Next Article in Journal
Quantitative Analysis of Core Lipid Production in Methanothermobacter marburgensis at Different Scales
Next Article in Special Issue
Advances in Cardiac Tissue Engineering
Previous Article in Journal
A Comparison of Heart Pulsations Provided by Forcecardiography and Double Integration of Seismocardiogram
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells

1
Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
2
Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
*
Author to whom correspondence should be addressed.
Bioengineering 2022, 9(4), 168; https://doi.org/10.3390/bioengineering9040168
Submission received: 25 February 2022 / Revised: 6 April 2022 / Accepted: 8 April 2022 / Published: 10 April 2022
(This article belongs to the Special Issue Advances in Cardiac Tissue Engineering)

Abstract

:
Human induced pluripotent stem cells (hiPSCs) can be used to generate various cell types in the human body. Hence, hiPSC-derived cardiomyocytes (hiPSC-CMs) represent a significant cell source for disease modeling, drug testing, and regenerative medicine. The immaturity of hiPSC-CMs in two-dimensional (2D) culture limit their applications. Cardiac tissue engineering provides a new promise for both basic and clinical research. Advanced bioengineered cardiac in vitro models can create contractile structures that serve as exquisite in vitro heart microtissues for drug testing and disease modeling, thereby promoting the identification of better treatments for cardiovascular disorders. In this review, we will introduce recent advances of bioengineering technologies to produce in vitro cardiac tissues derived from hiPSCs.

1. Introduction

Cardiovascular diseases (CVD) remain the leading cause of mortality worldwide [1]. According to statistics of the American Heart Association, there are more than 2300 deaths from CVD each day; on average, someone dies of CVD every 36 seconds in the United States. Every year, the United States spend an estimated $363.4 billion on CVD and it is projected that these costs will increase to 1044 billion dollars by 2030 [2].
The differentiation of induced pluripotent stem cells (iPSCs) into cardiac cells over the past decade has provided a powerful tool for cardiovascular research [2]. In 2006, Takahashi and Yamanaka reported, in a seminal paper, the possibility to induce iPSCs through reprogramming adult fibroblast by c-Myc, Oct3/4, Sox2, and Klf4 transcription factors [3]. Since then, iPSCs have been shown to differentiate into any cell type of human body including neurons, hepatocytes, and cardiomyocytes (CMs). Thus, iPSC technology provides a great opportunity to produce patient specific cell lines that can be used to study mechanisms of diseases, drug discovery, and therapeutic testing in vitro [4,5]. Patient- specific iPSCs-derived CMs (hiPSC-CMs) can mimic aspects of genetic diseases, metabolic dysfunction, and drug responses. However, hiPSC-CMs show properties similar to fetal human CM rather than adult CM [6]. The process of testing new drugs for safety and efficacy is time consuming and expensive [7,8]. These procedures are often carried out on two-dimensional (2D) cell culture and animal models, which are unable to properly reproduce the human physiological conditions [9]. In 2D in vitro models, single cell types are used, which do not accurately mimic functional multicellular tissue in vivo. A three-dimensional (3D) culture environment is necessary to obtain a proper CM phenotype, in a way as similar as possible to in vivo cardiac cells.
Henceforward, there is a need to improve CM differentiation in order to properly recapitulate the physiological and pathological characteristics of adult heart cells. 3D culture and engineering strategies can overcome 2D culture limitations. A combination of iPSC with 3D culture models can better replicate an in vivo microenvironment, including cell–cell and cell–matrix interactions, chemical, mechanical properties, and drug activities. The physiological immaturity of CMs is attenuated using combination iPSC technology with advancements in engineered biomaterials, such as microelectromechanical systems devices, heart-on-chip, bioprinting, and organoid technology [10,11,12]. In this review, we will summarize the most updated bioengineering approaches harnessed to generate in vitro cardiac tissues (Figure 1).

2. 3D Culture Using Scaffold-Free and Scaffold-Based Approaches

The generation of mature human CMs is a focal challenge in cardiovascular research. Unfortunately, CMs differentiated from hiPSCs in 2D conditions are immature in terms of morphology, expression of sarcomeric proteins, metabolism, and electrophysiology properties, such as excitation–contraction coupling (ECC) and contraction velocity [13]. Adult CMs are elongated, multinucleated, rod-shaped with organized sarcomeres, and exhibit a well-developed calcium (Ca2+) handling system, T-tubule organization, and action potential (AP) [14].
In contrast, hiPSC-CMs are mononucleated with disorganized sarcomeres and immature in terms of electrical coupling, AP, and gene expression (MYH6 > MYH7, TNNI > TNNI3, low expression of electrophysiological channels including KCNJ2, KCNJ8, KCNH7, SCN5A, HCN4, GJA1, CACNA1C, RYR2, ATP2A2, CASQ2, and CAV3) [15,16,17,18]. Small molecule-based approaches allow the obtainment of healthy hiPSC-CMs and patient specific hiPSC-CMs in small-scale and large-scale. Temporal modulation of Wnt/β-catenin signaling by the GSK3 inhibitor CHIR99021 (leading to an activation of Wnt signaling) or IWR1 or IWP2 (inhibitors of Wnt signaling) orchestrate CM differentiation [19,20,21,22]. Other studies demonstrated that 3D culture of hiPSC-CMs increased maturation as indicated by organized sarcomere structures, a rise in MYH7/MYH6 and TNNI3/TNNI1 ratio, potassium (K+) channels such as KCHJ2, and a metabolic switch from glucose or lactate to fatty acids [23,24,25,26,27].
There are two main methods for 3D culture of hiPSC-CM: scaffold-free and scaffold-based. In the scaffold-free method, hiPSC-CMs are cultured by aggregation through spinner flasks, hanging drop, non-adhesive U-shaped wells, V-bottom 96 well microplates, agarose microwells, and agitation culture; spherical microtissues are formed by hiPSC-CM alone or in co-culture with stromal cells [28,29]. In the scaffold-based method, hiPSC-CMs are encapsulated into extracellular matrix (ECM), natural hydrogels (fibrin, collagen, gelatin, and chitosan), or synthetic hydrogels (polycaprolactone, polyethylene glycol, and poly vinyl alcohol) [28,30,31,32,33,34,35,36].
ECM composition has been determined as one of the instrumental factors necessary for the maturation of neonatal CM by affecting the assembly of myofibrils [37]. Decellularized ECM obtained from the heart offers a network of glycosaminoglycans, proteoglycans, and other proteins that are present in native tissues, representing ideal biochemical and mechanical characteristics for proliferation and differentiation of cardiac cells, expression of cardiac markers of cardiac progenitor cells, and ultimately cardiac regeneration [38].
Hydrogels derived from decellularized tissues mimicking the properties of the native extracellular matrix can create a natural tissue microenvironment to support cellular processes [39,40,41]. Various studies showed that culturing hiPSC-CMs on ECM increased force contraction, cellular organization, and expression of late markers [42,43,44,45]. Decellularized ECMs are used to generate cardiac patches [46]. Decellularized tissues have already been tested in therapeutic applications such as heart valve implantation [47]. There is a high viability of hiPSC-CM within ECM or other hydrogels, which is very useful for cardiac drug testing [48,49].
The combination of 3D culture with other approaches, including co-culture with non-myocytes (endothelial cells, fibroblasts, and mesenchymal cells) [15,30,50,51,52,53,54,55,56], mechanical [57,58,59] and electrical [60,61] stimulation, addition of hormones like insulin-like growth factor-1 (IGF-1), or thyroid hormone, [14,62,63], could enhance hiPSC-CMs maturity (Figure 2).
The molecular interactions between endothelial cells, fibroblasts, and vascular smooth muscle cells (VSMCs) are essential for CM maturation and contractility as well as disease modelling. Indeed, co-culturing hiPSC-CMs with fibroblasts improved contractility [28] and several studies demonstrated that the overexpression of KCNJ2 [64], CASQ [65], and a combination of miRNAs (e.g., miR-125b, miR-199a, miR-221, and miR-222) [66] increased maturation properties of CMs; for instance, in terms of hyperpolarized resting membrane potential and Ca2+ handling. Treatment of hiPSC-CMs with triiodothyronine (T3) increased cell size, organization of sarcomere, contraction, mitochondrial biogenesis, and reduced proliferation [62]. Additionally, a combination of thyroid hormone, dexamethasone, and insulin-like growth factor-1 (TDI) enhanced sarcomeric alignment, Ca2+-transient kinetics, expression of ion channels (KCNJ2, SCN1B and RYR2), density of mitochondria, and T-tubule formation [67]. Recently, Anna Skorska and coworkers used super-resolution microscopy and deep machine learning to evaluate the quality of sarcomere network in hiPSC-CM; to improve the maturity of CMs, prolonged in vitro culture, treatment with thyroid hormone and dexamethasone, and micro patterned surfaces were applied, detecting an increase in sarcomere density, sarcomere length, and sarcomere alignment [68]. Equally important, in order to model long QT syndrome type 3 (LQT3) and dilated cardiomyopathy with mutation in SCNA5A and RBM20, respectively, Feyen and collaborators developed a maturation medium consisting of DMEM (free glucose), albuMAX, L-carnitine, taurine, creatine, and ascorbic acid; this medium increased CM maturation in 2D and 3D cultures, as noted by reduction of beating rate and proliferation, high fatty acid uptake and respiration rates, enhanced inward rectifier K+ current (IK1), diastolic membrane potential, upstroke velocity of action potential (AP), and Ca2+ handling [69].
Bioengineering approaches are used to generate engineered heart tissue (EHT) (ring-shaped, cylindrical, or longitudinal structures) using ECM, hydrogels, scaffolds, and microwell molds [70]. Culturing EHT in a bioreactor with electrical stimulation enhances the expression of cardiac specific proteins [71]. Electrical stimulation of a 3D culture of hiPSC-CMs with human fibroblasts in fibrin hydrogel upregulated gene expression of RYR2, ITPR3, KCNH2, MYH7, and CAV3 [59]. Electrical stimulation can increase the protein expression of Cx43, N-cadherin (N-Cad), and ZO-1, which improve the interaction between cells and release of Ca2+ [72,73]. Joseph Wu’s team generated an engineered heart muscle (EHM) by mixing human iPSC-CMs with human IMR-90 fibroblasts in collagen hydrogel under different stretching conditions [57]; EHMs exhibited upregulation of CAV (caveolin-3), KCNJ2, TNNT2, β-adrenergic receptors (ADRB1 and ADRB2), and the Ca2+ Voltage-Gated Channel Subunit Alpha1 C (CACNA1C), with a reduction of beating rate and a higher degree of Ca2+ amplitude [57]. Willem De Lange and colleagues introduced a novel model in which hiPSC-CMs and hiPSC-derived cardiac fibroblasts were cocultured in a 3D fibrin matrix to form engineered cardiac tissue constructs (hiPSC-ECTs), which are responsive to well-established physiological stimuli, including β-adrenergic stimulation, and stretch, and display Ca2+-handling and contractile kinetics that are similar to the human myocardium [74]. The combination of a PPARα agonist, dexamethasone, T3 and palmitate in media containing low glucose was shown to be very effective at inducing a mature phenotype in human pluripotent stem cell (hPSC)-derived ventricular CMs, increasing the expression of CX43 [75]. Recent work from the lab led by Jan Buikema elegantly illustrates that there is a complex biology behind the cellular and nuclear division of mono- and bi-nuclear CMs, with a shared-phenomenon of sarcomere disassembly during mitosis [76].
Innovative engineering approaches have made remarkable contributions to promote CM maturation. In the following section, we discuss some advanced technologies to create cardiac tissue.

3. Cardiac Organoids

The intrinsic capability of pluripotent stem cell (PSC) to discriminate various cell forms that are able to self-organize has implemented the possibility to generate ‘organs-in-a-dish’ known as organoids [77]. The word “organoid”, meaning ‘resembling an organ’, was first introduced by Smith and Cochrane [78] in 1946 to define a cystic teratoma. The term organoid refers to a 3D self-organized structure containing several cell types (organ specific) resulting from stem cells that summarize the in vivo organ’s architecture and function [79,80,81,82]. Since the organoid creation is a tremendous breakthrough in biological research, scientists are particularly interested in developing advancements in this model system to produce 3D tissues that can better mimic the organ’s physiology. The PSC and ASC (adult stem cells) generate the organoid and each of these components has specific applications. Organoids represent ideal suitable tools for disease modeling, regenerative medicine, and drug testing [83,84,85,86].
The first PSC-derived organoids were produced for the brain [87] and then other organoids have been generated for many other organs including optic cup [88,89], lung [90], kidney [91], liver [92,93], intestine [94,95], and stomach [96,97]. Despite extensive research on a variety of organoids, cardiac organoids have been less examined [98]. Cardiac organoids can reflect in vivo cardiogenesis and cardiac organoid models can ideally help us to better understand the mechanism of heart disease [70,99]. Organoids derived from patient-specific or genome-edited human iPSC lines provide a personalized platform for drug testing, disease modeling, and organ transplant [100].
There are two main methods to generate cardiac organoids, directed assembly and self-organization (also known as self-assembly). In the directed assembly method, CMs, endothelial cells, VSMCs, and fibroblasts are aggregated utilizing hydrogels, Matrigel or ECM [9,98,101,102]. In the second approach, hPSCs are triggered toward cardiac lineage after spheroid formation [103,104,105,106,107]. Direct assembly of cardiac organoids has been reported from co-culturing of human embryonic stem cells (hESC)-derived cardiac progenitor cells (hESC-CPC), endothelial cells, and mesenchymal stem cells, leading to improved maturation of CMs in terms of the expression of proteins involved in the typical CM structure and some key ion channel genes [108,109]. After co-aggregation, endothelial cells form vessel-like structures within the cardiac organoid.
One of the main problems of organoids is hypoxia in their central portion [110,111,112]. This issue can be disentangled by favoring the formation of vessel structures within spheroids and organoids. Vascularization of 3D hiPSC-CM tissue is essential for in vitro and in vivo survival [113,114,115] and can be achieved by providing an exquisite combination of growth factors, cytokines, and adhesion molecules triggering proliferation, differentiation, and regeneration of endothelial cells into 3D structures [116,117,118,119,120,121,122].
The LEFTY-PITX2 signaling pathway plays crucial roles in the maturation of cardiac organoids derived from hiPSC-cardiac mesoderm [123,124]. Using a specific cocktail of growth factors, Sasha Mendjan and coworkers were able to generate self-organizing cardioids using seeding hESCs and hiPSCs in 96-well plates (3D culture) followed by cardiac differentiation; the authors demonstrated that cavity morphogenesis is controlled by the mesodermal Wnt-BMP signaling axis [107], proving that cardioids represent a suitable platform to study mechanisms of cardiogenesis and heart diseases. 2-Cl-C.OXT-A (COA-Cl) increased contraction of cardiac organoid generated by co-culturing human dermal fibroblasts (HDFBs) with hiPSC- endothelial cells and hiPSC-CMs onto a 96-well plate with a spindle-shaped bottom [125]. As mentioned above, there is an ongoing challenge to recapitulate postnatal maturation of hPSC-CMs, overcoming an important limitation of their application for cell therapy or drug discovery. Mills and collaborators generated cardiac organoids by culturing hPSC-CMs and fibroblasts within heart-dyno platform; they found that key proliferation pathways including β-catenin and Yes-associated protein 1 (YAP1) were repressed during maturation [126]. The same group screened 5000 compounds on cardiac organoids to find activators of CM proliferation, identifying the mevalonate pathway as fundamental to reenter the cell cycle, both in vivo and in vitro [127]. In a recent study, Todd McDevitt’s research group described multilineage organoids emphasizing how the presence of gut tissue in an organoid provides paracrine factors and enhances CM maturation within hiPSC- organoids [128]. Similarly, Elisa Giacomelli and associates verified the relevance of cardiac fibroblasts in the maturation of 3D cardiac microtissues [56]: maturation of hiPSC-CMs was markedly promoted by culturing them with hiPSC endothelial cells and hiPSC-cardiac fibroblasts, with high reproducibility across lines, batches, and samples [56]. Taken together, all these studies demonstrate that organoid technology is a powerful tool both in biological studies and for clinical applications.

4. 3D Bioprinting

3D printing and bioprinting technologies have emerged as promising methods for the production of functional tissue and organ regeneration [129]. Bioprinting creates cardiac models by recapitulating in vivo cell structure, geometry, and chemical and physiomechanical properties through a precise spatial control of cells and biomaterials [130]. To mimic the native structure of heart tissue, the most used biomaterials in bioprinting are synthetic or natural hydrogel or decellularized matrices such as collagen, fibronectin, and gelatin [131]. Researchers have attempted to vascularize in vitro cardiac tissue structures by 3D printers and a Layer-by-Layer approaches [132,133]. Layer-by-Layer techniques are used to preserve cell-cell and cell-ECM interactions [134,135,136,137]. For instance, Yuto Amano and coworkers developed vascularized hiPSC-CM tissue by applying a filtration-Layer-by-Layer technique [138].
To support contraction and vascularization, normal human cardiac fibroblasts and endothelial cells have been introduced into the 3D hiPSC-CMs tissue together with fibronectin and collagen coating [138,139]. Schaefer and collaborators have generated a bi-layer patch composed of hiPSC-CMs, endothelial cells, and pericytes; the bi-layer approach revealed an increase in force production, maturation, and viability compared to hiPSC-CMs alone [140]. Using bioprinting, Narutoshi Hibino and colleagues were able to generate biomaterial-free 3D cardiac patches: cardiac spheroids were generated by co-culture hiPSC-CMs with endothelial cells and fibroblast and 3D cardiac patches were fabricated by 3D printer; both CX-43 (as a gap junction protein) and CD31 (as a blood vessel marker) were observed in these structures [141]. Similarly, Khademhosseini’s research team obtained an endothelialized myocardium based on 3D printing and organ-on-a-chip; this group bioprinted microfibrous scaffolds consisting of bioink and endothelial cells and then CMs were seeded into an endothelialized scaffold; then, the endothelialized myocardium was embedded into microfluidic systems to screen CVD-related drugs [142].

5. Heart-on-a-Chip

3D cell culture presents some limitations concerning the use of hydrogels, natural extracellular matrices, synthetic polymers, microtissues, or organoids, including variation in tissue size and shape, as well as insufficient nutrient supply [143,144,145]; the lack of proper vascular perfusion and tissue–tissue interfaces, such as the interfaces between vascular endothelium, connective tissues, and stromal cells, represent other critical aspects [146]. Microfluidic technology has provided an opportunity to develop organ-on-a-chip (OOAC) and overcome these limitations. Microfluidic systems allow the supply of culture media and removal of debris cells [147]. The combination of 3D models with microfluidics creates complex multi-organ intercommunication via metabolite and chemical exchange [148]. These microfluidic minimal functional units can reflect structural and functional characteristics of human tissues, including key parameters such as shear stress and culture medium flow rate [149,150], pH level [151], and organ-tissue interactions [149,150,151]. Polydimethylsiloxane (PDMS) is often used to create microfluidic channels [152,153,154].
Milica Radisic and colleagues reported a new approach to engineering organ-on-a-chip; they used a synthetic polymeric elastomer, a scaffold named Angio Tube, to fabricate microchannels [155]. This system, known as AngioChip, allows the incorporation of organoids into organ-on-a-chip and supports perfusable vascular system [155]. Examples reported in recent years show that the heart-on-a-chip can be used to assess drug toxicity [36,131,142]. The combination of heart-on-chip with electrical or mechanical stimulation, or microenvironmental cues [60,156,157] resulted in enhanced maturation of cardiac tissues at the cellular and electrophysiological level. Of note, Sakai and associates [158] cultured hiPSC-CMs and rat sympathetic neurons in separate microchambers connected by microtunnels, confirming that CM beating can be controlled by sympathetic neurons [159], and that this method could be useful for evaluating sympathetic induced-cardiotoxicity in cardiac tissue [158]. In another study, myocardium-on-chip was created via a three-channel device fabrication: hiPSC-CMs were encapsulated into UV cross-linkable methacrylated gelatin (GelMA) and then seeded within the central channel of the chip; hiPSC-derived endothelial cells were cultured into two side channels to better replicate the in vivo microvasculature [160].
A cardiac microphysiological system was successfully generated through means of the fabrication of microfluidic devices and loading beating CMs derived from hiPSC within the central part of chip [161]. There were “endothelial-like” barriers between the cell chamber and nutrient channels to mimic the human vasculature [161]. Intriguingly, heteropolar biowire chips were made by seeding atrial and ventricular CMs to the opposite ends of microchannels; electrical stimulation of cells within the biowire effectively enhanced the maturation of CMs towards adult-like properties [162].
Other investigators generated humanized multi-tissue organ-on-chip platforms including liver, brain, lung, and cardiac tissues for systemic toxicity screening [163,164,165,166,167,168,169,170,171]. Despite significant progress in cardiac tissue engineering, we need to better assess its potential for disease modeling.

6. Cardiac Disease Modeling by 3D Engineering Tissue

3D models of hiPSC-CM provide a valuable platform for disease modeling, studying mechanism of diseases, drug testing, and toxicity screening, especially when compared to 2D cultures. Modeling of cardiac diseases including myocardial fibrosis, dilated cardiomyopathy, and LQT syndrome has been reported using stem cell biology and tissue engineering approaches [18,172]. The Seidman laboratory at Harvard generated cardiac tissue engineered by culturing hiPSC-CMs with mutations in titin-truncating variants and mesenchymal stem cells in micropatterned substrates; these structures recapitulated characteristics of patients with dilated cardiomyopathy properties, including decreased contractile force and disability of truncated protein to assemble with sarcomeric component [173]. Consistent with these observations, 3D cardiac structures with mutations in PLN [174] and α-actinin 2 (ACTN2) [175] genes have been reported. Most recently, Richards and associates developed a cardiac organoid model of myocardial infarction by adding norepinephrine; they demonstrated that these cardiac organoids exhibit properties generally observed following myocardial infarction, such as fibrosis and alterations in Ca2+ handling, and can also recapitulate in vivo cardiac responses to drugs [98,176].

7. Quality Control of hiPSC-CMs by Artificial Intelligence and Machine Learning

The functional assessment of hiPSC-CMs is mainly based on the evaluation of changes in morphology, structure, gene expression, and electrophysiological patterns. In the last years, researchers have proposed to assess the quality of hiPSC-CMs by advanced methods such as artificial intelligence and machine learning [177,178]. This screening is performed in the reprogramming stage of iPSCs to exclude abnormal cells and in the differentiation stage to select mature CMs for cell therapy applications [179].
Ca2+ transients play a central role in ECC and functionality of CMs [13,180]. Changes in Ca2+ cycling can be monitored by machine learning methods in order to find valid cell lines for drug testing and eventually improve disease diagnostics and treatment. For instance, Juhola and collaborators studied the abnormalities of Ca2+ transient signals in different CVD, including catecholaminergic polymorphic ventricular tachycardia (CPVT), long QT syndrome (LQT), and hypertrophic cardiomyopathy (HCM), by signal analysis and machine learning methods [181,182,183,184]. The effects of adrenaline and dantrolene on Ca2+ cycling properties of (CPVT)-specific iPSC-CMs have also been assessed by machine learning [185]. These machine learning techniques with iPSC-CMs provide an outstanding platform to diagnose cardiac disease and to better understand drug toxicity for CVD [186].

8. Conclusions and Future Directions

Significant progress in iPSC technology and hiPSC-CM generation has provided advances in understanding genetic and pathology of diseases. However, hiPSC-CM are immature in 2D culture. 3D cardiac models show improvement in CM maturation, as shown by their ability to reliably recapitulate in vivo cardiac microenvironment.
Bioprinting, heart-on-chip, organoids, and natural and synthetic scaffolds systems are used to generate 3D cardiac tissue. Although there is still a need to improve maturation and tissue vascularization, we are confident that enhancements in bioengineering approaches and the integration of rapidly advancing technologies will soon allow the generation of excellent systems for personalized medicine.

Author Contributions

Writing—original draft preparation, F.V.; writing—review and editing, P.M. and G.S. All authors have read and agreed to the published version of the manuscript.

Funding

The Santulli’s Lab is supported in part by the National Institutes of Health (NIH: R01-HL146691, R01-HL159062, R01-DK033823, R01-DK123259, and T32-HL144456, to G.S.), by the Irma T. Hirschl and Monique Weill-Caulier Trusts (to G.S.), by the Diabetes Action Research and Education Foundation (to G.S.), and by the American Heart Association (AHA-22POST915561 to F.V.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update From the GBD 2019 Study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef]
  2. Mozaffarian, D.; Benjamin, E.J.; Go, A.S.; Arnett, D.K.; Blaha, M.J.; Cushman, M.; de Ferranti, S.; Despres, J.P.; Fullerton, H.J.; Howard, V.J.; et al. Heart disease and stroke statistics—2015 update: A report from the American Heart Association. Circulation 2015, 131, e29–e322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhao, Q.; Cai, H.; Zhan, Y.; Li, B.; Hu, S.; Sun, N. Applications of human-induced pluripotent stem cells in the investigation of inherited cardiomyopathy. Int. J. Cardiol. 2014, 177, 604–606. [Google Scholar] [CrossRef] [PubMed]
  5. Suh, T.C.; Amanah, A.Y.; Gluck, J.M. Electrospun Scaffolds and Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Cardiac Tissue Engineering Applications. Bioengineering 2020, 7, 105. [Google Scholar] [CrossRef] [PubMed]
  6. Karakikes, I.; Ameen, M.; Termglinchan, V.; Wu, J.C. Human induced pluripotent stem cell-derived cardiomyocytes: Insights into molecular, cellular, and functional phenotypes. Circ. Res. 2015, 117, 80–88. [Google Scholar] [CrossRef] [Green Version]
  7. Pimpaneau, V.; Voisin, E. Innovative medicine development. Technol. Health Care 2022, 30, 499–503. [Google Scholar] [CrossRef]
  8. Sardella, M.; Belcher, G.; Lungu, C.; Ignoni, T.; Camisa, M.; Stenver, D.I.; Porcelli, P.; D’Antuono, M.; Castiglione, N.G.; Adams, A.; et al. Monitoring the manufacturing and quality of medicines: A fundamental task of pharmacovigilance. Ther. Adv. Drug Saf. 2021, 12, 20420986211038436. [Google Scholar] [CrossRef]
  9. Buono, M.F.; von Boehmer, L.; Strang, J.; Hoerstrup, S.P.; Emmert, M.Y.; Nugraha, B. Human Cardiac Organoids for Modeling Genetic Cardiomyopathy. Cells 2020, 9, 1733. [Google Scholar] [CrossRef]
  10. Abulaiti, M.; Yalikun, Y.; Murata, K.; Sato, A.; Sami, M.M.; Sasaki, Y.; Fujiwara, Y.; Minatoya, K.; Shiba, Y.; Tanaka, Y.; et al. Establishment of a heart-on-a-chip microdevice based on human iPS cells for the evaluation of human heart tissue function. Sci. Rep. 2020, 10, 19201. [Google Scholar] [CrossRef]
  11. Hnatiuk, A.P.; Briganti, F.; Staudt, D.W.; Mercola, M. Human iPSC modeling of heart disease for drug development. Cell Chem. Biol. 2021, 28, 271–282. [Google Scholar] [CrossRef] [PubMed]
  12. Huch, M.; Knoblich, J.A.; Lutolf, M.P.; Martinez-Arias, A. The hope and the hype of organoid research. Development 2017, 144, 938–941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kansakar, U.; Varzideh, F.; Jankauskas, S.S.; Gambardella, J.; Trimarco, B.; Santulli, G. Advances in the understanding of excitation-contraction coupling: The pulsing quest for drugs against heart failure and arrhythmias. Eur. Heart J. Cardiovasc. Pharmacother. 2021, 7, e91–e93. [Google Scholar] [CrossRef] [PubMed]
  14. Guo, Y.; Pu, W.T. Cardiomyocyte Maturation: New Phase in Development. Circ. Res. 2020, 126, 1086–1106. [Google Scholar] [CrossRef] [PubMed]
  15. Veerman, C.C.; Kosmidis, G.; Mummery, C.L.; Casini, S.; Verkerk, A.O.; Bellin, M. Immaturity of human stem-cell-derived cardiomyocytes in culture: Fatal flaw or soluble problem? Stem Cells Dev. 2015, 24, 1035–1052. [Google Scholar] [CrossRef] [Green Version]
  16. Scuderi, G.J.; Butcher, J. Naturally Engineered Maturation of Cardiomyocytes. Front. Cell Dev. Biol. 2017, 5, 50. [Google Scholar] [CrossRef]
  17. Tenreiro, M.F.; Louro, A.F.; Alves, P.M.; Serra, M. Next generation of heart regenerative therapies: Progress and promise of cardiac tissue engineering. NPJ Regen. Med. 2021, 6, 30. [Google Scholar] [CrossRef]
  18. Li, J.; Hua, Y.; Miyagawa, S.; Zhang, J.; Li, L.; Liu, L.; Sawa, Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int. J. Mol. Sci. 2020, 21, 8893. [Google Scholar] [CrossRef]
  19. Deng, Y.; Lao, Y.; Ruan, Q.; Zhang, J.; Luo, C.; Shi, D.; Lu, F. Activation of Wnt/beta-Catenin Signaling Pathway Enhances the Derivation of Buffalo (Bubalus bubalis) Embryonic Stem Cell-Like Cells. Cell Reprogram. 2020, 22, 217–225. [Google Scholar] [CrossRef]
  20. Laco, F.; Woo, T.L.; Zhong, Q.; Szmyd, R.; Ting, S.; Khan, F.J.; Chai, C.L.L.; Reuveny, S.; Chen, A.; Oh, S. Unraveling the Inconsistencies of Cardiac Differentiation Efficiency Induced by the GSK3beta Inhibitor CHIR99021 in Human Pluripotent Stem Cells. Stem Cell Rep. 2018, 10, 1851–1866. [Google Scholar] [CrossRef] [Green Version]
  21. Halloin, C.; Schwanke, K.; Lobel, W.; Franke, A.; Szepes, M.; Biswanath, S.; Wunderlich, S.; Merkert, S.; Weber, N.; Osten, F.; et al. Continuous WNT Control Enables Advanced hPSC Cardiac Processing and Prognostic Surface Marker Identification in Chemically Defined Suspension Culture. Stem Cell Rep. 2019, 13, 775. [Google Scholar] [CrossRef] [PubMed]
  22. Mazzotta, S.; Neves, C.; Bonner, R.J.; Bernardo, A.S.; Docherty, K.; Hoppler, S. Distinctive Roles of Canonical and Noncanonical Wnt Signaling in Human Embryonic Cardiomyocyte Development. Stem Cell Rep. 2016, 7, 764–776. [Google Scholar] [CrossRef] [Green Version]
  23. Branco, M.A.; Cotovio, J.P.; Rodrigues, C.A.V.; Vaz, S.H.; Fernandes, T.G.; Moreira, L.M.; Cabral, J.M.S.; Diogo, M.M. Transcriptomic analysis of 3D Cardiac Differentiation of Human Induced Pluripotent Stem Cells Reveals Faster Cardiomyocyte Maturation Compared to 2D Culture. Sci. Rep. 2019, 9, 9229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Correia, C.; Koshkin, A.; Duarte, P.; Hu, D.; Carido, M.; Sebastiao, M.J.; Gomes-Alves, P.; Elliott, D.A.; Domian, I.J.; Teixeira, A.P.; et al. 3D aggregate culture improves metabolic maturation of human pluripotent stem cell derived cardiomyocytes. Biotechnol. Bioeng. 2018, 115, 630–644. [Google Scholar] [CrossRef] [PubMed]
  25. Li, J.; Zhang, L.; Yu, L.; Minami, I.; Miyagawa, S.; Horning, M.; Dong, J.; Qiao, J.; Qu, X.; Hua, Y.; et al. Circulating re-entrant waves promote maturation of hiPSC-derived cardiomyocytes in self-organized tissue ring. Commun. Biol. 2020, 3, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Zhang, D.; Shadrin, I.Y.; Lam, J.; Xian, H.Q.; Snodgrass, H.R.; Bursac, N. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 2013, 34, 5813–5820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Wu, P.; Deng, G.; Sai, X.; Guo, H.; Huang, H.; Zhu, P. Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci. Rep. 2021, 41, BSR20200833. [Google Scholar] [CrossRef] [PubMed]
  28. Beauchamp, P.; Jackson, C.B.; Ozhathil, L.C.; Agarkova, I.; Galindo, C.L.; Sawyer, D.B.; Suter, T.M.; Zuppinger, C. 3D Co-culture of hiPSC-Derived Cardiomyocytes With Cardiac Fibroblasts Improves Tissue-Like Features of Cardiac Spheroids. Front. Mol. Biosci. 2020, 7, 14. [Google Scholar] [CrossRef]
  29. Laco, F.; Lam, A.T.; Woo, T.L.; Tong, G.; Ho, V.; Soong, P.L.; Grishina, E.; Lin, K.H.; Reuveny, S.; Oh, S.K. Selection of human induced pluripotent stem cells lines optimization of cardiomyocytes differentiation in an integrated suspension microcarrier bioreactor. Stem Cell Res. Ther. 2020, 11, 118. [Google Scholar] [CrossRef] [Green Version]
  30. Varzideh, F.; Mahmoudi, E.; Pahlavan, S. Coculture with noncardiac cells promoted maturation of human stem cell-derived cardiomyocyte microtissues. J. Cell Biochem. 2019, 120, 16681–16691. [Google Scholar] [CrossRef]
  31. Archer, C.R.; Sargeant, R.; Basak, J.; Pilling, J.; Barnes, J.R.; Pointon, A. Characterization and Validation of a Human 3D Cardiac Microtissue for the Assessment of Changes in Cardiac Pathology. Sci. Rep. 2018, 8, 10160. [Google Scholar] [CrossRef] [PubMed]
  32. Mattapally, S.; Zhu, W.; Fast, V.G.; Gao, L.; Worley, C.; Kannappan, R.; Borovjagin, A.V.; Zhang, J. Spheroids of cardiomyocytes derived from human-induced pluripotent stem cells improve recovery from myocardial injury in mice. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H327–H339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lee, S.; Serpooshan, V.; Tong, X.; Venkatraman, S.; Lee, M.; Lee, J.; Chirikian, O.; Wu, J.C.; Wu, S.M.; Yang, F. Contractile force generation by 3D hiPSC-derived cardiac tissues is enhanced by rapid establishment of cellular interconnection in matrix with muscle-mimicking stiffness. Biomaterials 2017, 131, 111–120. [Google Scholar] [CrossRef] [PubMed]
  34. Khanna, A.; Zamani, M.; Huang, N.F. Extracellular Matrix-Based Biomaterials for Cardiovascular Tissue Engineering. J. Cardiovasc. Dev. Dis. 2021, 8, 137. [Google Scholar] [CrossRef] [PubMed]
  35. Almeida, H.V.; Tenreiro, M.F.; Louro, A.F.; Abecasis, B.; Santinha, D.; Calmeiro, T.; Fortunato, E.; Ferreira, L.; Alves, P.M.; Serra, M. Human Extracellular-Matrix Functionalization of 3D hiPSC-Based Cardiac Tissues Improves Cardiomyocyte Maturation. ACS Appl. Bio. Mater. 2021, 4, 1888–1899. [Google Scholar] [CrossRef]
  36. Andrysiak, K.; Stepniewski, J.; Dulak, J. Human-induced pluripotent stem cell-derived cardiomyocytes, 3D cardiac structures, and heart-on-a-chip as tools for drug research. Pflug. Arch. 2021, 473, 1061–1085. [Google Scholar] [CrossRef]
  37. Bayomy, A.F.; Bauer, M.; Qiu, Y.; Liao, R. Regeneration in heart disease-Is ECM the key? Life Sci. 2012, 91, 823–827. [Google Scholar] [CrossRef] [Green Version]
  38. Bejleri, D.; Davis, M.E. Decellularized Extracellular Matrix Materials for Cardiac Repair and Regeneration. Adv. Healthc. Mater. 2019, 8, e1801217. [Google Scholar] [CrossRef]
  39. Kim, Y.S.; Majid, M.; Melchiorri, A.J.; Mikos, A.G. Applications of decellularized extracellular matrix in bone and cartilage tissue engineering. Bioeng. Transl. Med. 2019, 4, 83–95. [Google Scholar] [CrossRef] [Green Version]
  40. Sackett, S.D.; Tremmel, D.M.; Ma, F.; Feeney, A.K.; Maguire, R.M.; Brown, M.E.; Zhou, Y.; Li, X.; O’Brien, C.; Li, L.; et al. Extracellular matrix scaffold and hydrogel derived from decellularized and delipidized human pancreas. Sci. Rep. 2018, 8, 10452. [Google Scholar] [CrossRef]
  41. Giobbe, G.G.; Crowley, C.; Luni, C.; Campinoti, S.; Khedr, M.; Kretzschmar, K.; De Santis, M.M.; Zambaiti, E.; Michielin, F.; Meran, L.; et al. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture. Nat. Commun. 2019, 10, 5658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hansen, A.; Eder, A.; Bonstrup, M.; Flato, M.; Mewe, M.; Schaaf, S.; Aksehirlioglu, B.; Schwoerer, A.P.; Uebeler, J.; Eschenhagen, T. Development of a drug screening platform based on engineered heart tissue. Circ. Res. 2010, 107, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Masumoto, H.; Yamashita, J.K. Human iPS cell-engineered three-dimensional cardiac tissues perfused by capillary networks between host and graft. Inflamm. Regen. 2018, 38, 26. [Google Scholar] [CrossRef] [PubMed]
  44. Turnbull, I.C.; Karakikes, I.; Serrao, G.W.; Backeris, P.; Lee, J.J.; Xie, C.; Senyei, G.; Gordon, R.E.; Li, R.A.; Akar, F.G.; et al. Advancing functional engineered cardiac tissues toward a preclinical model of human myocardium. FASEB J. 2014, 28, 644–654. [Google Scholar] [CrossRef] [Green Version]
  45. Jackman, C.P.; Carlson, A.L.; Bursac, N. Dynamic culture yields engineered myocardium with near-adult functional output. Biomaterials 2016, 111, 66–79. [Google Scholar] [CrossRef] [Green Version]
  46. Wang, Q.; Yang, H.; Bai, A.; Jiang, W.; Li, X.; Wang, X.; Mao, Y.; Lu, C.; Qian, R.; Guo, F.; et al. Functional engineered human cardiac patches prepared from nature’s platform improve heart function after acute myocardial infarction. Biomaterials 2016, 105, 52–65. [Google Scholar] [CrossRef]
  47. VeDepo, M.C.; Detamore, M.S.; Hopkins, R.A.; Converse, G.L. Recellularization of decellularized heart valves: Progress toward the tissue-engineered heart valve. J. Tissue Eng. 2017, 8, 2041731417726327. [Google Scholar] [CrossRef]
  48. Navaei, A.; Truong, D.; Heffernan, J.; Cutts, J.; Brafman, D.; Sirianni, R.W.; Vernon, B.; Nikkhah, M. PNIPAAm-based biohybrid injectable hydrogel for cardiac tissue engineering. Acta Biomater. 2016, 32, 10–23. [Google Scholar] [CrossRef]
  49. Blazeski, A.; Lowenthal, J.; Zhu, R.; Ewoldt, J.; Boheler, K.R.; Tung, L. Functional Properties of Engineered Heart Slices Incorporating Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cell Rep. 2019, 12, 982–995. [Google Scholar] [CrossRef] [Green Version]
  50. Tulloch, N.L.; Muskheli, V.; Razumova, M.V.; Korte, F.S.; Regnier, M.; Hauch, K.D.; Pabon, L.; Reinecke, H.; Murry, C.E. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ. Res. 2011, 109, 47–59. [Google Scholar] [CrossRef]
  51. Zhang, W.; Kong, C.W.; Tong, M.H.; Chooi, W.H.; Huang, N.; Li, R.A.; Chan, B.P. Maturation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) in 3D collagen matrix: Effects of niche cell supplementation and mechanical stimulation. Acta Biomater. 2017, 49, 204–217. [Google Scholar] [CrossRef] [PubMed]
  52. Burridge, P.W.; Metzler, S.A.; Nakayama, K.H.; Abilez, O.J.; Simmons, C.S.; Bruce, M.A.; Matsuura, Y.; Kim, P.; Wu, J.C.; Butte, M.; et al. Multi-cellular interactions sustain long-term contractility of human pluripotent stem cell-derived cardiomyocytes. Am. J. Transl. Res. 2014, 6, 724–735. [Google Scholar] [PubMed]
  53. Caspi, O.; Lesman, A.; Basevitch, Y.; Gepstein, A.; Arbel, G.; Habib, I.H.; Gepstein, L.; Levenberg, S. Tissue engineering of vascularized cardiac muscle from human embryonic stem cells. Circ. Res. 2007, 100, 263–272. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, C.; Majdi, M.; Xia, P.; Wei, K.A.; Talantova, M.; Spiering, S.; Nelson, B.; Mercola, M.; Chen, H.S. Non-cardiomyocytes influence the electrophysiological maturation of human embryonic stem cell-derived cardiomyocytes during differentiation. Stem Cells Dev. 2010, 19, 783–795. [Google Scholar] [CrossRef] [PubMed]
  55. Giacomelli, E.; Bellin, M.; Sala, L.; van Meer, B.J.; Tertoolen, L.G.; Orlova, V.V.; Mummery, C.L. Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development 2017, 144, 1008–1017. [Google Scholar] [CrossRef] [Green Version]
  56. Giacomelli, E.; Meraviglia, V.; Campostrini, G.; Cochrane, A.; Cao, X.; van Helden, R.W.J.; Krotenberg Garcia, A.; Mircea, M.; Kostidis, S.; Davis, R.P.; et al. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease. Cell Stem Cell 2020, 26, 862–879.e811. [Google Scholar] [CrossRef]
  57. Abilez, O.J.; Tzatzalos, E.; Yang, H.; Zhao, M.T.; Jung, G.; Zollner, A.M.; Tiburcy, M.; Riegler, J.; Matsa, E.; Shukla, P.; et al. Passive Stretch Induces Structural and Functional Maturation of Engineered Heart Muscle as Predicted by Computational Modeling. Stem Cells 2018, 36, 265–277. [Google Scholar] [CrossRef] [Green Version]
  58. Leonard, A.; Bertero, A.; Powers, J.D.; Beussman, K.M.; Bhandari, S.; Regnier, M.; Murry, C.E.; Sniadecki, N.J. Afterload promotes maturation of human induced pluripotent stem cell derived cardiomyocytes in engineered heart tissues. J. Mol. Cell Cardiol. 2018, 118, 147–158. [Google Scholar] [CrossRef]
  59. Ronaldson-Bouchard, K.; Ma, S.P.; Yeager, K.; Chen, T.; Song, L.; Sirabella, D.; Morikawa, K.; Teles, D.; Yazawa, M.; Vunjak-Novakovic, G. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 2018, 556, 239–243. [Google Scholar] [CrossRef]
  60. Nunes, S.S.; Miklas, J.W.; Liu, J.; Aschar-Sobbi, R.; Xiao, Y.; Zhang, B.; Jiang, J.; Masse, S.; Gagliardi, M.; Hsieh, A.; et al. Biowire: A platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat. Methods 2013, 10, 781–787. [Google Scholar] [CrossRef] [Green Version]
  61. Eng, G.; Lee, B.W.; Protas, L.; Gagliardi, M.; Brown, K.; Kass, R.S.; Keller, G.; Robinson, R.B.; Vunjak-Novakovic, G. Autonomous beating rate adaptation in human stem cell-derived cardiomyocytes. Nat. Commun. 2016, 7, 10312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Yang, X.; Rodriguez, M.; Pabon, L.; Fischer, K.A.; Reinecke, H.; Regnier, M.; Sniadecki, N.J.; Ruohola-Baker, H.; Murry, C.E. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 2014, 72, 296–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ribeiro, M.C.; Tertoolen, L.G.; Guadix, J.A.; Bellin, M.; Kosmidis, G.; D’Aniello, C.; Monshouwer-Kloots, J.; Goumans, M.J.; Wang, Y.L.; Feinberg, A.W.; et al. Functional maturation of human pluripotent stem cell derived cardiomyocytes in vitro—Correlation between contraction force and electrophysiology. Biomaterials 2015, 51, 138–150. [Google Scholar] [CrossRef] [PubMed]
  64. Lieu, D.K.; Fu, J.D.; Chiamvimonvat, N.; Tung, K.C.; McNerney, G.P.; Huser, T.; Keller, G.; Kong, C.W.; Li, R.A. Mechanism-based facilitated maturation of human pluripotent stem cell-derived cardiomyocytes. Circ. Arrhythm. Electrophysiol. 2013, 6, 191–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, J.; Lieu, D.K.; Siu, C.W.; Fu, J.D.; Tse, H.F.; Li, R.A. Facilitated maturation of Ca2+ handling properties of human embryonic stem cell-derived cardiomyocytes by calsequestrin expression. Am. J. Physiol. Cell Physiol. 2009, 297, C152–C159. [Google Scholar] [CrossRef] [Green Version]
  66. Lee, D.S.; Chen, J.H.; Lundy, D.J.; Liu, C.H.; Hwang, S.M.; Pabon, L.; Shieh, R.C.; Chen, C.C.; Wu, S.N.; Yan, Y.T.; et al. Defined MicroRNAs Induce Aspects of Maturation in Mouse and Human Embryonic-Stem-Cell-Derived Cardiomyocytes. Cell Rep. 2015, 12, 1960–1967. [Google Scholar] [CrossRef] [Green Version]
  67. Huang, C.Y.; Peres Moreno Maia-Joca, R.; Ong, C.S.; Wilson, I.; DiSilvestre, D.; Tomaselli, G.F.; Reich, D.H. Enhancement of human iPSC-derived cardiomyocyte maturation by chemical conditioning in a 3D environment. J. Mol. Cell. Cardiol. 2020, 138, 1–11. [Google Scholar] [CrossRef]
  68. Skorska, A.; Johann, L.; Chabanovska, O.; Vasudevan, P.; Kussauer, S.; Hillemanns, M.; Wolfien, M.; Jonitz-Heincke, A.; Wolkenhauer, O.; Bader, R.; et al. Monitoring the maturation of the sarcomere network: A super-resolution microscopy-based approach. Cell. Mol. Life Sci. 2022, 79, 149. [Google Scholar] [CrossRef]
  69. Feyen, D.A.M.; McKeithan, W.L.; Bruyneel, A.A.N.; Spiering, S.; Hormann, L.; Ulmer, B.; Zhang, H.; Briganti, F.; Schweizer, M.; Hegyi, B.; et al. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep. 2020, 32, 107925. [Google Scholar] [CrossRef]
  70. Seguret, M.; Vermersch, E.; Jouve, C.; Hulot, J.S. Cardiac Organoids to Model and Heal Heart Failure and Cardiomyopathies. Biomedicines 2021, 9, 563. [Google Scholar] [CrossRef]
  71. Visone, R.; Talo, G.; Lopa, S.; Rasponi, M.; Moretti, M. Enhancing all-in-one bioreactors by combining interstitial perfusion, electrical stimulation, on-line monitoring and testing within a single chamber for cardiac constructs. Sci. Rep. 2018, 8, 16944. [Google Scholar] [CrossRef] [PubMed]
  72. Ruan, J.L.; Tulloch, N.L.; Razumova, M.V.; Saiget, M.; Muskheli, V.; Pabon, L.; Reinecke, H.; Regnier, M.; Murry, C.E. Mechanical Stress Conditioning and Electrical Stimulation Promote Contractility and Force Maturation of Induced Pluripotent Stem Cell-Derived Human Cardiac Tissue. Circulation 2016, 134, 1557–1567. [Google Scholar] [CrossRef] [PubMed]
  73. LaBarge, W.; Mattappally, S.; Kannappan, R.; Fast, V.G.; Pretorius, D.; Berry, J.L.; Zhang, J. Maturation of three-dimensional, hiPSC-derived cardiomyocyte spheroids utilizing cyclic, uniaxial stretch and electrical stimulation. PLoS ONE 2019, 14, e0219442. [Google Scholar] [CrossRef]
  74. de Lange, W.J.; Farrell, E.T.; Kreitzer, C.R.; Jacobs, D.R.; Lang, D.; Glukhov, A.V.; Ralphe, J.C. Human iPSC-engineered cardiac tissue platform faithfully models important cardiac physiology. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1670–H1686. [Google Scholar] [CrossRef] [PubMed]
  75. Funakoshi, S.; Fernandes, I.; Mastikhina, O.; Wilkinson, D.; Tran, T.; Dhahri, W.; Mazine, A.; Yang, D.; Burnett, B.; Lee, J.; et al. Generation of mature compact ventricular cardiomyocytes from human pluripotent stem cells. Nat. Commun. 2021, 12, 3155. [Google Scholar] [CrossRef]
  76. Yuan, Q.; Maas, R.G.C.; Brouwer, E.C.J.; Pei, J.; Blok, C.S.; Popovic, M.A.; Paauw, N.J.; Bovenschen, N.; Hjortnaes, J.; Harakalova, M.; et al. Sarcomere Disassembly and Transfection Efficiency in Proliferating Human iPSC-Derived Cardiomyocytes. J. Cardiovasc. Dev. Dis. 2022, 9, 43. [Google Scholar] [CrossRef]
  77. Murrow, L.M.; Weber, R.J.; Gartner, Z.J. Dissecting the stem cell niche with organoid models: An engineering-based approach. Development 2017, 144, 998–1007. [Google Scholar] [CrossRef] [Green Version]
  78. Smith, E.; Cochrane, W. Cystic organoid teratoma:(report of a case). Can. Med. Assoc. J. 1946, 55, 151. [Google Scholar]
  79. Clevers, H. Modeling Development and Disease with Organoids. Cell 2016, 165, 1586–1597. [Google Scholar] [CrossRef] [Green Version]
  80. Sasai, Y. Cytosystems dynamics in self-organization of tissue architecture. Nature 2013, 493, 318–326. [Google Scholar] [CrossRef]
  81. Sasai, Y. Next-generation regenerative medicine: Organogenesis from stem cells in 3D culture. Cell Stem Cell 2013, 12, 520–530. [Google Scholar] [CrossRef] [Green Version]
  82. Woodford, C.; Zandstra, P.W. Tissue engineering 2.0: Guiding self-organization during pluripotent stem cell differentiation. Curr. Opin. Biotechnol. 2012, 23, 810–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Xu, H.; Jiao, Y.; Qin, S.; Zhao, W.; Chu, Q.; Wu, K. Organoid technology in disease modelling, drug development, personalized treatment and regeneration medicine. Exp. Hematol. Oncol. 2018, 7, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lancaster, M.A.; Knoblich, J.A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef] [PubMed]
  85. Forsythe, S.D.; Devarasetty, M.; Shupe, T.; Bishop, C.; Atala, A.; Soker, S.; Skardal, A. Environmental Toxin Screening Using Human-Derived 3D Bioengineered Liver and Cardiac Organoids. Front. Public Health 2018, 6, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Nugraha, B.; Buono, M.F.; von Boehmer, L.; Hoerstrup, S.P.; Emmert, M.Y. Human Cardiac Organoids for Disease Modeling. Clin. Pharmacol. Ther. 2019, 105, 79–85. [Google Scholar] [CrossRef] [Green Version]
  87. Eiraku, M.; Watanabe, K.; Matsuo-Takasaki, M.; Kawada, M.; Yonemura, S.; Matsumura, M.; Wataya, T.; Nishiyama, A.; Muguruma, K.; Sasai, Y. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 2008, 3, 519–532. [Google Scholar] [CrossRef] [Green Version]
  88. Eiraku, M.; Takata, N.; Ishibashi, H.; Kawada, M.; Sakakura, E.; Okuda, S.; Sekiguchi, K.; Adachi, T.; Sasai, Y. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 2011, 472, 51–56. [Google Scholar] [CrossRef]
  89. Nakano, T.; Ando, S.; Takata, N.; Kawada, M.; Muguruma, K.; Sekiguchi, K.; Saito, K.; Yonemura, S.; Eiraku, M.; Sasai, Y. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell 2012, 10, 771–785. [Google Scholar] [CrossRef] [Green Version]
  90. Miller, A.J.; Dye, B.R.; Ferrer-Torres, D.; Hill, D.R.; Overeem, A.W.; Shea, L.D.; Spence, J.R. Generation of lung organoids from human pluripotent stem cells in vitro. Nat. Protoc. 2019, 14, 518–540. [Google Scholar] [CrossRef]
  91. Zeng, Z.; Huang, B.; Parvez, R.K.; Li, Y.; Chen, J.; Vonk, A.C.; Thornton, M.E.; Patel, T.; Rutledge, E.A.; Kim, A.D.; et al. Generation of patterned kidney organoids that recapitulate the adult kidney collecting duct system from expandable ureteric bud progenitors. Nat. Commun. 2021, 12, 3641. [Google Scholar] [CrossRef] [PubMed]
  92. Takebe, T.; Sekine, K.; Enomura, M.; Koike, H.; Kimura, M.; Ogaeri, T.; Zhang, R.R.; Ueno, Y.; Zheng, Y.W.; Koike, N.; et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013, 499, 481–484. [Google Scholar] [CrossRef] [PubMed]
  93. Takebe, T.; Zhang, R.R.; Koike, H.; Kimura, M.; Yoshizawa, E.; Enomura, M.; Koike, N.; Sekine, K.; Taniguchi, H. Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant. Nat. Protoc. 2014, 9, 396–409. [Google Scholar] [CrossRef] [PubMed]
  94. Lukonin, I.; Serra, D.; Challet Meylan, L.; Volkmann, K.; Baaten, J.; Zhao, R.; Meeusen, S.; Colman, K.; Maurer, F.; Stadler, M.B.; et al. Phenotypic landscape of intestinal organoid regeneration. Nature 2020, 586, 275–280. [Google Scholar] [CrossRef]
  95. Watanabe, S.; Kobayashi, S.; Ogasawara, N.; Okamoto, R.; Nakamura, T.; Watanabe, M.; Jensen, K.B.; Yui, S. Transplantation of intestinal organoids into a mouse model of colitis. Nat. Protoc. 2022, 17, 649–671. [Google Scholar] [CrossRef]
  96. McCauley, H.A.; Wells, J.M. Pluripotent stem cell-derived organoids: Using principles of developmental biology to grow human tissues in a dish. Development 2017, 144, 958–962. [Google Scholar] [CrossRef] [Green Version]
  97. McCracken, K.W.; Cata, E.M.; Crawford, C.M.; Sinagoga, K.L.; Schumacher, M.; Rockich, B.E.; Tsai, Y.H.; Mayhew, C.N.; Spence, J.R.; Zavros, Y.; et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature 2014, 516, 400–404. [Google Scholar] [CrossRef] [Green Version]
  98. Richards, D.J.; Coyle, R.C.; Tan, Y.; Jia, J.; Wong, K.; Toomer, K.; Menick, D.R.; Mei, Y. Inspiration from heart development: Biomimetic development of functional human cardiac organoids. Biomaterials 2017, 142, 112–123. [Google Scholar] [CrossRef]
  99. Zhao, D.; Lei, W.; Hu, S. Cardiac organoid—A promising perspective of preclinical model. Stem Cell Res. Ther. 2021, 12, 272. [Google Scholar] [CrossRef]
  100. Hulot, J.S. Modeling Cardiac Arrhythmias With Organoids. J. Am. Coll. Cardiol. 2019, 73, 2325–2327. [Google Scholar] [CrossRef]
  101. Polonchuk, L.; Chabria, M.; Badi, L.; Hoflack, J.C.; Figtree, G.; Davies, M.J.; Gentile, C. Cardiac spheroids as promising in vitro models to study the human heart microenvironment. Sci. Rep. 2017, 7, 7005. [Google Scholar] [CrossRef] [PubMed]
  102. Brassard, J.A.; Lutolf, M.P. Engineering Stem Cell Self-organization to Build Better Organoids. Cell Stem Cell 2019, 24, 860–876. [Google Scholar] [CrossRef] [PubMed]
  103. Dang, S.M.; Kyba, M.; Perlingeiro, R.; Daley, G.Q.; Zandstra, P.W. Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems. Biotechnol. Bioeng. 2002, 78, 442–453. [Google Scholar] [CrossRef]
  104. Shkumatov, A.; Baek, K.; Kong, H. Matrix rigidity-modulated cardiovascular organoid formation from embryoid bodies. PLoS ONE 2014, 9, e94764. [Google Scholar] [CrossRef] [PubMed]
  105. Lewis-Israeli, Y.R.; Wasserman, A.H.; Gabalski, M.A.; Volmert, B.D.; Ming, Y.; Ball, K.A.; Yang, W.; Zou, J.; Ni, G.; Pajares, N.; et al. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat. Commun. 2021, 12, 5142. [Google Scholar] [CrossRef]
  106. Andersen, P.; Tampakakis, E.; Jimenez, D.V.; Kannan, S.; Miyamoto, M.; Shin, H.K.; Saberi, A.; Murphy, S.; Sulistio, E.; Chelko, S.P.; et al. Precardiac organoids form two heart fields via Bmp/Wnt signaling. Nat. Commun. 2018, 9, 3140. [Google Scholar] [CrossRef]
  107. Hofbauer, P.; Jahnel, S.M.; Papai, N.; Giesshammer, M.; Deyett, A.; Schmidt, C.; Penc, M.; Tavernini, K.; Grdseloff, N.; Meledeth, C.; et al. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 2021, 184, 3299–3317.e22. [Google Scholar] [CrossRef]
  108. Varzideh, F.; Pahlavan, S.; Ansari, H.; Halvaei, M.; Kostin, S.; Feiz, M.S.; Latifi, H.; Aghdami, N.; Braun, T.; Baharvand, H. Human cardiomyocytes undergo enhanced maturation in embryonic stem cell-derived organoid transplants. Biomaterials 2019, 192, 537–550. [Google Scholar] [CrossRef]
  109. Kerr, C.M.; Richards, D.; Menick, D.R.; Deleon-Pennell, K.Y.; Mei, Y. Multicellular Human Cardiac Organoids Transcriptomically Model Distinct Tissue-Level Features of Adult Myocardium. Int. J. Mol. Sci. 2021, 22, 8482. [Google Scholar] [CrossRef]
  110. Hubert, C.G.; Rivera, M.; Spangler, L.C.; Wu, Q.; Mack, S.C.; Prager, B.C.; Couce, M.; McLendon, R.E.; Sloan, A.E.; Rich, J.N. A Three-Dimensional Organoid Culture System Derived from Human Glioblastomas Recapitulates the Hypoxic Gradients and Cancer Stem Cell Heterogeneity of Tumors Found In Vivo. Cancer Res. 2016, 76, 2465–2477. [Google Scholar] [CrossRef] [Green Version]
  111. Daviaud, N.; Chevalier, C.; Friedel, R.H.; Zou, H. Distinct Vulnerability and Resilience of Human Neuroprogenitor Subtypes in Cerebral Organoid Model of Prenatal Hypoxic Injury. Front. Cell. Neurosci. 2019, 13, 336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kong, J.; Wen, S.; Cao, W.; Yue, P.; Xu, X.; Zhang, Y.; Luo, L.; Chen, T.; Li, L.; Wang, F.; et al. Lung organoids, useful tools for investigating epithelial repair after lung injury. Stem Cell Res. Ther. 2021, 12, 95. [Google Scholar] [CrossRef]
  113. Zhao, X.; Xu, Z.; Xiao, L.; Shi, T.; Xiao, H.; Wang, Y.; Li, Y.; Xue, F.; Zeng, W. Review on the Vascularization of Organoids and Organoids-on-a-Chip. Front. Bioeng. Biotechnol. 2021, 9, 637048. [Google Scholar] [CrossRef] [PubMed]
  114. Yu, J. Vascularized Organoids: A More Complete Model. Int. J. Stem Cells 2021, 14, 127–137. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, E.P.; Toksoy, Z.; Davis, B.A.; Geibel, J.P. 3D Bioprinting of Vascularized Tissues for in vitro and in vivo Applications. Front. Bioeng. Biotechnol. 2021, 9, 664188. [Google Scholar] [CrossRef]
  116. Narmoneva, D.A.; Vukmirovic, R.; Davis, M.E.; Kamm, R.D.; Lee, R.T. Endothelial cells promote cardiac myocyte survival and spatial reorganization: Implications for cardiac regeneration. Circulation 2004, 110, 962–968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Brutsaert, D.L. Cardiac endothelial-myocardial signaling: Its role in cardiac growth, contractile performance, and rhythmicity. Physiol. Rev. 2003, 83, 59–115. [Google Scholar] [CrossRef]
  118. Nolan, D.J.; Ginsberg, M.; Israely, E.; Palikuqi, B.; Poulos, M.G.; James, D.; Ding, B.S.; Schachterle, W.; Liu, Y.; Rosenwaks, Z.; et al. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev. Cell 2013, 26, 204–219. [Google Scholar] [CrossRef] [Green Version]
  119. Saini, H.; Navaei, A.; Van Putten, A.; Nikkhah, M. 3D cardiac microtissues encapsulated with the co-culture of cardiomyocytes and cardiac fibroblasts. Adv. Healthc. Mater. 2015, 4, 1961–1971. [Google Scholar] [CrossRef]
  120. Roberts, M.A.; Tran, D.; Coulombe, K.L.; Razumova, M.; Regnier, M.; Murry, C.E.; Zheng, Y. Stromal Cells in Dense Collagen Promote Cardiomyocyte and Microvascular Patterning in Engineered Human Heart Tissue. Tissue Eng. Part A 2016, 22, 633–644. [Google Scholar] [CrossRef]
  121. Peters, E.B. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. Tissue Eng. Part B Rev. 2018, 24, 1–24. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, F.M.; Liu, X. Advancing biomaterials of human origin for tissue engineering. Prog. Polym. Sci. 2016, 53, 86–168. [Google Scholar] [CrossRef] [Green Version]
  123. Song, M.H.; Choi, S.C.; Noh, J.M.; Joo, H.J.; Park, C.Y.; Cha, J.J.; Ahn, T.H.; Ko, T.H.; Choi, J.I.; Na, J.E.; et al. LEFTY-PITX2 signaling pathway is critical for generation of mature and ventricular cardiac organoids in human pluripotent stem cell-derived cardiac mesoderm cells. Biomaterials 2021, 278, 121133. [Google Scholar] [CrossRef] [PubMed]
  124. Lozano-Velasco, E.; Garcia-Padilla, C.; Del Mar Munoz-Gallardo, M.; Martinez-Amaro, F.J.; Cano-Carrillo, S.; Castillo-Casas, J.M.; Sanchez-Fernandez, C.; Aranega, A.E.; Franco, D. Post-Transcriptional Regulation of Molecular Determinants during Cardiogenesis. Int. J. Mol. Sci. 2022, 23, 2839. [Google Scholar] [CrossRef] [PubMed]
  125. Kitsuka, T.; Itoh, M.; Amamoto, S.; Arai, K.I.; Oyama, J.; Node, K.; Toda, S.; Morita, S.; Nishida, T.; Nakayama, K. 2-Cl-C.OXT-A stimulates contraction through the suppression of phosphodiesterase activity in human induced pluripotent stem cell-derived cardiac organoids. PLoS ONE 2019, 14, e0213114. [Google Scholar] [CrossRef] [Green Version]
  126. Mills, R.J.; Titmarsh, D.M.; Koenig, X.; Parker, B.L.; Ryall, J.G.; Quaife-Ryan, G.A.; Voges, H.K.; Hodson, M.P.; Ferguson, C.; Drowley, L.; et al. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. Proc. Natl. Acad. Sci. USA 2017, 114, E8372–E8381. [Google Scholar] [CrossRef] [Green Version]
  127. Mills, R.J.; Parker, B.L.; Quaife-Ryan, G.A.; Voges, H.K.; Needham, E.J.; Bornot, A.; Ding, M.; Andersson, H.; Polla, M.; Elliott, D.A.; et al. Drug Screening in Human PSC-Cardiac Organoids Identifies Pro-proliferative Compounds Acting via the Mevalonate Pathway. Cell Stem Cell 2019, 24, 895–907.e6. [Google Scholar] [CrossRef]
  128. Silva, A.C.; Matthys, O.B.; Joy, D.A.; Kauss, M.A.; Natarajan, V.; Lai, M.H.; Turaga, D.; Blair, A.P.; Alexanian, M.; Bruneau, B.G.; et al. Co-emergence of cardiac and gut tissues promotes cardiomyocyte maturation within human iPSC-derived organoids. Cell Stem Cell 2021, 28, 2137–2152.e6. [Google Scholar] [CrossRef]
  129. Kozaniti, F.K.; Metsiou, D.N.; Manara, A.E.; Athanassiou, G.; Deligianni, D.D. Recent Advancements in 3D Printing and Bioprinting Methods for Cardiovascular Tissue Engineering. Bioengineering 2021, 8, 133. [Google Scholar] [CrossRef]
  130. Hu, J.B.; Tomov, M.L.; Buikema, J.W.; Chen, C.; Mahmoudi, M.; Wu, S.M.; Serpooshan, V. Cardiovascular tissue bioprinting: Physical and chemical processes. Appl. Phys. Rev. 2018, 5, 041106. [Google Scholar] [CrossRef]
  131. Cui, H.; Miao, S.; Esworthy, T.; Zhou, X.; Lee, S.J.; Liu, C.; Yu, Z.X.; Fisher, J.P.; Mohiuddin, M.; Zhang, L.G. 3D bioprinting for cardiovascular regeneration and pharmacology. Adv. Drug Deliv. Rev. 2018, 132, 252–269. [Google Scholar] [CrossRef] [PubMed]
  132. Tsukamoto, Y.; Akagi, T.; Akashi, M. Vascularized cardiac tissue construction with orientation by layer-by-layer method and 3D printer. Sci. Rep. 2020, 10, 5484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Puluca, N.; Lee, S.; Doppler, S.; Munsterer, A.; Dressen, M.; Krane, M.; Wu, S.M. Bioprinting Approaches to Engineering Vascularized 3D Cardiac Tissues. Curr. Cardiol. Rep. 2019, 21, 90. [Google Scholar] [CrossRef] [PubMed]
  134. Hatami, J.; Silva, S.G.; Oliveira, M.B.; Costa, R.R.; Reis, R.L.; Mano, J.F. Multilayered Films Produced by Layer-by-Layer Assembly of Chitosan and Alginate as a Potential Platform for the Formation of Human Adipose-Derived Stem Cell aggregates. Polymers 2017, 9, 440. [Google Scholar] [CrossRef] [Green Version]
  135. Wang, L.; Zhang, J. Layer-By-Layer Fabrication of Thicker and Larger Human Cardiac Muscle Patches for Cardiac Repair in Mice. Front. Cardiovasc. Med. 2021, 8, 800667. [Google Scholar] [CrossRef]
  136. Al-Jubori, A.A.; Sulaiman, G.M.; Tawfeeq, A.T.; Mohammed, H.A.; Khan, R.A.; Mohammed, S.A.A. Layer-by-Layer Nanoparticles of Tamoxifen and Resveratrol for Dual Drug Delivery System and Potential Triple-Negative Breast Cancer Treatment. Pharmaceutics 2021, 13, 1098. [Google Scholar] [CrossRef]
  137. Takeda, M.; Miyagawa, S.; Akashi, M.; Sawa, Y. Construction of Three-Dimensional Cardiac Tissues Using Layer-by-Layer Method. Methods Mol. Biol. 2021, 2320, 75–79. [Google Scholar] [CrossRef]
  138. Amano, Y.; Nishiguchi, A.; Matsusaki, M.; Iseoka, H.; Miyagawa, S.; Sawa, Y.; Seo, M.; Yamaguchi, T.; Akashi, M. Development of vascularized iPSC derived 3D-cardiomyocyte tissues by filtration Layer-by-Layer technique and their application for pharmaceutical assays. Acta Biomater. 2016, 33, 110–121. [Google Scholar] [CrossRef]
  139. Tadano, K.; Miyagawa, S.; Takeda, M.; Tsukamoto, Y.; Kazusa, K.; Takamatsu, K.; Akashi, M.; Sawa, Y. Cardiotoxicity assessment using 3D vascularized cardiac tissue consisting of human iPSC-derived cardiomyocytes and fibroblasts. Mol. Ther. Methods Clin. Dev. 2021, 22, 338–349. [Google Scholar] [CrossRef]
  140. Schaefer, J.A.; Guzman, P.A.; Riemenschneider, S.B.; Kamp, T.J.; Tranquillo, R.T. A cardiac patch from aligned microvessel and cardiomyocyte patches. J. Tissue Eng. Regen. Med. 2018, 12, 546–556. [Google Scholar] [CrossRef]
  141. Ong, C.S.; Fukunishi, T.; Zhang, H.; Huang, C.Y.; Nashed, A.; Blazeski, A.; DiSilvestre, D.; Vricella, L.; Conte, J.; Tung, L.; et al. Biomaterial-Free Three-Dimensional Bioprinting of Cardiac Tissue using Human Induced Pluripotent Stem Cell Derived Cardiomyocytes. Sci. Rep. 2017, 7, 4566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Zhang, Y.S.; Arneri, A.; Bersini, S.; Shin, S.R.; Zhu, K.; Goli-Malekabadi, Z.; Aleman, J.; Colosi, C.; Busignani, F.; Dell’Erba, V.; et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 2016, 110, 45–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Chaicharoenaudomrung, N.; Kunhorm, P.; Noisa, P. Three-dimensional cell culture systems as an in vitro platform for cancer and stem cell modeling. World J. Stem Cells 2019, 11, 1065–1083. [Google Scholar] [CrossRef] [PubMed]
  144. Russo, M.; Cejas, C.M.; Pitingolo, G. Advances in microfluidic 3D cell culture for preclinical drug development. Prog. Mol. Biol. Transl. Sci. 2022, 187, 163–204. [Google Scholar] [CrossRef] [PubMed]
  145. Tibbitt, M.W.; Anseth, K.S. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol. Bioeng. 2009, 103, 655–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Bhatia, S.N.; Ingber, D.E. Microfluidic organs-on-chips. Nat. Biotechnol. 2014, 32, 760–772. [Google Scholar] [CrossRef] [PubMed]
  147. Cho, S.; Lee, C.; Skylar-Scott, M.A.; Heilshorn, S.C.; Wu, J.C. Reconstructing the heart using iPSCs: Engineering strategies and applications. J. Mol. Cell. Cardiol. 2021, 157, 56–65. [Google Scholar] [CrossRef] [PubMed]
  148. Doherty, E.L.; Aw, W.Y.; Hickey, A.J.; Polacheck, W.J. Microfluidic and Organ-on-a-Chip Approaches to Investigate Cellular and Microenvironmental Contributions to Cardiovascular Function and Pathology. Front. Bioeng. Biotechnol. 2021, 9, 624435. [Google Scholar] [CrossRef]
  149. Vollert, I.; Seiffert, M.; Bachmair, J.; Sander, M.; Eder, A.; Conradi, L.; Vogelsang, A.; Schulze, T.; Uebeler, J.; Holnthoner, W.; et al. In vitro perfusion of engineered heart tissue through endothelialized channels. Tissue Eng. Part A 2014, 20, 854–863. [Google Scholar] [CrossRef]
  150. Schneider, O.; Zeifang, L.; Fuchs, S.; Sailer, C.; Loskill, P. User-Friendly and Parallelized Generation of Human Induced Pluripotent Stem Cell-Derived Microtissues in a Centrifugal Heart-on-a-Chip. Tissue Eng. Part A 2019, 25, 786–798. [Google Scholar] [CrossRef] [Green Version]
  151. Mousavi Shaegh, S.A.; De Ferrari, F.; Zhang, Y.S.; Nabavinia, M.; Binth Mohammad, N.; Ryan, J.; Pourmand, A.; Laukaitis, E.; Banan Sadeghian, R.; Nadhman, A.; et al. A microfluidic optical platform for real-time monitoring of pH and oxygen in microfluidic bioreactors and organ-on-chip devices. Biomicrofluidics 2016, 10, 044111. [Google Scholar] [CrossRef] [PubMed]
  152. Mercader, A.; Ye, S.H.; Kim, S.; Orizondo, R.A.; Cho, S.K.; Wagner, W.R. PDMS-Zwitterionic Hybrid for Facile, Antifouling Microfluidic Device Fabrication. Langmuir 2022, 38, 3775–3784. [Google Scholar] [CrossRef] [PubMed]
  153. Choi, H.J.; Lee, J.H.; Jeong, O.C. Pneumatically Driven Microfluidic Platform for Micro-Particle Concentration. J. Vis. Exp. 2022, 180. [Google Scholar] [CrossRef] [PubMed]
  154. Xu, T.; Lim, Y.J.; Zheng, Y.; Jung, M.; Gaus, K.; Gardiner, E.E.; Lee, W.M. Modified inverted selective plane illumination microscopy for sub-micrometer imaging resolution in polydimethylsiloxane soft lithography devices. Lab Chip 2020, 20, 3960–3969. [Google Scholar] [CrossRef]
  155. Lai, B.F.L.; Lu, R.X.Z.; Davenport Huyer, L.; Kakinoki, S.; Yazbeck, J.; Wang, E.Y.; Wu, Q.; Zhang, B.; Radisic, M. A well plate-based multiplexed platform for incorporation of organoids into an organ-on-a-chip system with a perfusable vasculature. Nat. Protoc. 2021, 16, 2158–2189. [Google Scholar] [CrossRef]
  156. Marsano, A.; Conficconi, C.; Lemme, M.; Occhetta, P.; Gaudiello, E.; Votta, E.; Cerino, G.; Redaelli, A.; Rasponi, M. Beating heart on a chip: A novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 2016, 16, 599–610. [Google Scholar] [CrossRef]
  157. Zhao, Y.; Rafatian, N.; Wang, E.Y.; Feric, N.T.; Lai, B.F.L.; Knee-Walden, E.J.; Backx, P.H.; Radisic, M. Engineering microenvironment for human cardiac tissue assembly in heart-on-a-chip platform. Matrix Biol. 2020, 85–86, 189–204. [Google Scholar] [CrossRef]
  158. Sakai, K.; Shimba, K.; Ishizuka, K.; Yang, Z.; Oiwa, K.; Takeuchi, A.; Kotani, K.; Jimbo, Y. Functional innervation of human induced pluripotent stem cell-derived cardiomyocytes by co-culture with sympathetic neurons developed using a microtunnel technique. Biochem. Biophys. Res. Commun. 2017, 494, 138–143. [Google Scholar] [CrossRef]
  159. Carnevale, D. Neuroimmune axis of cardiovascular control: Mechanisms and therapeutic implications. Nat. Rev. Cardiol. 2022. [Google Scholar] [CrossRef]
  160. Ellis, B.W.; Acun, A.; Can, U.I.; Zorlutuna, P. Human iPSC-derived myocardium-on-chip with capillary-like flow for personalized medicine. Biomicrofluidics 2017, 11, 024105. [Google Scholar] [CrossRef] [Green Version]
  161. Mathur, A.; Loskill, P.; Shao, K.; Huebsch, N.; Hong, S.; Marcus, S.G.; Marks, N.; Mandegar, M.; Conklin, B.R.; Lee, L.P.; et al. Human iPSC-based cardiac microphysiological system for drug screening applications. Sci. Rep. 2015, 5, 8883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Zhao, Y.; Rafatian, N.; Feric, N.T.; Cox, B.J.; Aschar-Sobbi, R.; Wang, E.Y.; Aggarwal, P.; Zhang, B.; Conant, G.; Ronaldson-Bouchard, K.; et al. A Platform for Generation of Chamber-Specific Cardiac Tissues and Disease Modeling. Cell 2019, 176, 913–927.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Rajan, S.A.P.; Aleman, J.; Wan, M.; Pourhabibi Zarandi, N.; Nzou, G.; Murphy, S.; Bishop, C.E.; Sadri-Ardekani, H.; Shupe, T.; Atala, A.; et al. Probing prodrug metabolism and reciprocal toxicity with an integrated and humanized multi-tissue organ-on-a-chip platform. Acta Biomater. 2020, 106, 124–135. [Google Scholar] [CrossRef] [PubMed]
  164. Oleaga, C.; Bernabini, C.; Smith, A.S.; Srinivasan, B.; Jackson, M.; McLamb, W.; Platt, V.; Bridges, R.; Cai, Y.; Santhanam, N.; et al. Multi-Organ toxicity demonstration in a functional human in vitro system composed of four organs. Sci. Rep. 2016, 6, 20030. [Google Scholar] [CrossRef] [PubMed]
  165. Ma, C.; Peng, Y.; Li, H.; Chen, W. Organ-on-a-Chip: A New Paradigm for Drug Development. Trends Pharmacol. Sci. 2021, 42, 119–133. [Google Scholar] [CrossRef]
  166. Low, L.A.; Sutherland, M.; Lumelsky, N.; Selimovic, S.; Lundberg, M.S.; Tagle, D.A. Organs-on-a-Chip. Adv. Exp. Med. Biol. 2020, 1230, 27–42. [Google Scholar] [CrossRef]
  167. Young, R.E.; Huh, D.D. Organ-on-a-chip technology for the study of the female reproductive system. Adv. Drug Deliv. Rev. 2021, 173, 461–478. [Google Scholar] [CrossRef]
  168. Jalili-Firoozinezhad, S.; Miranda, C.C.; Cabral, J.M.S. Modeling the Human Body on Microfluidic Chips. Trends Biotechnol. 2021, 39, 838–852. [Google Scholar] [CrossRef]
  169. Jodat, Y.A.; Kang, M.G.; Kiaee, K.; Kim, G.J.; Martinez, A.F.H.; Rosenkranz, A.; Bae, H.; Shin, S.R. Human-Derived Organ-on-a-Chip for Personalized Drug Development. Curr. Pharm. Des. 2018, 24, 5471–5486. [Google Scholar] [CrossRef]
  170. Ajalik, R.E.; Alenchery, R.G.; Cognetti, J.S.; Zhang, V.Z.; McGrath, J.L.; Miller, B.L.; Awad, H.A. Human Organ-on-a-Chip Microphysiological Systems to Model Musculoskeletal Pathologies and Accelerate Therapeutic Discovery. Front. Bioeng. Biotechnol. 2022, 10, 846230. [Google Scholar] [CrossRef]
  171. Ingber, D.E. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat. Rev. Genet. 2022. [Google Scholar] [CrossRef] [PubMed]
  172. Parrotta, E.I.; Lucchino, V.; Scaramuzzino, L.; Scalise, S.; Cuda, G. Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. Int. J. Mol. Sci. 2020, 21, 4354. [Google Scholar] [CrossRef] [PubMed]
  173. Hinson, J.T.; Chopra, A.; Nafissi, N.; Polacheck, W.J.; Benson, C.C.; Swist, S.; Gorham, J.; Yang, L.; Schafer, S.; Sheng, C.C.; et al. HEART DISEASE. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 2015, 349, 982–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Stillitano, F.; Turnbull, I.C.; Karakikes, I.; Nonnenmacher, M.; Backeris, P.; Hulot, J.S.; Kranias, E.G.; Hajjar, R.J.; Costa, K.D. Genomic correction of familial cardiomyopathy in human engineered cardiac tissues. Eur. Heart J. 2016, 37, 3282–3284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Prondzynski, M.; Lemoine, M.D.; Zech, A.T.; Horvath, A.; Di Mauro, V.; Koivumaki, J.T.; Kresin, N.; Busch, J.; Krause, T.; Kramer, E.; et al. Disease modeling of a mutation in alpha-actinin 2 guides clinical therapy in hypertrophic cardiomyopathy. EMBO Mol. Med. 2019, 11, e11115. [Google Scholar] [CrossRef]
  176. Richards, D.J.; Li, Y.; Kerr, C.M.; Yao, J.; Beeson, G.C.; Coyle, R.C.; Chen, X.; Jia, J.; Damon, B.; Wilson, R.; et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat. Biomed. Eng. 2020, 4, 446–462. [Google Scholar] [CrossRef]
  177. Orita, K.; Sawada, K.; Matsumoto, N.; Ikegaya, Y. Machine-learning-based quality control of contractility of cultured human-induced pluripotent stem-cell-derived cardiomyocytes. Biochem. Biophys. Res. Commun. 2020, 526, 751–755. [Google Scholar] [CrossRef]
  178. Orita, K.; Sawada, K.; Koyama, R.; Ikegaya, Y. Deep learning-based quality control of cultured human-induced pluripotent stem cell-derived cardiomyocytes. J. Pharmacol. Sci. 2019, 140, 313–316. [Google Scholar] [CrossRef]
  179. Coronnello, C.; Francipane, M.G. Moving Towards Induced Pluripotent Stem Cell-based Therapies with Artificial Intelligence and Machine Learning. Stem Cell Rev. Rep. 2022, 18, 559–569. [Google Scholar] [CrossRef]
  180. Santulli, G.; Lewis, D.R.; Marks, A.R. Physiology and pathophysiology of excitation-contraction coupling: The functional role of ryanodine receptor. J. Muscle Res. Cell Motil. 2017, 38, 37–45. [Google Scholar] [CrossRef] [Green Version]
  181. Juhola, M.; Penttinen, K.; Joutsijoki, H.; Varpa, K.; Saarikoski, J.; Rasku, J.; Siirtola, H.; Iltanen, K.; Laurikkala, J.; Hyyro, H.; et al. Signal analysis and classification methods for the calcium transient data of stem cell-derived cardiomyocytes. Comput. Biol. Med. 2015, 61, 1–7. [Google Scholar] [CrossRef] [PubMed]
  182. Juhola, M.; Joutsijoki, H.; Penttinen, K.; Aalto-Setala, K. Detection of genetic cardiac diseases by Ca(2+) transient profiles using machine learning methods. Sci. Rep. 2018, 8, 9355. [Google Scholar] [CrossRef] [PubMed]
  183. Joutsijoki, H.; Penttinen, K.; Juhola, M.; Aalto-Setala, K. Separation of HCM and LQT Cardiac Diseases with Machine Learning of Ca2+ Transient Profiles. Methods Inf. Med. 2019, 58, 167–178. [Google Scholar] [CrossRef] [PubMed]
  184. Joutsijoki, H.; Haponen, M.; Rasku, J.; Aalto-Setala, K.; Juhola, M. Machine Learning Approach to Automated Quality Identification of Human Induced Pluripotent Stem Cell Colony Images. Comput. Math Methods Med. 2016, 2016, 3091039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Juhola, M.; Penttinen, K.; Joutsijoki, H.; Aalto-Setala, K. Analysis of Drug Effects on iPSC Cardiomyocytes with Machine Learning. Ann. Biomed. Eng. 2021, 49, 129–138. [Google Scholar] [CrossRef]
  186. Aghasafari, P.; Yang, P.C.; Kernik, D.C.; Sakamoto, K.; Kanda, Y.; Kurokawa, J.; Vorobyov, I.; Clancy, C.E. A deep learning algorithm to translate and classify cardiac electrophysiology. eLife 2021, 10, e68335. [Google Scholar] [CrossRef]
Figure 1. Tissue engineering approaches to create in vitro 3D cardiac tissue derived from hiPSCs.
Figure 1. Tissue engineering approaches to create in vitro 3D cardiac tissue derived from hiPSCs.
Bioengineering 09 00168 g001
Figure 2. Strategies to increase maturation of hiPSC-CMs and their applications in basic research and in the clinical scenario. Electrical, mechanical, and biochemical factors, alongside genetic modifications and approaches aiming at adjusting substrate stiffness can be harnessed to enhance the maturation of hiPSC-CMs. Adult-like hiPSC-CMs are characterized by morphological, structural, genetic, and electrophysiological modifications.
Figure 2. Strategies to increase maturation of hiPSC-CMs and their applications in basic research and in the clinical scenario. Electrical, mechanical, and biochemical factors, alongside genetic modifications and approaches aiming at adjusting substrate stiffness can be harnessed to enhance the maturation of hiPSC-CMs. Adult-like hiPSC-CMs are characterized by morphological, structural, genetic, and electrophysiological modifications.
Bioengineering 09 00168 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Varzideh, F.; Mone, P.; Santulli, G. Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells. Bioengineering 2022, 9, 168. https://doi.org/10.3390/bioengineering9040168

AMA Style

Varzideh F, Mone P, Santulli G. Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells. Bioengineering. 2022; 9(4):168. https://doi.org/10.3390/bioengineering9040168

Chicago/Turabian Style

Varzideh, Fahimeh, Pasquale Mone, and Gaetano Santulli. 2022. "Bioengineering Strategies to Create 3D Cardiac Constructs from Human Induced Pluripotent Stem Cells" Bioengineering 9, no. 4: 168. https://doi.org/10.3390/bioengineering9040168

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop