. J Clin Invest 115, 2018-2024
    Pubmed KoreaMed CrossRef
  79. Goldfracht I, Efraim Y, Shinnawi R et al (2019) Engineered heart tissue models from hiPSC-derived cardiomyocytes and cardiac ECM for disease modeling and drug testing applications. Acta Biomater 92, 145-159
    Pubmed CrossRef
  80. Goldfracht I, Protze S, Shiti A et al (2020) Generating ring-shaped engineered heart tissues from ventricular and atrial human pluripotent stem cell-derived cardiomyocytes. Nat Commun 11, 75
    Pubmed KoreaMed CrossRef
  81. Schwartz PJ and Woosley RL (2016) Predicting the unpredictable: drug-induced QT prolongation and torsades de pointes. J Am Coll Cardiol 67, 1639-1650
    Pubmed CrossRef
  82. Lemoine MD, Krause T, Koivumaki JT et al (2018) Human induced pluripotent stem cell-derived engineered heart tissue as a sensitive test system for QT prolongation and arrhythmic triggers. Circ Arrhythm Electrophysiol 11, e006035
    Pubmed CrossRef
  83. Fermini B, Hancox JC, Abi-Gerges N et al (2016) A new perspective in the field of cardiac safety testing through the comprehensive in vitro proarrhythmia assay paradigm. J Biomol Screen 21, 1-11
    Pubmed CrossRef
  84. Kawatou M, Masumoto H, Fukushima H et al (2017) Modelling torsade de pointes arrhythmias in vitro in 3D human iPS cell-engineered heart tissue. Nat Commun 8, 1078
    Pubmed KoreaMed CrossRef
  85. MacQueen LA, Sheehy SP, Chantre CO et al (2018) A tissue-engineered scale model of the heart ventricle. Nat Biomed Eng 2, 930-941
    Pubmed KoreaMed CrossRef
  86. Munderere R, Kim SH, Kim C and Park SH (2022) The progress of stem cell therapy in myocardial-infarcted heart regeneration: cell sheet technology. Tissue Eng Regen Med 19, 969-986
    Pubmed KoreaMed CrossRef
  87. Acun A, Nguyen TD and Zorlutuna P (2019) In vitro aged, hiPSC-origin engineered heart tissue models with age-dependent functional deterioration to study myocardial infarction. Acta Biomater 94, 372-391
    Pubmed KoreaMed CrossRef
  88. Richards DJ, Li Y, Kerr CM et al (2020) Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng 4, 446-462
    Pubmed KoreaMed CrossRef
  89. Chen T and Vunjak-Novakovic G (2019) Human tissue-engineered model of myocardial ischemia-reperfusion injury. Tissue Eng Part A 25, 711-724
    Pubmed KoreaMed CrossRef
  90. Sebastiao MJ, Gomes-Alves P, Reis I et al (2020) Bioreactor-based 3D human myocardial ischemia/reperfusion in vitro model: a novel tool to unveil key paracrine factors upon acute myocardial infarction. Transl Res 215, 57-74
    Pubmed CrossRef
  91. Singhal AK, Symons JD, Boudina S, Jaishy B and Shiu YT (2010) Role of endothelial cells in myocardial ischemia-reperfusion injury. Vasc Dis Prev 7, 1-14
    Pubmed KoreaMed CrossRef
  92. Yadid M, Lind JU, Ardona HAM et al (2020) Endothelial extracellular vesicles contain protective proteins and rescue ischemia-reperfusion injury in a human heart-on-chip. Sci Transl Med 12, eaax8005
    Pubmed KoreaMed CrossRef
  93. Ellis BW, Ronan G, Ren X et al (2022) Human Heart Anoxia and Reperfusion Tissue (HEART) model for the rapid study of exosome bound miRNA expression as biomarkers for myocardial infarction. Small 18, e2201330
    Pubmed KoreaMed CrossRef
  94. Travers JG, Kamal FA, Robbins J, Yutzey KE and Blaxall BC (2016) Cardiac fibrosis: the fibroblast awakens. Circ Res 118, 1021-1040
    Pubmed KoreaMed CrossRef
  95. Saadat S, Noureddini M, Mahjoubin-Tehran M et al (2020) Pivotal role of TGF-beta/Smad signaling in cardiac fibrosis: non-coding RNAs as effectual players. Front Cardiovasc Med 7, 588347
    Pubmed KoreaMed CrossRef
  96. Sadeghi AH, Shin SR, Deddens JC et al (2017) Engineered 3D cardiac fibrotic tissue to study fibrotic remodeling. Adv Healthc Mater 6, 201601434
    Pubmed KoreaMed CrossRef
  97. Kong M, Lee J, Yazdi IK et al (2019) Cardiac fibrotic remodeling on a chip with dynamic mechanical stimulation. Adv Healthc Mater 8, e1801146
    Pubmed KoreaMed CrossRef
  98. Mastikhina O, Moon BU, Williams K et al (2020) Human cardiac fibrosis-on-a-chip model recapitulates disease hallmarks and can serve as a platform for drug testing. Biomaterials 233, 119741
    Pubmed CrossRef
  99. Wang EY, Rafatian N, Zhao Y et al (2019) Biowire model of interstitial and focal cardiac fibrosis. ACS Cent Sci 5, 1146-1158
    Pubmed KoreaMed CrossRef
  100. Daly AC, Davidson MD and Burdick JA (2021) 3D bioprinting of high cell-density heterogeneous tissue models through spheroid fusion within self-healing hydrogels. Nat Commun 12, 753
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Author ORCID Information

Funding Information
  • National Research Foundation of Korea
      10.13039/501100003725
      2022M3A9B6082675
  • Ministry of Science and ICT, South Korea
      10.13039/501100014188
     
  • Yonsei Signature Research Cluster Program
      2021-22-0014

Collections

Services
Social Network Service

e-submission

Archives