1
|
Sleiman Y, Reisqs JB, Boutjdir M. Differentiation of Sinoatrial-like Cardiomyocytes as a Biological Pacemaker Model. Int J Mol Sci 2024; 25:9155. [PMID: 39273104 PMCID: PMC11394733 DOI: 10.3390/ijms25179155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are widely used for disease modeling and pharmacological screening. However, their application has mainly focused on inherited cardiopathies affecting ventricular cardiomyocytes, leading to extensive knowledge on generating ventricular-like hiPSC-CMs. Electronic pacemakers, despite their utility, have significant disadvantages, including lack of hormonal responsiveness, infection risk, limited battery life, and inability to adapt to changes in heart size. Therefore, developing an in vitro multiscale model of the human sinoatrial node (SAN) pacemaker using hiPSC-CM and SAN-like cardiomyocyte differentiation protocols is essential. This would enhance the understanding of SAN-related pathologies and support targeted therapies. Generating SAN-like cardiomyocytes offers the potential for biological pacemakers and specialized conduction tissues, promising significant benefits for patients with conduction system defects. This review focuses on arrythmias related to pacemaker dysfunction, examining protocols' advantages and drawbacks for generating SAN-like cardiomyocytes from hESCs/hiPSCs, and discussing therapeutic approaches involving their engraftment in animal models.
Collapse
Affiliation(s)
- Yvonne Sleiman
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
| | - Jean-Baptiste Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, New York, NY 11203, USA
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
2
|
Wulkan F, Romagnuolo R, Qiang B, Valdman Sadikov T, Kim KP, Quesnel E, Jiang W, Andharia N, Weyers JJ, Ghugre NR, Ozcan B, Alibhai FJ, Laflamme MA. Stem cell-derived cardiomyocytes expressing a dominant negative pacemaker HCN4 channel do not reduce the risk of graft-related arrhythmias. Front Cardiovasc Med 2024; 11:1374881. [PMID: 39045008 PMCID: PMC11263024 DOI: 10.3389/fcvm.2024.1374881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Background Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) show tremendous promise for cardiac regeneration following myocardial infarction (MI), but their transplantation gives rise to transient ventricular tachycardia (VT) in large-animal MI models, representing a major hurdle to translation. Our group previously reported that these arrhythmias arise from a focal mechanism whereby graft tissue functions as an ectopic pacemaker; therefore, we hypothesized that hPSC-CMs engineered with a dominant negative form of the pacemaker ion channel HCN4 (dnHCN4) would exhibit reduced automaticity and arrhythmogenic risk following transplantation. Methods We used CRISPR/Cas9-mediated gene-editing to create transgenic dnHCN4 hPSC-CMs, and their electrophysiological behavior was evaluated in vitro by patch-clamp recordings and optical mapping. Next, we transplanted WT and homozygous dnHCN4 hPSC-CMs in a pig MI model and compared post-transplantation outcomes including the incidence of spontaneous arrhythmias and graft structure by immunohistochemistry. Results In vitro dnHCN4 hPSC-CMs exhibited significantly reduced automaticity and pacemaker funny current (I f ) density relative to wildtype (WT) cardiomyocytes. Following transplantation with either dnHCN4 or WT hPSC-CMs, all recipient hearts showed transmural infarct scar that was partially remuscularized by scattered islands of human myocardium. However, in contrast to our hypothesis, both dnHCN4 and WT hPSC-CM recipients exhibited frequent episodes of ventricular tachycardia (VT). Conclusions While genetic silencing of the pacemaker ion channel HCN4 suppresses the automaticity of hPSC-CMs in vitro, this intervention is insufficient to reduce VT risk post-transplantation in the pig MI model, implying more complex mechanism(s) are operational in vivo.
Collapse
Affiliation(s)
- Fanny Wulkan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Beiping Qiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | | | | | - Elya Quesnel
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Wenlei Jiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Naaz Andharia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Jill J. Weyers
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nilesh R. Ghugre
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Bilgehan Ozcan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J. Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Nguyen T, Rosa-Garrido M, Sadek H, Garry DJ, Zhang JJ. Promoting cardiomyocyte proliferation for myocardial regeneration in large mammals. J Mol Cell Cardiol 2024; 188:52-60. [PMID: 38340541 PMCID: PMC11018144 DOI: 10.1016/j.yjmcc.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/29/2023] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
From molecular and cellular perspectives, heart failure is caused by the loss of cardiomyocytes-the fundamental contractile units of the heart. Because mammalian cardiomyocytes exit the cell cycle shortly after birth, the cardiomyocyte damage induced by myocardial infarction (MI) typically leads to dilatation of the left ventricle (LV) and often progresses to heart failure. However, recent findings indicate that the hearts of neonatal pigs completely regenerated the cardiomyocytes that were lost to MI when the injury occurred on postnatal day 1 (P1). This recovery was accompanied by increases in the expression of markers for cell-cycle activity in cardiomyocytes. These results suggest that the repair process was driven by cardiomyocyte proliferation. This review summarizes findings from recent studies that found evidence of cardiomyocyte proliferation in 1) the uninjured hearts of newborn pigs on P1, 2) neonatal pig hearts after myocardial injury on P1, and 3) the hearts of pigs that underwent apical resection surgery (AR) on P1 followed by MI on postnatal day 28 (P28). Analyses of cardiomyocyte single-nucleus RNA sequencing data collected from the hearts of animals in these three experimental groups, their corresponding control groups, and fetal pigs suggested that although the check-point regulators and other molecules that direct cardiomyocyte cell-cycle progression and proliferation in fetal, newborn, and postnatal pigs were identical, the mechanisms that activated cardiomyocyte proliferation in response to injury may differ from those that regulate cardiomyocyte proliferation during development.
Collapse
Affiliation(s)
- Thanh Nguyen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Hesham Sadek
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel J Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jianyi Jay Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA; Department of Medicine, Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
4
|
Butler D, Reyes DR. Heart-on-a-chip systems: disease modeling and drug screening applications. LAB ON A CHIP 2024; 24:1494-1528. [PMID: 38318723 DOI: 10.1039/d3lc00829k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, casting a substantial economic footprint and burdening the global healthcare system. Historically, pre-clinical CVD modeling and therapeutic screening have been performed using animal models. Unfortunately, animal models oftentimes fail to adequately mimic human physiology, leading to a poor translation of therapeutics from pre-clinical trials to consumers. Even those that make it to market can be removed due to unforeseen side effects. As such, there exists a clinical, technological, and economical need for systems that faithfully capture human (patho)physiology for modeling CVD, assessing cardiotoxicity, and evaluating drug efficacy. Heart-on-a-chip (HoC) systems are a part of the broader organ-on-a-chip paradigm that leverages microfluidics, tissue engineering, microfabrication, electronics, and gene editing to create human-relevant models for studying disease, drug-induced side effects, and therapeutic efficacy. These compact systems can be capable of real-time measurements and on-demand characterization of tissue behavior and could revolutionize the drug development process. In this review, we highlight the key components that comprise a HoC system followed by a review of contemporary reports of their use in disease modeling, drug toxicity and efficacy assessment, and as part of multi-organ-on-a-chip platforms. We also discuss future perspectives and challenges facing the field, including a discussion on the role that standardization is expected to play in accelerating the widespread adoption of these platforms.
Collapse
Affiliation(s)
- Derrick Butler
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| | - Darwin R Reyes
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| |
Collapse
|
5
|
Lin Z, Garbern JC, Liu R, Li Q, Mancheño Juncosa E, Elwell HL, Sokol M, Aoyama J, Deumer US, Hsiao E, Sheng H, Lee RT, Liu J. Tissue-embedded stretchable nanoelectronics reveal endothelial cell-mediated electrical maturation of human 3D cardiac microtissues. SCIENCE ADVANCES 2023; 9:eade8513. [PMID: 36888704 PMCID: PMC9995081 DOI: 10.1126/sciadv.ade8513] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Clinical translation of stem cell therapies for heart disease requires electrical integration of transplanted cardiomyocytes. Generation of electrically matured human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) is critical for electrical integration. Here, we found that hiPSC-derived endothelial cells (hiPSC-ECs) promoted the expression of selected maturation markers in hiPSC-CMs. Using tissue-embedded stretchable mesh nanoelectronics, we achieved a long-term stable map of human three-dimensional (3D) cardiac microtissue electrical activity. The results revealed that hiPSC-ECs accelerated the electrical maturation of hiPSC-CMs in 3D cardiac microtissues. Machine learning-based pseudotime trajectory inference of cardiomyocyte electrical signals further revealed the electrical phenotypic transition path during development. Guided by the electrical recording data, single-cell RNA sequencing identified that hiPSC-ECs promoted cardiomyocyte subpopulations with a more mature phenotype, and multiple ligand-receptor interactions were up-regulated between hiPSC-ECs and hiPSC-CMs, revealing a coordinated multifactorial mechanism of hiPSC-CM electrical maturation. Collectively, these findings show that hiPSC-ECs drive hiPSC-CM electrical maturation via multiple intercellular pathways.
Collapse
Affiliation(s)
- Zuwan Lin
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Department of Cardiology, Boston Children’s Hospital, Boston, MA, USA
| | - Ren Liu
- School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Qiang Li
- School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | | | - Hannah L.T. Elwell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Morgan Sokol
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Junya Aoyama
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Undine-Sophie Deumer
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Emma Hsiao
- School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Hao Sheng
- School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Corresponding author. (J.L.), (R.T.L.)
| | - Jia Liu
- School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Corresponding author. (J.L.), (R.T.L.)
| |
Collapse
|
6
|
Pan Z, Liang P. Human-Induced Pluripotent Stem Cell-Based Differentiation of Cardiomyocyte Subtypes for Drug Discovery and Cell Therapy. Handb Exp Pharmacol 2023; 281:209-233. [PMID: 37421443 DOI: 10.1007/164_2023_663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Drug attrition rates have increased over the past few years, accompanied with growing costs for the pharmaceutical industry and consumers. Lack of in vitro models connecting the results of toxicity screening assays with clinical outcomes accounts for this high attrition rate. The emergence of cardiomyocytes derived from human pluripotent stem cells provides an amenable source of cells for disease modeling, drug discovery, and cardiotoxicity screening. Functionally similar to to embryonic stem cells, but with fewer ethical concerns, induced pluripotent stem cells (iPSCs) can recapitulate patient-specific genetic backgrounds, which would be a huge revolution for personalized medicine. The generated iPSC-derived cardiomyocytes (iPSC-CMs) represent different subtypes including ventricular-, atrial-, and nodal-like cardiomyocytes. Purifying these subtypes for chamber-specific drug screening presents opportunities and challenges. In this chapter, we discuss the strategies for the purification of iPSC-CMs, the use of iPSC-CMs for drug discovery and cardiotoxicity test, and the current limitations of iPSC-CMs that should be overcome for wider and more precise cardiovascular applications.
Collapse
Affiliation(s)
- Ziwei Pan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
7
|
Brogi S, Tabanelli R, Calderone V. Combinatorial approaches for novel cardiovascular drug discovery: a review of the literature. Expert Opin Drug Discov 2022; 17:1111-1129. [PMID: 35853260 DOI: 10.1080/17460441.2022.2104247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION In this article, authors report an inclusive discussion about the combinatorial approach for the treatment of cardiovascular diseases (CVDs) and for counteracting the cardiovascular risk factors. The mentioned strategy was demonstrated to be useful for improving the efficacy of pharmacological treatments and in CVDs showed superior efficacy with respect to the classical monotherapeutic approach. AREAS COVERED According to this topic, authors analyzed the combinatorial treatments that are available on the market, highlighting clinical studies that demonstrated the efficacy of combinatorial drug strategies to cure CVDs and related risk factors. Furthermore, the review gives an outlook on the future perspective of this therapeutic option, highlighting novel drug targets and disease models that could help the future cardiovascular drug discovery. EXPERT OPINION The use of specifically designed and increasingly rational and effective drug combination therapies can therefore be considered the evolution of polypharmacy in cardiometabolic and CVDs. This approach can allow to intervene on multiple etiopathogenetic mechanisms of the disease or to act simultaneously on different pathologies/risk factors, using the combinations most suitable from a pharmacodynamic, pharmacokinetic, and toxicological perspective, thus finding the most appropriate therapeutic option.
Collapse
Affiliation(s)
- Simone Brogi
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
8
|
Wang Y, Wei J, Zhang P, Zhang X, Wang Y, Chen W, Zhao Y, Cui X. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022; 13:945206. [PMID: 36120374 PMCID: PMC9471952 DOI: 10.3389/fphar.2022.945206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
NRG1 (Neuregulin-1) is an effective cardiomyocyte proliferator, secreted and released by endothelial vascular cells, and affects the cardiovascular system. It plays a major role in heart growth, proliferation, differentiation, apoptosis, and other cardiovascular processes. Numerous experiments have shown that NRG1 can repair the heart in the pathophysiology of atherosclerosis, myocardial infarction, ischemia reperfusion, heart failure, cardiomyopathy and other cardiovascular diseases. NRG1 can connect related signaling pathways through the NRG1/ErbB pathway, which form signal cascades to improve the myocardial microenvironment, such as regulating cardiac inflammation, oxidative stress, necrotic apoptosis. Here, we summarize recent research advances on the molecular mechanisms of NRG1, elucidate the contribution of NRG1 to cardiovascular disease, discuss therapeutic approaches targeting NRG1 associated with cardiovascular disease, and highlight areas for future research.
Collapse
Affiliation(s)
- Yan Wang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianliang Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peng Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifei Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanan Zhao
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| |
Collapse
|
9
|
Yu D, Zhu D, Wang X, Li B, Li J, Lu P, Ji Y, Wang X. Prenatal di-(2-ethylhexyl) phthalate exposure induced myocardial cytotoxicity via the regulation of the NRG1-dependent ErbB2/ErbB4-PI3K/AKT signaling pathway in fetal mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113771. [PMID: 35716406 DOI: 10.1016/j.ecoenv.2022.113771] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
Environmental sanitation of maternal contact during pregnancy is extremely important for the development of different fetal tissues and organs. In particular, during early pregnancy, any adverse exposure may cause abnormal fetal growth or inhibit the development of embryogenic organs. The potential risks of phthalate exposure, which affects the development of humans and animals, are becoming a serious concern worldwide. However, the specific molecular mechanism of di-(2-ethylhexyl) phthalate (DEHP)-induced cardiotoxicity in fetal mice remains unclear. In this study, animal models of DEHP gavage at concentrations of 250, 500, and 1000 mg/kg/day within 8.5-18.5 days of pregnancy were established. The cell proliferation, survival, and apoptosis rates were evaluated using CCK8, EdU, TUNEL and flow cytometry. The molecular mechanism was assessed via transcriptome sequencing, immunohistochemistry, immunofluorescence, reverse transcription-quantitative polymerase chain reaction, and Western blot analysis. In vivo, DEHP increased apoptosis, decreased Ki67 and CD31 expression, reduced heart weight and area, slowed down myocardial sarcomere development, and caused cardiac septal defect in fetal mice heart. Transcriptome sequencing showed that DEHP decreased NRG1 expression and downregulated the ErbB2/ErbB4-PI3K/AKT signaling pathway-related target genes. In vitro, primary cardiomyocytes were cultured with DEHP at a concentration of 150 μg/mL combined with ErbB inhibitor (AG1478, 10 μmol/L) and/or NRG1 protein (100 ng/mL) for 72 h. After DEHP intervention, the expression of NRG1 and the phosphorylation level of ErbB2, ErbB4, PI3K, and AKT decreased, and the apoptosis-related protein levels increased. Moreover, the apoptosis rate increased. After adding exogenous NRG1, the phosphorylation level of the NRG1/ERbB2/ERbB4-PI3K/AKT pathway increased, and the apoptosis-related protein levels decreased. Further, the apoptosis rate reduced. Interestingly, after exposure to DEHP and AG1478 + NRG1, the anti-apoptotic effect of NRG1 and cardiomyocyte proliferation decreased by inhibiting the NRG1/ERbB2/ERbB4-PI3K/AKT pathway. Hence, the NRG1-dependent regulation of the ERbB2/ERbB4-PI3K/AKT signaling pathway may be a key mechanism of DEHP-induced myocardial cytotoxicity.
Collapse
Affiliation(s)
- Dongmin Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Dawei Zhu
- Department of Cardiothoracic Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211000, China
| | - Xufeng Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Ben Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jinghang Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Peng Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yumeng Ji
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
10
|
Nguyen T, Wei Y, Nakada Y, Zhou Y, Zhang J. Cardiomyocyte Cell-Cycle Regulation in Neonatal Large Mammals: Single Nucleus RNA-Sequencing Data Analysis via an Artificial-Intelligence–Based Pipeline. Front Bioeng Biotechnol 2022; 10:914450. [PMID: 35860330 PMCID: PMC9289371 DOI: 10.3389/fbioe.2022.914450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/18/2022] [Indexed: 11/20/2022] Open
Abstract
Adult mammalian cardiomyocytes have very limited capacity to proliferate and repair the myocardial infarction. However, when apical resection (AR) was performed in pig hearts on postnatal day (P) 1 (ARP1) and acute myocardial infarction (MI) was induced on P28 (MIP28), the animals recovered with no evidence of myocardial scarring or decline in contractile performance. Furthermore, the repair process appeared to be driven by cardiomyocyte proliferation, but the regulatory molecules that govern the ARP1-induced enhancement of myocardial recovery remain unclear. Single-nucleus RNA sequencing (snRNA-seq) data collected from fetal pig hearts and the hearts of pigs that underwent ARP1, MIP28, both ARP1 and MI, or neither myocardial injury were evaluated via autoencoder, cluster analysis, sparse learning, and semisupervised learning. Ten clusters of cardiomyocytes (CM1–CM10) were identified across all experimental groups and time points. CM1 was only observed in ARP1 hearts on P28 and was enriched for the expression of T-box transcription factors 5 and 20 (TBX5 and TBX20, respectively), Erb-B2 receptor tyrosine kinase 4 (ERBB4), and G Protein-Coupled Receptor Kinase 5 (GRK5), as well as genes associated with the proliferation and growth of cardiac muscle. CM1 cardiomyocytes also highly expressed genes for glycolysis while lowly expressed genes for adrenergic signaling, which suggested that CM1 were immature cardiomyocytes. Thus, we have identified a cluster of cardiomyocytes, CM1, in neonatal pig hearts that appeared to be generated in response to AR injury on P1 and may have been primed for activation of CM cell-cycle activation and proliferation by the upregulation of TBX5, TBX20, ERBB4, and GRK5.
Collapse
Affiliation(s)
- Thanh Nguyen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Wei
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuji Nakada
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yang Zhou
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Jianyi Zhang,
| |
Collapse
|
11
|
Abstract
An ensemble of in vitro cardiac tissue models has been developed over the past several decades to aid our understanding of complex cardiovascular disorders using a reductionist approach. These approaches often rely on recapitulating single or multiple clinically relevant end points in a dish indicative of the cardiac pathophysiology. The possibility to generate disease-relevant and patient-specific human induced pluripotent stem cells has further leveraged the utility of the cardiac models as screening tools at a large scale. To elucidate biological mechanisms in the cardiac models, it is critical to integrate physiological cues in form of biochemical, biophysical, and electromechanical stimuli to achieve desired tissue-like maturity for a robust phenotyping. Here, we review the latest advances in the directed stem cell differentiation approaches to derive a wide gamut of cardiovascular cell types, to allow customization in cardiac model systems, and to study diseased states in multiple cell types. We also highlight the recent progress in the development of several cardiovascular models, such as cardiac organoids, microtissues, engineered heart tissues, and microphysiological systems. We further expand our discussion on defining the context of use for the selection of currently available cardiac tissue models. Last, we discuss the limitations and challenges with the current state-of-the-art cardiac models and highlight future directions.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Suji Choi
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
| | - Christina Alamana
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kevin K. Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston MA 02134
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
12
|
Jiang CL, Goyal Y, Jain N, Wang Q, Truitt RE, Coté AJ, Emert B, Mellis IA, Kiani K, Yang W, Jain R, Raj A. Cell type determination for cardiac differentiation occurs soon after seeding of human-induced pluripotent stem cells. Genome Biol 2022; 23:90. [PMID: 35382863 PMCID: PMC8985385 DOI: 10.1186/s13059-022-02654-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 03/16/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cardiac differentiation of human-induced pluripotent stem (hiPS) cells consistently produces a mixed population of cardiomyocytes and non-cardiac cell types, even when using well-characterized protocols. We sought to determine whether different cell types might result from intrinsic differences in hiPS cells prior to the onset of differentiation. RESULTS By associating individual differentiated cells that share a common hiPS cell precursor, we tested whether expression variability is predetermined from the hiPS cell state. In a single experiment, cells that shared a progenitor were more transcriptionally similar to each other than to other cells in the differentiated population. However, when the same hiPS cells were differentiated in parallel, we did not observe high transcriptional similarity across differentiations. Additionally, we found that substantial cell death occurs during differentiation in a manner that suggested all cells were equally likely to survive or die, suggesting that there is no intrinsic selection bias for cells descended from particular hiPS cell progenitors. We thus wondered how cells grow spatially during differentiation, so we labeled cells by expression of marker genes and found that cells expressing the same marker tended to occur in patches. Our results suggest that cell type determination across multiple cell types, once initiated, is maintained in a cell-autonomous manner for multiple divisions. CONCLUSIONS Altogether, our results show that while substantial heterogeneity exists in the initial hiPS cell population, it is not responsible for the variability observed in differentiated outcomes; instead, factors specifying the various cell types likely act during a window that begins shortly after the seeding of hiPS cells for differentiation.
Collapse
Affiliation(s)
- Connie L Jiang
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogesh Goyal
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Naveen Jain
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qiaohong Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel E Truitt
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Allison J Coté
- Cell Biology, Physiology, and Metabolism, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Emert
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian A Mellis
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karun Kiani
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenli Yang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rajan Jain
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Ghazizadeh Z, Zhu J, Fattahi F, Tang A, Sun X, Amin S, Tsai SY, Khalaj M, Zhou T, Samuel RM, Zhang T, Ortega FA, Gordillo M, Moroziewicz D, Paull D, Noggle SA, Xiang JZ, Studer L, Christini DJ, Pitt GS, Evans T, Chen S. A dual SHOX2:GFP; MYH6:mCherry knockin hESC reporter line for derivation of human SAN-like cells. iScience 2022; 25:104153. [PMID: 35434558 PMCID: PMC9010642 DOI: 10.1016/j.isci.2022.104153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/25/2022] [Accepted: 03/22/2022] [Indexed: 01/19/2023] Open
Abstract
The sinoatrial node (SAN) is the primary pacemaker of the heart. The human SAN is poorly understood due to limited primary tissue access and limitations in robust in vitro derivation methods. We developed a dual SHOX2:GFP; MYH6:mCherry knockin human embryonic stem cell (hESC) reporter line, which allows the identification and purification of SAN-like cells. Using this line, we performed several rounds of chemical screens and developed an efficient strategy to generate and purify hESC-derived SAN-like cells (hESC-SAN). The derived hESC-SAN cells display molecular and electrophysiological characteristics of bona fide nodal cells, which allowed exploration of their transcriptional profile at single-cell level. In sum, our dual reporter system facilitated an effective strategy for deriving human SAN-like cells, which can potentially be used for future disease modeling and drug discovery.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA,Corresponding author
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Faranak Fattahi
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alice Tang
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xiaolu Sun
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sadaf Amin
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Su-Yi Tsai
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Mona Khalaj
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ting Zhou
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ryan M. Samuel
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medical College, New York, NY 10065, USA
| | - Francis A. Ortega
- Physiology, Biophysics, and Systems Biology Graduate Program, Weill Cornell Medical College, New York, NY 10065, USA,Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Miriam Gordillo
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019, USA
| | | | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019, USA
| | - Scott A. Noggle
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019, USA
| | - Jenny Zhaoying Xiang
- Genomic Resource Core Facility, Weill Cornell Medical College, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David J. Christini
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA,Corresponding author
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA,Corresponding author
| |
Collapse
|
14
|
Mitrečić D, Hribljan V, Jagečić D, Isaković J, Lamberto F, Horánszky A, Zana M, Foldes G, Zavan B, Pivoriūnas A, Martinez S, Mazzini L, Radenovic L, Milasin J, Chachques JC, Buzanska L, Song MS, Dinnyés A. Regenerative Neurology and Regenerative Cardiology: Shared Hurdles and Achievements. Int J Mol Sci 2022; 23:855. [PMID: 35055039 PMCID: PMC8776151 DOI: 10.3390/ijms23020855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/24/2021] [Accepted: 01/09/2022] [Indexed: 02/05/2023] Open
Abstract
From the first success in cultivation of cells in vitro, it became clear that developing cell and/or tissue specific cultures would open a myriad of new opportunities for medical research. Expertise in various in vitro models has been developing over decades, so nowadays we benefit from highly specific in vitro systems imitating every organ of the human body. Moreover, obtaining sufficient number of standardized cells allows for cell transplantation approach with the goal of improving the regeneration of injured/disease affected tissue. However, different cell types bring different needs and place various types of hurdles on the path of regenerative neurology and regenerative cardiology. In this review, written by European experts gathered in Cost European action dedicated to neurology and cardiology-Bioneca, we present the experience acquired by working on two rather different organs: the brain and the heart. When taken into account that diseases of these two organs, mostly ischemic in their nature (stroke and heart infarction), bring by far the largest burden of the medical systems around Europe, it is not surprising that in vitro models of nervous and heart muscle tissue were in the focus of biomedical research in the last decades. In this review we describe and discuss hurdles which still impair further progress of regenerative neurology and cardiology and we detect those ones which are common to both fields and some, which are field-specific. With the goal to elucidate strategies which might be shared between regenerative neurology and cardiology we discuss methodological solutions which can help each of the fields to accelerate their development.
Collapse
Affiliation(s)
- Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Valentina Hribljan
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Denis Jagečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | | | - Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, 2100 Godollo, Hungary
| | - Alex Horánszky
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, 2100 Godollo, Hungary
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
| | - Gabor Foldes
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Salvador Martinez
- Instituto de Neurociencias UMH-CSIC, 03550 San Juan de Alicante, Spain
| | - Letizia Mazzini
- ALS Center, Department of Neurology, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy
| | - Lidija Radenovic
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Milasin
- Laboratory for Stem Cell Research, School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Juan Carlos Chachques
- Laboratory of Biosurgical Research, Pompidou Hospital, University of Paris, 75006 Paris, France
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Min Suk Song
- Omnion Research International Ltd., 10000 Zagreb, Croatia
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Gordillo, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, 2100 Godollo, Hungary
- HCEMM-USZ Stem Cell Research Group, Department of Cell Biology and Molecular Medicine, University of Szeged, 6720 Szeged, Hungary
- College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
15
|
Dastidar S, Majumdar D, Tipanee J, Singh K, Klein AF, Furling D, Chuah MK, VandenDriessche T. Comprehensive transcriptome-wide analysis of spliceopathy correction of myotonic dystrophy using CRISPR-Cas9 in iPSCs-derived cardiomyocytes. Mol Ther 2022; 30:75-91. [PMID: 34371182 PMCID: PMC8753376 DOI: 10.1016/j.ymthe.2021.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 07/01/2021] [Accepted: 07/26/2021] [Indexed: 01/07/2023] Open
Abstract
CTG repeat expansion (CTGexp) is associated with aberrant alternate splicing that contributes to cardiac dysfunction in myotonic dystrophy type 1 (DM1). Excision of this CTGexp repeat using CRISPR-Cas resulted in the disappearance of punctate ribonuclear foci in cardiomyocyte-like cells derived from DM1-induced pluripotent stem cells (iPSCs). This was associated with correction of the underlying spliceopathy as determined by RNA sequencing and alternate splicing analysis. Certain genes were of particular interest due to their role in cardiac development, maturation, and function (TPM4, CYP2J2, DMD, MBNL3, CACNA1H, ROCK2, ACTB) or their association with splicing (SMN2, GCFC2, MBNL3). Moreover, while comparing isogenic CRISPR-Cas9-corrected versus non-corrected DM1 cardiomyocytes, a prominent difference in the splicing pattern for a number of candidate genes was apparent pertaining to genes that are associated with cardiac function (TNNT, TNNT2, TTN, TPM1, SYNE1, CACNA1A, MTMR1, NEBL, TPM1), cellular signaling (NCOR2, CLIP1, LRRFIP2, CLASP1, CAMK2G), and other DM1-related genes (i.e., NUMA1, MBNL2, LDB3) in addition to the disease-causing DMPK gene itself. Subsequent validation using a selected gene subset, including MBNL1, MBNL2, INSR, ADD3, and CRTC2, further confirmed correction of the spliceopathy following CTGexp repeat excision. To our knowledge, the present study provides the first comprehensive unbiased transcriptome-wide analysis of the differential splicing landscape in DM1 patient-derived cardiac cells after excision of the CTGexp repeat using CRISPR-Cas9, showing reversal of the abnormal cardiac spliceopathy in DM1.
Collapse
Affiliation(s)
- Sumitava Dastidar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Debanjana Majumdar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Jaitip Tipanee
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Kshitiz Singh
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Arnaud F. Klein
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Denis Furling
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marinee K. Chuah
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium,Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium,Corresponding author: Marinee K. Chuah, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium,Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium,Corresponding author: Thierry VandenDriessche, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| |
Collapse
|
16
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
17
|
Martínez-Falguera D, Iborra-Egea O, Gálvez-Montón C. iPSC Therapy for Myocardial Infarction in Large Animal Models: Land of Hope and Dreams. Biomedicines 2021; 9:1836. [PMID: 34944652 PMCID: PMC8698445 DOI: 10.3390/biomedicines9121836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction is the main driver of heart failure due to ischemia and subsequent cell death, and cell-based strategies have emerged as promising therapeutic methods to replace dead tissue in cardiovascular diseases. Research in this field has been dramatically advanced by the development of laboratory-induced pluripotent stem cells (iPSCs) that harbor the capability to become any cell type. Like other experimental strategies, stem cell therapy must meet multiple requirements before reaching the clinical trial phase, and in vivo models are indispensable for ensuring the safety of such novel therapies. Specifically, translational studies in large animal models are necessary to fully evaluate the therapeutic potential of this approach; to empirically determine the optimal combination of cell types, supplementary factors, and delivery methods to maximize efficacy; and to stringently assess safety. In the present review, we summarize the main strategies employed to generate iPSCs and differentiate them into cardiomyocytes in large animal species; the most critical differences between using small versus large animal models for cardiovascular studies; and the strategies that have been pursued regarding implanted cells' stage of differentiation, origin, and technical application.
Collapse
Affiliation(s)
- Daina Martínez-Falguera
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain;
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| |
Collapse
|
18
|
Mellis IA, Edelstein HI, Truitt R, Goyal Y, Beck LE, Symmons O, Dunagin MC, Linares Saldana RA, Shah PP, Pérez-Bermejo JA, Padmanabhan A, Yang W, Jain R, Raj A. Responsiveness to perturbations is a hallmark of transcription factors that maintain cell identity in vitro. Cell Syst 2021; 12:885-899.e8. [PMID: 34352221 PMCID: PMC8522198 DOI: 10.1016/j.cels.2021.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/27/2020] [Accepted: 07/09/2021] [Indexed: 02/07/2023]
Abstract
Identifying the particular transcription factors that maintain cell type in vitro is important for manipulating cell type. Identifying such transcription factors by their cell-type-specific expression or their involvement in developmental regulation has had limited success. We hypothesized that because cell type is often resilient to perturbations, the transcriptional response to perturbations would reveal identity-maintaining transcription factors. We developed perturbation panel profiling (P3) as a framework for perturbing cells across many conditions and measuring gene expression responsiveness transcriptome-wide. In human iPSC-derived cardiac myocytes, P3 showed that transcription factors important for cardiac myocyte differentiation and maintenance were among the most frequently upregulated (most responsive). We reasoned that one function of responsive genes may be to maintain cellular identity. We identified responsive transcription factors in fibroblasts using P3 and found that suppressing their expression led to enhanced reprogramming. We propose that responsiveness to perturbations is a property of transcription factors that help maintain cellular identity in vitro. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Ian A Mellis
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Genomics and Computational Biology Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hailey I Edelstein
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Truitt
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogesh Goyal
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren E Beck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Orsolya Symmons
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Margaret C Dunagin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ricardo A Linares Saldana
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Parisha P Shah
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Arun Padmanabhan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA; Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Wenli Yang
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rajan Jain
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Arjun Raj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Gao R, Ren J. Zebrafish Models in Therapeutic Research of Cardiac Conduction Disease. Front Cell Dev Biol 2021; 9:731402. [PMID: 34422842 PMCID: PMC8371477 DOI: 10.3389/fcell.2021.731402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/20/2021] [Indexed: 01/04/2023] Open
Abstract
Malfunction in the cardiac conduction system (CCS) due to congenital anomalies or diseases can cause cardiac conduction disease (CCD), which results in disturbances in cardiac rhythm, leading to syncope and even sudden cardiac death. Insights into development of the CCS components, including pacemaker cardiomyocytes (CMs), atrioventricular node (AVN) and the ventricular conduction system (VCS), can shed light on the pathological and molecular mechanisms underlying CCD, provide approaches for generating human pluripotent stem cell (hPSC)-derived CCS cells, and thus improve therapeutic treatment for such a potentially life-threatening disorder of the heart. However, the cellular and molecular mechanisms controlling CCS development remain elusive. The zebrafish has become a valuable vertebrate model to investigate early development of CCS components because of its unique features such as external fertilization, embryonic optical transparency and the ability to survive even with severe cardiovascular defects during development. In this review, we highlight how the zebrafish has been utilized to dissect the cellular and molecular mechanisms of CCS development, and how the evolutionarily conserved developmental mechanisms discovered in zebrafish could be applied to directing the creation of hPSC-derived CCS cells, therefore providing potential therapeutic strategies that may contribute to better treatment for CCD patients.
Collapse
Affiliation(s)
- Rui Gao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - Jie Ren
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
20
|
Chu X, Wang M, Qiu X, Huang Y, Li T, Otieno E, Li N, Luo L, Xiao X. Strategies for constructing pluripotent stem cell- and progenitor cell-derived three-dimensional cardiac micro-tissues. J Biomed Mater Res A 2021; 110:488-503. [PMID: 34397148 DOI: 10.1002/jbm.a.37298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cardiac micro-tissue is a promising model for simulating the structural and functional features of heart in vitro. This scientific achievement provides a platform for exploration about the mechanisms on the development, damage, and regeneration of tissue, hence, paving a way toward development of novel therapies for heart diseases. However, 3D micro-tissue technology is still in its infant stages faced with many challenges such as incompleteness of the tissue microarchitecture, loss of the resident immune cells, poor reproducibility, and deficiencies in continuously feeding the nutrients and removing wastes during micro-tissue culturing. There is an urgent need to optimize the construction of 3D cardiac micro-tissue and improve functions of the involved cells. Therefore, scaffolds and cell resources for building 3D cardiac micro-tissues, strategies for inducing the maturation and functionalization of pluripotent stem cell- or cardiac progenitor cell-derived cardiomyocytes, and the major challenges were reviewed in this writing to enable future fabrication of 3D cardiac micro-tissues or organoids for drug screening, disease modeling, regeneration treatment, and so on.
Collapse
Affiliation(s)
- Xinyue Chu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mingyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China.,Institute of Laboratory Animals Science, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Husbandry Engineering, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yun Huang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Tong Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Edward Otieno
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Na Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Li Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
21
|
Zhang J, Chou OHI, Tse YL, Ng KM, Tse HF. Application of Patient-Specific iPSCs for Modelling and Treatment of X-Linked Cardiomyopathies. Int J Mol Sci 2021; 22:ijms22158132. [PMID: 34360897 PMCID: PMC8347533 DOI: 10.3390/ijms22158132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/11/2022] Open
Abstract
Inherited cardiomyopathies are among the major causes of heart failure and associated with significant mortality and morbidity. Currently, over 70 genes have been linked to the etiology of various forms of cardiomyopathy, some of which are X-linked. Due to the lack of appropriate cell and animal models, it has been difficult to model these X-linked cardiomyopathies. With the advancement of induced pluripotent stem cell (iPSC) technology, the ability to generate iPSC lines from patients with X-linked cardiomyopathy has facilitated in vitro modelling and drug testing for the condition. Nonetheless, due to the mosaicism of the X-chromosome inactivation, disease phenotypes of X-linked cardiomyopathy in heterozygous females are also usually more heterogeneous, with a broad spectrum of presentation. Recent advancements in iPSC procedures have enabled the isolation of cells with different lyonisation to generate isogenic disease and control cell lines. In this review, we will summarise the current strategies and examples of using an iPSC-based model to study different types of X-linked cardiomyopathy. The potential application of isogenic iPSC lines derived from a female patient with heterozygous Danon disease and drug screening will be demonstrated by our preliminary data. The limitations of an iPSC-derived cardiomyocyte-based platform will also be addressed.
Collapse
Affiliation(s)
- Jennifer Zhang
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
| | - Oscar Hou-In Chou
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
| | - Yiu-Lam Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
- Correspondence: (K.-M.N.); (H.-F.T.); Tel.: +852-3917-9955 (K.-M.N.); +852-2255-3598 (H.-F.T.)
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.Z.); (O.H.-I.C.); (Y.-L.T.)
- Centre of Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong, China
- Correspondence: (K.-M.N.); (H.-F.T.); Tel.: +852-3917-9955 (K.-M.N.); +852-2255-3598 (H.-F.T.)
| |
Collapse
|
22
|
Jiang Y, Bao X, Lian XL. A dual cardiomyocyte reporter model derived from human pluripotent stem cells. Stem Cell Res Ther 2021; 12:305. [PMID: 34051863 PMCID: PMC8164304 DOI: 10.1186/s13287-021-02341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 04/19/2021] [Indexed: 11/10/2022] Open
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death in the USA. Cardiomyocytes (CMs) derived from human pluripotent stem cells (hPSCs) provide a valuable cell source for regenerative therapy, disease modeling, and drug screening. Here, we established a hPSC line integrated with a mCherry fluorescent protein driven by the alpha myosin heavy chain (aMHC) promoter, which could be used to purify CMs based on the aMHC promoter activity in these cells. Combined with a fluorescent voltage indicator, ASAP2f, we achieved a dual reporter CM platform, which enables purification and characterization of CM subtypes and holds great potential for disease modeling and drug discovery of CVD.
Collapse
Affiliation(s)
- Yuqian Jiang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
| | - Xiaojun Lance Lian
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA.
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
23
|
Lang D, Glukhov AV. Cellular and Molecular Mechanisms of Functional Hierarchy of Pacemaker Clusters in the Sinoatrial Node: New Insights into Sick Sinus Syndrome. J Cardiovasc Dev Dis 2021; 8:jcdd8040043. [PMID: 33924321 PMCID: PMC8069964 DOI: 10.3390/jcdd8040043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/17/2022] Open
Abstract
The sinoatrial node (SAN), the primary pacemaker of the heart, consists of a heterogeneous population of specialized cardiac myocytes that can spontaneously produce action potentials, generating the rhythm of the heart and coordinating heart contractions. Spontaneous beating can be observed from very early embryonic stage and under a series of genetic programing, the complex heterogeneous SAN cells are formed with specific biomarker proteins and generate robust automaticity. The SAN is capable to adjust its pacemaking rate in response to environmental and autonomic changes to regulate the heart's performance and maintain physiological needs of the body. Importantly, the origin of the action potential in the SAN is not static, but rather dynamically changes according to the prevailing conditions. Changes in the heart rate are associated with a shift of the leading pacemaker location within the SAN and accompanied by alterations in P wave morphology and PQ interval on ECG. Pacemaker shift occurs in response to different interventions: neurohormonal modulation, cardiac glycosides, pharmacological agents, mechanical stretch, a change in temperature, and a change in extracellular electrolyte concentrations. It was linked with the presence of distinct anatomically and functionally defined intranodal pacemaker clusters that are responsible for the generation of the heart rhythm at different rates. Recent studies indicate that on the cellular level, different pacemaker clusters rely on a complex interplay between the calcium (referred to local subsarcolemmal Ca2+ releases generated by the sarcoplasmic reticulum via ryanodine receptors) and voltage (referred to sarcolemmal electrogenic proteins) components of so-called "coupled clock pacemaker system" that is used to describe a complex mechanism of SAN pacemaking. In this review, we examine the structural, functional, and molecular evidence for hierarchical pacemaker clustering within the SAN. We also demonstrate the unique molecular signatures of intranodal pacemaker clusters, highlighting their importance for physiological rhythm regulation as well as their role in the development of SAN dysfunction, also known as sick sinus syndrome.
Collapse
|
24
|
Wang Y, Lei W, Yang J, Ni X, Ye L, Shen Z, Hu S. The updated view on induced pluripotent stem cells for cardiovascular precision medicine. Pflugers Arch 2021; 473:1137-1149. [DOI: 10.1007/s00424-021-02530-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
|
25
|
Krigerts J, Salmina K, Freivalds T, Zayakin P, Rumnieks F, Inashkina I, Giuliani A, Hausmann M, Erenpreisa J. Differentiating cancer cells reveal early large-scale genome regulation by pericentric domains. Biophys J 2021; 120:711-724. [PMID: 33453273 PMCID: PMC7896032 DOI: 10.1016/j.bpj.2021.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Finding out how cells prepare for fate change during differentiation commitment was our task. To address whether the constitutive pericentromere-associated domains (PADs) may be involved, we used a model system with known transcriptome data, MCF-7 breast cancer cells treated with the ErbB3 ligand heregulin (HRG), which induces differentiation and is used in the therapy of cancer. PAD-repressive heterochromatin (H3K9me3), centromere-associated-protein-specific, and active euchromatin (H3K4me3) antibodies, real-time PCR, acridine orange DNA structural test (AOT), and microscopic image analysis were applied. We found a two-step DNA unfolding after 15-20 and 60 min of HRG treatment, respectively. This behavior was consistent with biphasic activation of the early response genes (c-fos - fosL1/myc) and the timing of two transcriptome avalanches reported in the literature. In control, the average number of PADs negatively correlated with their size by scale-free distribution, and centromere clustering in turn correlated with PAD size, both indicating that PADs may create and modulate a suprachromosomal network by fusing and splitting a constant proportion of the constitutive heterochromatin. By 15 min of HRG treatment, the bursting unraveling of PADs from the nucleolus boundary occurred, coinciding with the first step of H3K4me3 chromatin unfolding, confirmed by AOT. The second step after 60 min of HRG treatment was associated with transcription of long noncoding RNA from PADs and peaking of fosL1/c-myc response. We hypothesize that the bursting of PAD clusters under a critical silencing threshold pushes the first transcription avalanche, whereas the destruction of the PAD network enables genome rewiring needed for differentiation repatterning, mediated by early response bivalent genes.
Collapse
Affiliation(s)
- Jekabs Krigerts
- Latvian Biomedicine Research and Study Centre, Riga, Latvia; University of Latvia, Riga, Latvia
| | | | - Talivaldis Freivalds
- Institute of Cardiology and Regenerative Medicine, University of Latvia, Riga, Latvia
| | - Pawel Zayakin
- Latvian Biomedicine Research and Study Centre, Riga, Latvia
| | - Felikss Rumnieks
- Latvian Biomedicine Research and Study Centre, Riga, Latvia; University of Latvia, Riga, Latvia
| | - Inna Inashkina
- Latvian Biomedicine Research and Study Centre, Riga, Latvia
| | - Alessandro Giuliani
- Environment and Health Department, Italian National Institute of Health, Rome, Italy
| | - Michael Hausmann
- Kirchhoff Institute for Physics, Heidelberg University, Heidelberg, Germany.
| | | |
Collapse
|
26
|
Sontayananon N, Redwood C, Davies B, Gehmlich K. Fluorescent PSC-Derived Cardiomyocyte Reporter Lines: Generation Approaches and Their Applications in Cardiovascular Medicine. BIOLOGY 2020; 9:biology9110402. [PMID: 33207727 PMCID: PMC7697758 DOI: 10.3390/biology9110402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
Recent advances have made pluripotent stem cell (PSC)-derived cardiomyocytes an attractive option to model both normal and diseased cardiac function at the single-cell level. However, in vitro differentiation yields heterogeneous populations of cardiomyocytes and other cell types, potentially confounding phenotypic analyses. Fluorescent PSC-derived cardiomyocyte reporter systems allow specific cell lineages to be labelled, facilitating cell isolation for downstream applications including drug testing, disease modelling and cardiac regeneration. In this review, the different genetic strategies used to generate such reporter lines are presented with an emphasis on their relative technical advantages and disadvantages. Next, we explore how the fluorescent reporter lines have provided insights into cardiac development and cardiomyocyte physiology. Finally, we discuss how exciting new approaches using PSC-derived cardiomyocyte reporter lines are contributing to progress in cardiac cell therapy with respect to both graft adaptation and clinical safety.
Collapse
Affiliation(s)
- Naeramit Sontayananon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK; (N.S.); (C.R.)
| | - Charles Redwood
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK; (N.S.); (C.R.)
| | - Benjamin Davies
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
- Correspondence: (B.D.); (K.G.)
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK; (N.S.); (C.R.)
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Correspondence: (B.D.); (K.G.)
| |
Collapse
|
27
|
Fukushima H, Yoshioka M, Kawatou M, López-Dávila V, Takeda M, Kanda Y, Sekino Y, Yoshida Y, Yamashita JK. Specific induction and long-term maintenance of high purity ventricular cardiomyocytes from human induced pluripotent stem cells. PLoS One 2020; 15:e0241287. [PMID: 33137106 PMCID: PMC7605685 DOI: 10.1371/journal.pone.0241287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022] Open
Abstract
Currently, cardiomyocyte (CM) differentiation methods require a purification step after CM induction to ensure the high purity of the cell population. Here we show an improved human CM differentiation protocol with which high-purity ventricular-type CMs can be obtained and maintained without any CM purification process. We induced and collected a mesodermal cell population (platelet-derived growth factor receptor-α (PDGFRα)-positive cells) that can respond to CM differentiation cues, and then stimulated CM differentiation by means of Wnt inhibition. This method reproducibly generated CMs with purities above 95% in several human pluripotent stem cell lines. Furthermore, these CM populations were maintained in culture at such high purity without any further CM purification step for over 200 days. The majority of these CMs (>95%) exhibited a ventricular-like phenotype with a tendency to structural and electrophysiological maturation, including T-tubule-like structure formation and the ability to respond to QT prolongation drugs. This is a simple and valuable method to stably generate CM populations suitable for cardiac toxicology testing, disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Hiroyuki Fukushima
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Miki Yoshioka
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masahide Kawatou
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Víctor López-Dávila
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masafumi Takeda
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yuko Sekino
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Jun K. Yamashita
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
28
|
Yechikov S, Kao HKJ, Chang CW, Pretto D, Zhang XD, Sun YH, Smithers R, Sirish P, Nolta JA, Chan JW, Chiamvimonvat N, Lieu DK. NODAL inhibition promotes differentiation of pacemaker-like cardiomyocytes from human induced pluripotent stem cells. Stem Cell Res 2020; 49:102043. [PMID: 33128951 PMCID: PMC7814970 DOI: 10.1016/j.scr.2020.102043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/17/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
Directed cardiomyogenesis from human induced pluripotent stem cells (hiPSCs) has been greatly improved in the last decade but directed differentiation to pacemaking cardiomyocytes (CMs) remains incompletely understood. In this study, we demonstrated that inhibition of NODAL signaling by a specific NODAL inhibitor (SB431542) in the cardiac mesoderm differentiation stage downregulated PITX2c, a transcription factor that is known to inhibit the formation of the sinoatrial node in the left atrium during cardiac development. The resulting hiPSC-CMs were smaller in cell size, expressed higher pro-pacemaking transcription factors, TBX3 and TBX18, and exhibited pacemaking-like electrophysiological characteristics compared to control hiPSC-CMs differentiated from established Wnt-based protocol. The pacemaker-like subtype increased up to 2.4-fold in hiPSC-CMs differentiated with the addition of SB431542 relative to the control. Hence, Nodal inhibition in the cardiac mesoderm stage promoted pacemaker-like CM differentiation from hiPSCs. Improving the yield of human pacemaker-like CMs is a critical first step in the development of functional human cell-based biopacemakers.
Collapse
Affiliation(s)
- Sergey Yechikov
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Hillary K J Kao
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Che-Wei Chang
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Dalyir Pretto
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Xiao-Dong Zhang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Yao-Hui Sun
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Regan Smithers
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA; Bridges to Stem Cell Research Program, California State University Sacramento, Sacramento, CA 95819, USA
| | - Padmini Sirish
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Jan A Nolta
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA; Department of Hematology and Oncology, University of California, Davis, Sacramento, CA 95817, USA
| | - James W Chan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Deborah K Lieu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
29
|
Ravindran D, Kok C, Farraha M, Selvakumar D, Clayton ZE, Kumar S, Chong J, Kizana E. Gene and Cell Therapy for Cardiac Arrhythmias. Clin Ther 2020; 42:1911-1922. [PMID: 32988632 DOI: 10.1016/j.clinthera.2020.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE In the last decade, interest in gene therapy as a therapeutic technology has increased, largely driven by an exciting yet modest number of successful applications for monogenic diseases. Setbacks in the use of gene therapy for cardiac disease have motivated efforts to develop vectors with enhanced tropism for the heart and more efficient delivery methods. Although monogenic diseases are the logical target, cardiac arrhythmias represent a group of conditions amenable to gene therapy because of focal targets (biological pacemakers, nodal conduction, or stem cell-related arrhythmias) or bystander effects on cells not directly transduced because of electrical coupling. METHODS This review provides a contemporary narrative of the field of gene therapy for experimental cardiac arrhythmias, including those associated with stem cell transplant. Recent articles published in the English language and available through the PubMed database and other prominent literature are discussed. FINDINGS The promise of gene therapy has been realized for a handful of monogenic diseases and is actively being pursued for cardiac applications in preclinical models. With improved vectors, it is likely that cardiac disease will also benefit from this technology. Cardiac arrhythmias, whether inherited or acquired, are a group of conditions with a potentially lower threshold for phenotypic correction and as such hold unique potential as targets for cardiac gene therapy. IMPLICATIONS There has been a proliferation of research on the potential of gene therapy for cardiac arrhythmias. This body of investigation forms a strong basis on which further developments, particularly with viral vectors, are likely to help this technology progress along its translational trajectory.
Collapse
Affiliation(s)
- Dhanya Ravindran
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Cindy Kok
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Melad Farraha
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Saurabh Kumar
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - James Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Eddy Kizana
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
30
|
Filice D, Dhahri W, Solan JL, Lampe PD, Steele E, Milani N, Van Biber B, Zhu WZ, Valdman TS, Romagnuolo R, Otero-Cruz JD, Hauch KD, Kay MW, Sarvazyan N, Laflamme MA. Optical mapping of human embryonic stem cell-derived cardiomyocyte graft electrical activity in injured hearts. Stem Cell Res Ther 2020; 11:417. [PMID: 32988411 PMCID: PMC7523067 DOI: 10.1186/s13287-020-01919-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/13/2020] [Accepted: 09/01/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) show tremendous promise for cardiac regeneration, but the successful development of hESC-CM-based therapies requires improved tools to investigate their electrical behavior in recipient hearts. While optical voltage mapping is a powerful technique for studying myocardial electrical activity ex vivo, we have previously shown that intra-cardiac hESC-CM grafts are not labeled by conventional voltage-sensitive fluorescent dyes. We hypothesized that the water-soluble voltage-sensitive dye di-2-ANEPEQ would label engrafted hESC-CMs and thereby facilitate characterization of graft electrical function and integration. METHODS We developed and validated a novel optical voltage mapping strategy based on the simultaneous imaging of the calcium-sensitive fluorescent protein GCaMP3, a graft-autonomous reporter of graft activation, and optical action potentials (oAPs) derived from di-2-ANEPEQ, which labels both graft and host myocardium. Cardiomyocytes from three different GCaMP3+ hESC lines (H7, RUES2, or ESI-17) were transplanted into guinea pig models of subacute and chronic infarction, followed by optical mapping at 2 weeks post-transplantation. RESULTS Use of a water-soluble voltage-sensitive dye revealed pro-arrhythmic properties of GCaMP3+ hESC-CM grafts from all three lines including slow conduction velocity, incomplete host-graft coupling, and spatially heterogeneous patterns of activation that varied beat-to-beat. GCaMP3+ hESC-CMs from the RUES2 and ESI-17 lines both showed prolonged oAP durations both in vitro and in vivo. Although hESC-CMs partially remuscularize the injured hearts, histological evaluation revealed immature graft structure and impaired gap junction expression at this early timepoint. CONCLUSION Simultaneous imaging of GCaMP3 and di-2-ANEPEQ allowed us to acquire the first unambiguously graft-derived oAPs from hESC-CM-engrafted hearts and yielded critical insights into their arrhythmogenic potential and line-to-line variation.
Collapse
Affiliation(s)
- Dominic Filice
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Wahiba Dhahri
- McEwen Stem Cell Institute, University Health Network, 101 College Street, Rm 3-908, Toronto, ON, M5G 1L7, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, M5G 2N2, Canada
| | - Joell L Solan
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Erin Steele
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Biology, University of Washington, Seattle, WA, 98195, USA
| | - Nikita Milani
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Benjamin Van Biber
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Wei-Zhong Zhu
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Tamilla Sadikov Valdman
- McEwen Stem Cell Institute, University Health Network, 101 College Street, Rm 3-908, Toronto, ON, M5G 1L7, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, M5G 2N2, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, 101 College Street, Rm 3-908, Toronto, ON, M5G 1L7, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, M5G 2N2, Canada
| | - José David Otero-Cruz
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Kip D Hauch
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Matthew W Kay
- Department of Biomedical Engineering, G. Washington University, Washington, DC, 20052, USA
| | - Narine Sarvazyan
- Department of Pharmacology & Physiology, G. Washington University, Washington, DC, 20052, USA
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, 101 College Street, Rm 3-908, Toronto, ON, M5G 1L7, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, M5G 2N2, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
31
|
Protze SI, Lee JH, Keller GM. Human Pluripotent Stem Cell-Derived Cardiovascular Cells: From Developmental Biology to Therapeutic Applications. Cell Stem Cell 2020; 25:311-327. [PMID: 31491395 DOI: 10.1016/j.stem.2019.07.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in our understanding of cardiovascular development have provided a roadmap for the directed differentiation of human pluripotent stem cells (hPSCs) to the major cell types found in the heart. In this Perspective, we review the state of the field in generating and maturing cardiovascular cells from hPSCs based on our fundamental understanding of heart development. We then highlight their applications for studying human heart development, modeling disease-performing drug screening, and cell replacement therapy. With the advancements highlighted here, the promise that hPSCs will deliver new treatments for degenerative and debilitating diseases may soon be fulfilled.
Collapse
Affiliation(s)
- Stephanie I Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jee Hoon Lee
- BlueRock Therapeutics ULC, Toronto, ON M5G 1L7, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
32
|
Assessment of Cardiotoxicity With Stem Cell-based Strategies. Clin Ther 2020; 42:1892-1910. [PMID: 32938533 DOI: 10.1016/j.clinthera.2020.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Adverse cardiovascular drug effects pose a substantial medical risk and represent a common cause of drug withdrawal from the market. Thus, current in vitro assays and in vivo animal models still have shortcomings in assessing cardiotoxicity. A human model for more accurate preclinical cardiotoxicity assessment is highly desirable. Current differentiation protocols allow for the generation of human pluripotent stem cell-derived cardiomyocytes in basically unlimited numbers and offer the opportunity to study drug effects on human cardiomyocytes. The purpose of this review is to provide a brief overview of the current approaches to translate studies with pluripotent stem cell-derived cardiomyocytes from basic science to preclinical risk assessment. METHODS A review of the literature was performed to gather data on the pathophysiology of cardiotoxicity, the current cardiotoxicity screening assays, stem cell-derived cardiomyocytes, and their application in cardiotoxicity screening. FINDINGS There is increasing evidence that stem cell-derived cardiomyocytes predict arrhythmogenicity with high accuracy. Cardiomyocyte immaturity represents the major limitation so far. However, strategies are being developed to overcome this hurdle, such as tissue engineering. In addition, stem cell-based strategies offer the possibility to assess structural drug toxicity (eg, by anticancer drugs) on complex models that more closely mirror the structure of the heart and contain endothelial cells and fibroblasts. IMPLICATIONS Pluripotent stem cell-derived cardiomyocytes have the potential to substantially change how preclinical cardiotoxicity screening is performed. To which extent they will replace or complement current approaches is being evaluated.
Collapse
|
33
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
34
|
Lippi M, Stadiotti I, Pompilio G, Sommariva E. Human Cell Modeling for Cardiovascular Diseases. Int J Mol Sci 2020; 21:E6388. [PMID: 32887493 PMCID: PMC7503257 DOI: 10.3390/ijms21176388] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 11/17/2022] Open
Abstract
The availability of appropriate and reliable in vitro cell models recapitulating human cardiovascular diseases has been the aim of numerous researchers, in order to retrace pathologic phenotypes, elucidate molecular mechanisms, and discover therapies using simple and reproducible techniques. In the past years, several human cell types have been utilized for these goals, including heterologous systems, cardiovascular and non-cardiovascular primary cells, and embryonic stem cells. The introduction of induced pluripotent stem cells and their differentiation potential brought new prospects for large-scale cardiovascular experiments, bypassing ethical concerns of embryonic stem cells and providing an advanced tool for disease modeling, diagnosis, and therapy. Each model has its advantages and disadvantages in terms of accessibility, maintenance, throughput, physiological relevance, recapitulation of the disease. A higher level of complexity in diseases modeling has been achieved with multicellular co-cultures. Furthermore, the important progresses reached by bioengineering during the last years, together with the opportunities given by pluripotent stem cells, have allowed the generation of increasingly advanced in vitro three-dimensional tissue-like constructs mimicking in vivo physiology. This review provides an overview of the main cell models used in cardiovascular research, highlighting the pros and cons of each, and describing examples of practical applications in disease modeling.
Collapse
Affiliation(s)
- Melania Lippi
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
| | - Ilaria Stadiotti
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.L.); (I.S.); (G.P.)
| |
Collapse
|
35
|
Huang Y, Wang T, López MEU, Hirano M, Hasan A, Shin SR. Recent advancements of human iPSC derived cardiomyocytes in drug screening and tissue regeneration. MICROPHYSIOLOGICAL SYSTEMS 2020; 4:2. [PMID: 39430371 PMCID: PMC11488690 DOI: 10.21037/mps-20-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Myocardial infarction together with subsequent heart failures are among the main reasons for death related to cardiovascular diseases (CVD). Restoring cardiac function and replacing scar tissue with healthy regenerated cardiomyocytes (CMs) is a hopeful therapy for heart failure. Human-induced pluripotent stem cell (hiPSC) derived CMs (hiPSC-CMs) offer the advantages of not having significant ethical issues and having negligible immunological rejection compared to other myocardial regeneration methods. hiPSCs can also produce an unlimited number of human CMs, another advantage they have compared with other cell sources for cardiac regeneration. Numerous researchers have focused their work on promoting the functional maturity of hiPSC-CMs, as well as finding out the precise regulatory mechanisms of each differentiation stage together with the economical and practical methods of acquisition and purification. However, the clinical applications of hiPSC-CMs in drug discovery and cardiac regeneration therapy have yet to be achieved. In this review, we present an overview of various methods for improving the differentiation efficiency of hiPSC-CMs and discuss the differences of electrophysiological characteristics between hiPSC-CMs and matured native CMs. We also introduce approaches for obtaining a large quantity of iPSC-CMs, which are needed to achieve biomanufacturing strategies for building biomimetic three-dimensional tissue constructs using combinations of biomaterials and advanced microfabrication techniques. Recent advances in specific iPSC technology-based drug screening platforms and regeneration therapies can suggest future directions for personalized medicine in biomedical applications.
Collapse
Affiliation(s)
- Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Ting Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- The Department of Laboratory Medicine. The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - María Elizabeth Urbina López
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- Instituto Tecnológico de Estudios Superiores de Monterrey, Campus Puebla, Puebla, México
| | - Minoru Hirano
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- Future Vehicle Research Department, Toyota Research Institute North America, Toyota Motor North America Inc., Ann Arbor, MI, USA
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
- Biomedical Research Centre (BRC), Qatar University, Doha, Qatar
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
| |
Collapse
|
36
|
Increased predominance of the matured ventricular subtype in embryonic stem cell-derived cardiomyocytes in vivo. Sci Rep 2020; 10:11883. [PMID: 32681032 PMCID: PMC7368005 DOI: 10.1038/s41598-020-68373-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence suggests that human pluripotent stem cell-derived cardiomyocytes can affect “heart regeneration”, replacing injured cardiac scar tissue with concomitant electrical integration. However, electrically coupled graft cardiomyocytes were found to innately induce transient post-transplant ventricular tachycardia in recent large animal model transplantation studies. We hypothesised that these phenomena were derived from alterations in the grafted cardiomyocyte characteristics. In vitro experiments showed that human embryonic stem cell-derived cardiomyocytes (hESC-CMs) contain nodal-like cardiomyocytes that spontaneously contract faster than working-type cardiomyocytes. When transplanted into athymic rat hearts, proliferative capacity was lower for nodal-like than working-type cardiomyocytes with grafted cardiomyocytes eventually comprising only relatively matured ventricular cardiomyocytes. RNA-sequencing of engrafted hESC-CMs confirmed the increased expression of matured ventricular cardiomyocyte-related genes, and simultaneous decreased expression of nodal cardiomyocyte-related genes. Temporal engraftment of electrical excitable nodal-like cardiomyocytes may thus explain the transient incidence of post-transplant ventricular tachycardia, although further large animal model studies will be required to control post-transplant arrhythmia.
Collapse
|
37
|
Jiang B, Yan L, Shamul JG, Hakun M, He X. Stem cell therapy of myocardial infarction: a promising opportunity in bioengineering. ADVANCED THERAPEUTICS 2020; 3:1900182. [PMID: 33665356 PMCID: PMC7928435 DOI: 10.1002/adtp.201900182] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is a life-threatening disease resulting from irreversible death of cardiomyocytes (CMs) and weakening of the heart blood-pumping function. Stem cell-based therapies have been studied for MI treatment over the last two decades with promising outcome. In this review, we critically summarize the past work in this field to elucidate the advantages and disadvantages of treating MI using pluripotent stem cells (PSCs) including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), adult stem cells, and cardiac progenitor cells. The main advantage of the latter is their cytokine production capability to modulate immune responses and control the progression of healing. However, human adult stem cells have very limited (if not 'no') capacity to differentiate into functional CMs in vitro or in vivo. In contrast, PSCs can be differentiated into functional CMs although the protocols for the cardiac differentiation of PSCs are mainly for adherent cells under 2D culture. Derivation of PSC-CMs in 3D, allowing for large-scale production of CMs via modulation of the Wnt/β-catenin signal pathway with defined chemicals and medium, may be desired for clinical translation. Furthermore, the technology of purification and maturation of the PSC-CMs may need further improvements to eliminate teratoma formation after in vivo implantation of the PSC-CMs for treating MI. In addition, in vitro derived PSC-CMs may have mechanical and electrical mismatch with the patient's cardiac tissue, which causes arrhythmia. This supports the use of PSC-derived cells committed to cardiac lineage without beating for implantation to treat MI. In this case, the PSC derived cells may utilize the mechanical, electrical, and chemical cues in the heart to further differentiate into mature/functional CMs in situ. Another major challenge facing stem cell therapy of MI is the low retention/survival of stem cells or their derivatives (e.g., PSC-CMs) in the heart for MI treatment after injection in vivo. This may be resolved by using biomaterials to engineer stem cells for reduced immunogenicity, immobilization of the cells in the heart, and increased integration with the host cardiac tissue. Biomaterials have also been applied in the derivation of CMs in vitro to increase the efficiency and maturation of differentiation. Collectively, a lot has been learned from the past failure of simply injecting intact stem cells or their derivatives in vivo for treating MI, and bioengineering stem cells with biomaterials is expected to be a valuable strategy for advancing stem cell therapy towards its widespread application for treating MI in the clinic.
Collapse
Affiliation(s)
- Bin Jiang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Maxwell Hakun
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
38
|
miR-128a Acts as a Regulator in Cardiac Development by Modulating Differentiation of Cardiac Progenitor Cell Populations. Int J Mol Sci 2020; 21:ijms21031158. [PMID: 32050579 PMCID: PMC7038042 DOI: 10.3390/ijms21031158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 11/18/2022] Open
Abstract
MicroRNAs (miRs) appear to be major, yet poorly understood players in regulatory networks guiding cardiogenesis. We sought to identify miRs with unknown functions during cardiogenesis analyzing the miR-profile of multipotent Nkx2.5 enhancer cardiac progenitor cells (NkxCE-CPCs). Besides well-known candidates such as miR-1, we found about 40 miRs that were highly enriched in NkxCE-CPCs, four of which were chosen for further analysis. Knockdown in zebrafish revealed that only miR-128a affected cardiac development and function robustly. For a detailed analysis, loss-of-function and gain-of-function experiments were performed during in vitro differentiations of transgenic murine pluripotent stem cells. MiR-128a knockdown (1) increased Isl1, Sfrp5, and Hcn4 (cardiac transcription factors) but reduced Irx4 at the onset of cardiogenesis, (2) upregulated Isl1-positive CPCs, whereas NkxCE-positive CPCs were downregulated, and (3) increased the expression of the ventricular cardiomyocyte marker Myl2 accompanied by a reduced beating frequency of early cardiomyocytes. Overexpression of miR-128a (4) diminished the expression of Isl1, Sfrp5, Nkx2.5, and Mef2c, but increased Irx4, (5) enhanced NkxCE-positive CPCs, and (6) favored nodal-like cardiomyocytes (Tnnt2+, Myh6+, Shox2+) accompanied by increased beating frequencies. In summary, we demonstrated that miR-128a plays a so-far unknown role in early heart development by affecting the timing of CPC differentiation into various cardiomyocyte subtypes.
Collapse
|
39
|
Poetsch MS, Guan K. iPSCs for modeling of sarcomeric cardiomyopathies. RECENT ADVANCES IN IPSC DISEASE MODELING, VOLUME 1 2020:237-273. [DOI: 10.1016/b978-0-12-822227-0.00012-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Sung TC, Liu CH, Huang WL, Lee YC, Kumar SS, Chang Y, Ling QD, Hsu ST, Higuchi A. Efficient differentiation of human ES and iPS cells into cardiomyocytes on biomaterials under xeno-free conditions. Biomater Sci 2019; 7:5467-5481. [PMID: 31656967 DOI: 10.1039/c9bm00817a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current xeno-free and chemically defined methods for the differentiation of hPSCs (human pluripotent stem cells) into cardiomyocytes are not efficient and are sometimes not reproducible. Therefore, it is necessary to develop reliable and efficient methods for the differentiation of hPSCs into cardiomyocytes for future use in cardiovascular research related to drug discovery, cardiotoxicity screening, and disease modeling. We evaluated two representative differentiation methods that were reported previously, and we further developed original, more efficient methods for the differentiation of hPSCs into cardiomyocytes under xeno-free, chemically defined conditions. The developed protocol successively differentiated hPSCs into cardiomyocytes, approximately 90-97% of which expressed the cardiac marker cTnT, with beating speeds and sarcomere lengths that were similar to those of a healthy adult human heart. The optimal cell culture biomaterials for the cardiac differentiation of hPSCs were also evaluated using extracellular matrix-mimetic material-coated dishes. Synthemax II-coated and Laminin-521-coated dishes were found to be the most effective and efficient biomaterials for the cardiac differentiation of hPSCs according to the observation of hPSC-derived cardiomyocytes with high survival ratios, high beating colony numbers, a similar beating frequency to that of a healthy adult human heart, high purity levels (high cTnT expression) and longer sarcomere lengths similar to those of a healthy adult human heart.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Devalla HD, Passier R. Cardiac differentiation of pluripotent stem cells and implications for modeling the heart in health and disease. Sci Transl Med 2019; 10:10/435/eaah5457. [PMID: 29618562 DOI: 10.1126/scitranslmed.aah5457] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 07/15/2016] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
Cellular models comprising cardiac cell types derived from human pluripotent stem cells are valuable for studying heart development and disease. We discuss transcriptional differences that define cellular identity in the heart, current methods for generating different cardiomyocyte subtypes, and implications for disease modeling, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Harsha D Devalla
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, Netherlands.
| | - Robert Passier
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, Netherlands. .,Department of Applied Stem Cell Technologies, Technical Medical Center, University of Twente, 7500 AE Enschede, Netherlands
| |
Collapse
|
42
|
A comprehensive, multiscale framework for evaluation of arrhythmias arising from cell therapy in the whole post-myocardial infarcted heart. Sci Rep 2019; 9:9238. [PMID: 31239508 PMCID: PMC6592890 DOI: 10.1038/s41598-019-45684-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 06/12/2019] [Indexed: 12/19/2022] Open
Abstract
Direct remuscularization approaches to cell-based heart repair seek to restore ventricular contractility following myocardial infarction (MI) by introducing new cardiomyocytes (CMs) to replace lost or injured ones. However, despite promising improvements in cardiac function, high incidences of ventricular arrhythmias have been observed in animal models of MI injected with pluripotent stem cell-derived cardiomyocytes (PSC-CMs). The mechanisms of arrhythmogenesis remain unclear. Here, we present a comprehensive framework for computational modeling of direct remuscularization approaches to cell therapy. Our multiscale 3D whole-heart modeling framework integrates realistic representations of cell delivery and transdifferentiation therapy modalities as well as representation of spatial distributions of engrafted cells, enabling simulation of clinical therapy and the prediction of emergent electrophysiological behavior and arrhythmogenensis. We employ this framework to explore how varying parameters of cell delivery and transdifferentiation could result in three mechanisms of arrhythmogenesis: focal ectopy, heart block, and reentry.
Collapse
|
43
|
Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, Radisic M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019; 198:3-26. [PMID: 30343824 PMCID: PMC6397087 DOI: 10.1016/j.biomaterials.2018.09.036] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/11/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Although investment in drug discovery and development has been sky-rocketing, the number of approved drugs has been declining. Cardiovascular toxicity due to therapeutic drug use claims the highest incidence and severity of adverse drug reactions in late-stage clinical development. Therefore, to address this issue, new, additional, replacement and combinatorial approaches are needed to fill the gap in effective drug discovery and screening. The motivation for developing accurate, predictive models is twofold: first, to study and discover new treatments for cardiac pathologies which are leading in worldwide morbidity and mortality rates; and second, to screen for adverse drug reactions on the heart, a primary risk in drug development. In addition to in vivo animal models, in vitro and in silico models have been recently proposed to mimic the physiological conditions of heart and vasculature. Here, we describe current in vitro, in vivo, and in silico platforms for modelling healthy and pathological cardiac tissues and their advantages and disadvantages for drug screening and discovery applications. We review the pathophysiology and the underlying pathways of different cardiac diseases, as well as the new tools being developed to facilitate their study. We finally suggest a roadmap for employing these non-animal platforms in assessing drug cardiotoxicity and safety.
Collapse
Affiliation(s)
- Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Samad Ahadian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada.
| |
Collapse
|
44
|
Addressing Variability and Heterogeneity of Induced Pluripotent Stem Cell-Derived Cardiomyocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:1-29. [DOI: 10.1007/5584_2019_350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Chen Z, Xian W, Bellin M, Dorn T, Tian Q, Goedel A, Dreizehnter L, Schneider CM, Ward-van Oostwaard D, Ng JKM, Hinkel R, Pane LS, Mummery CL, Lipp P, Moretti A, Laugwitz KL, Sinnecker D. Subtype-specific promoter-driven action potential imaging for precise disease modelling and drug testing in hiPSC-derived cardiomyocytes. Eur Heart J 2019; 38:292-301. [PMID: 28182242 PMCID: PMC5381588 DOI: 10.1093/eurheartj/ehw189] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 03/18/2016] [Accepted: 04/19/2016] [Indexed: 12/30/2022] Open
Affiliation(s)
- Zhifen Chen
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Wenying Xian
- Institute for Molecular Cell Biology, Medical Faculty, University Homburg/Saar, Universität des Saarlandes, Homburg/Saar 66421, Germany
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Tatjana Dorn
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Qinghai Tian
- Institute for Molecular Cell Biology, Medical Faculty, University Homburg/Saar, Universität des Saarlandes, Homburg/Saar 66421, Germany
| | - Alexander Goedel
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Lisa Dreizehnter
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Christine M Schneider
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Dorien Ward-van Oostwaard
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Judy King Man Ng
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | | | - Luna Simona Pane
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Peter Lipp
- Institute for Molecular Cell Biology, Medical Faculty, University Homburg/Saar, Universität des Saarlandes, Homburg/Saar 66421, Germany
| | | | | | - Daniel Sinnecker
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| |
Collapse
|
46
|
Jara Avaca M, Gruh I. Bioengineered Cardiac Tissue Based on Human Stem Cells for Clinical Application. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:117-146. [PMID: 29218360 DOI: 10.1007/10_2017_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Engineered cardiac tissue might enable novel therapeutic strategies for the human heart in a number of acquired and congenital diseases. With recent advances in stem cell technologies, namely the availability of pluripotent stem cells, the generation of potentially autologous tissue grafts has become a realistic option. Nevertheless, a number of limitations still have to be addressed before clinical application of engineered cardiac tissue based on human stem cells can be realized. We summarize current progress and pending challenges regarding the optimal cell source, cardiomyogenic lineage specification, purification, safety of genetic cell engineering, and genomic stability. Cardiac cells should be combined with clinical grade scaffold materials for generation of functional myocardial tissue in vitro. Scale-up to clinically relevant dimensions is mandatory, and tissue vascularization is most probably required both for preclinical in vivo testing in suitable large animal models and for clinical application. Graphical Abstract.
Collapse
Affiliation(s)
- Monica Jara Avaca
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Vascular and Transplantation Surgery (HTTG), Hannover Medical School (MHH) & Cluster of Excellence REBIRTH, Hannover, Germany
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Vascular and Transplantation Surgery (HTTG), Hannover Medical School (MHH) & Cluster of Excellence REBIRTH, Hannover, Germany.
| |
Collapse
|
47
|
Scalable Cardiac Differentiation of Pluripotent Stem Cells Using Specific Growth Factors and Small Molecules. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:39-69. [PMID: 29071404 DOI: 10.1007/10_2017_30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The envisioned routine application of human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) for therapies and industry-compliant screening approaches will require efficient and highly reproducible processes for the mass production of well-characterized CM batches.On their way toward beating CMs, hPSCs initially undergo an epithelial-to-mesenchymal transition into a primitive-streak (PS)-like population that later gives rise to all endodermal and mesodermal lineages, including cardiovascular progenies (CVPs). CVPs are multipotent and possess the capability to give rise to all major cell types of the heart, including CMs, endothelial cells, cardiac fibroblasts, and smooth muscle cells. This article provides an historical overview and describes the stepwise development of protocols that typically result in the appearance of beating CMs within 7-12 days of hPSC differentiation.We describe the development of directed and closely controlled cardiomyogenic differentiation, which now enables the induction of >90% CM purity without further lineage enrichment. Although secreted lineage specifiers (revealed from developmental biology) were initially used, we outline the advantages of chemical pathway modulators, as defined by more recent screening approaches. Subsequently, we discuss the use of defined culture media for upscaling the production of hPSC-CMs in controlled bioreactors and how this, in principle, unlimited source of human CMs can be used to progress heart regeneration and stimulate the drug discovery pipeline. Graphical Abstract.
Collapse
|
48
|
Oikonomopoulos A, Kitani T, Wu JC. Pluripotent Stem Cell-Derived Cardiomyocytes as a Platform for Cell Therapy Applications: Progress and Hurdles for Clinical Translation. Mol Ther 2018; 26:1624-1634. [PMID: 29699941 PMCID: PMC6035734 DOI: 10.1016/j.ymthe.2018.02.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Regenerative therapy has been applied to restore lost cardiac muscle and cardiac performance. Induced pluripotent stem cells (iPSCs) can provide an unlimited source of cardiomyocytes and therefore play a key role in cardiac regeneration. Despite initial encouraging results from pre-clinical studies, progress toward clinical applications has been hampered by issues such as tumorigenesis, arrhythmogenesis, immune rejection, scalability, low graft-cell survival, and poor engraftment. Here, we review recent developments in iPSC research on regenerating injured heart tissue, including novel advances in cell therapy and potential strategies to overcome current obstacles in the field.
Collapse
Affiliation(s)
- Angelos Oikonomopoulos
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
49
|
Human-Induced Pluripotent Stem Cell Technology and Cardiomyocyte Generation: Progress and Clinical Applications. Cells 2018; 7:cells7060048. [PMID: 29799480 PMCID: PMC6025241 DOI: 10.3390/cells7060048] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/16/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) are reprogrammed cells that have hallmarks similar to embryonic stem cells including the capacity of self-renewal and differentiation into cardiac myocytes. The improvements in reprogramming and differentiating methods achieved in the past 10 years widened the use of hiPSCs, especially in cardiac research. hiPSC-derived cardiac myocytes (CMs) recapitulate phenotypic differences caused by genetic variations, making them attractive human disease models and useful tools for drug discovery and toxicology testing. In addition, hiPSCs can be used as sources of cells for cardiac regeneration in animal models. Here, we review the advances in the genetic and epigenetic control of cardiomyogenesis that underlies the significant improvement of the induced reprogramming of somatic cells to CMs; the methods used to improve scalability of throughput assays for functional screening and drug testing in vitro; the phenotypic characteristics of hiPSCs-derived CMs and their ability to rescue injured CMs through paracrine effects; we also cover the novel approaches in tissue engineering for hiPSC-derived cardiac tissue generation, and finally, their immunological features and the potential use in biomedical applications.
Collapse
|
50
|
Brandão KO, Tabel VA, Atsma DE, Mummery CL, Davis RP. Human pluripotent stem cell models of cardiac disease: from mechanisms to therapies. Dis Model Mech 2018; 10:1039-1059. [PMID: 28883014 PMCID: PMC5611968 DOI: 10.1242/dmm.030320] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is now a decade since human induced pluripotent stem cells (hiPSCs) were first described. The reprogramming of adult somatic cells to a pluripotent state has become a robust technology that has revolutionised our ability to study human diseases. Crucially, these cells capture all the genetic aspects of the patient from which they were derived. Combined with advances in generating the different cell types present in the human heart, this has opened up new avenues to study cardiac disease in humans and investigate novel therapeutic approaches to treat these pathologies. Here, we provide an overview of the current state of the field regarding the generation of cardiomyocytes from human pluripotent stem cells and methods to assess them functionally, an essential requirement when investigating disease and therapeutic outcomes. We critically evaluate whether treatments suggested by these in vitro models could be translated to clinical practice. Finally, we consider current shortcomings of these models and propose methods by which they could be further improved.
Collapse
Affiliation(s)
- Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Viola A Tabel
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|