101
|
Cpmer: A new conserved eEF1A2-binding partner that regulates Eomes translation and cardiomyocyte differentiation. Stem Cell Reports 2022; 17:1154-1169. [PMID: 35395174 PMCID: PMC9133893 DOI: 10.1016/j.stemcr.2022.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Previous studies have shown that eukaryotic elongation factor 1A2 (eEF1A2) serves as an essential heart-specific translation elongation element and that its mutation or knockout delays heart development and causes congenital heart disease and death among species. However, the function and regulatory mechanisms of eEF1A2 in mammalian heart development remain largely unknown. Here we identified the long noncoding RNA (lncRNA) Cpmer (cytoplasmic mesoderm regulator), which interacted with eEF1A2 to co-regulate differentiation of mouse and human embryonic stem cell-derived cardiomyocytes. Mechanistically, Cpmer specifically recognized Eomes mRNA by RNA-RNA pairing and facilitated binding of eEF1A2 with Eomes mRNA, guaranteeing Eomes mRNA translation and cardiomyocyte differentiation. Our data reveal a novel functionally conserved lncRNA that can specifically regulate Eomes translation and cardiomyocyte differentiation, which broadens our understanding of the mechanism of lncRNA involvement in the subtle translational regulation of eEF1A2 during mammalian heart development.
Collapse
|
102
|
Thomas D, de Jesus Perez VA, Sayed N. An evidence appraisal of heart organoids in a dish and commensurability to human heart development in vivo. BMC Cardiovasc Disord 2022; 22:122. [PMID: 35317745 PMCID: PMC8939187 DOI: 10.1186/s12872-022-02543-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Stem-cell derived in vitro cardiac models have provided profound insights into mechanisms in cardiac development and disease. Efficient differentiation of specific cardiac cell types from human pluripotent stem cells using a three-step Wnt signaling modulation has been one of the major discoveries that has enabled personalized cardiovascular disease modeling approaches. Generation of cardiac cell types follow key development stages during embryogenesis, they intuitively are excellent models to study cardiac tissue patterning in primitive cardiac structures. Here, we provide a brief overview of protocols that have laid the foundation for derivation of stem-cell derived three-dimensional cardiac models. Further this article highlights features and utility of the models to distinguish the advantages and trade-offs in modeling embryonic development and disease processes. Finally, we discuss the challenges in improving robustness in the current models and utilizing developmental principles to bring higher physiological relevance. In vitro human cardiac models are complimentary tools that allow mechanistic interrogation in a reductionist way. The unique advantage of utilizing patient specific stem cells and continued improvements in generating reliable organoid mimics of the heart will boost predictive power of these tools in basic and translational research.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- , Stanford, CA, USA.
| |
Collapse
|
103
|
Murata K, Masumoto H. Systems for the functional evaluation of human heart tissues derived from pluripotent stem cells. Stem Cells 2022; 40:537-545. [PMID: 35303744 PMCID: PMC9216506 DOI: 10.1093/stmcls/sxac022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/06/2022] [Indexed: 11/13/2022]
Abstract
Human pluripotent stem cells (hPSCs) are expected to be a promising cell source in regenerative medicine and drug discovery for the treatment of various intractable diseases. An approach for creating a three-dimensional (3D) structure from hPSCs that mimics human cardiac tissue functions has made it theoretically possible to conduct drug discovery and cardiotoxicity tests by assessing pharmacological responses in human cardiac tissues by a screening system using a compound library. The myocardium functions as a tissue composed of organized vascular networks, supporting stromal cells and cardiac muscle cells. Considering this, the reconstruction of tissue structure by various cells of cardiovascular lineages, such as vascular cells and cardiac muscle cells, is desirable for the ideal conformation of hPSC-derived cardiac tissues. Heart-on-a-chip, an organ-on-a-chip system to evaluate the physiological pump function of 3D cardiac tissues might hold promise in medical researches such as drug discovery and regenerative medicine. Here, we review various modalities to evaluate the function of human stem cell-derived cardiac tissues and introduce heart-on-a-chip systems that can recapitulate physiological parameters of hPSC-derived cardiac tissues.
Collapse
Affiliation(s)
- Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
104
|
Ruan T, Harney D, Koay YC, Loo L, Larance M, Caron L. Anabolic Factors and Myokines Improve Differentiation of Human Embryonic Stem Cell Derived Skeletal Muscle Cells. Cells 2022; 11:cells11060963. [PMID: 35326414 PMCID: PMC8946006 DOI: 10.3390/cells11060963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscle weakness is linked to many adverse health outcomes. Current research to identify new drugs has often been inconclusive due to lack of adequate cellular models. We previously developed a scalable monolayer system to differentiate human embryonic stem cells (hESCs) into mature skeletal muscle cells (SkMCs) within 26 days without cell sorting or genetic manipulation. Here, building on our previous work, we show that differentiation and fusion of myotubes can be further enhanced using the anabolic factors testosterone (T) and follistatin (F) in combination with a cocktail of myokines (C). Importantly, combined TFC treatment significantly enhanced both the hESC-SkMC fusion index and the expression levels of various skeletal muscle markers, including the motor protein myosin heavy chain (MyHC). Transcriptomic and proteomic analysis revealed oxidative phosphorylation as the most up-regulated pathway, and a significantly higher level of ATP and increased mitochondrial mass were also observed in TFC-treated hESC-SkMCs, suggesting enhanced energy metabolism is coupled with improved muscle differentiation. This cellular model will be a powerful tool for studying in vitro myogenesis and for drug discovery pertaining to further enhancing muscle development or treating muscle diseases.
Collapse
Affiliation(s)
- Travis Ruan
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (T.R.); (L.L.)
| | - Dylan Harney
- Larance Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (D.H.); (M.L.)
| | - Yen Chin Koay
- Cardiometabolic Disease Group, Heart Research Institute, Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Lipin Loo
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (T.R.); (L.L.)
| | - Mark Larance
- Larance Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (D.H.); (M.L.)
| | - Leslie Caron
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (T.R.); (L.L.)
- MMG, Marseille Medical Genetics, Aix Marseille Univ, INSERM U1251, 13005 Marseille, France
- Correspondence:
| |
Collapse
|
105
|
Cardiac Cell Therapy with Pluripotent Stem Cell-Derived Cardiomyocytes: What Has Been Done and What Remains to Do? Curr Cardiol Rep 2022; 24:445-461. [PMID: 35275365 PMCID: PMC9068652 DOI: 10.1007/s11886-022-01666-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Exciting pre-clinical data presents pluripotent stem cell-derived cardiomyocytes (PSC-CM) as a novel therapeutic prospect following myocardial infarction, and worldwide clinical trials are imminent. However, despite notable advances, several challenges remain. Here, we review PSC-CM pre-clinical studies, identifying key translational hurdles. We further discuss cell production and characterization strategies, identifying markers that may help generate cells which overcome these barriers. RECENT FINDINGS PSC-CMs can robustly repopulate infarcted myocardium with functional, force generating cardiomyocytes. However, current differentiation protocols produce immature and heterogenous cardiomyocytes, creating related issues such as arrhythmogenicity, immunogenicity and poor engraftment. Recent efforts have enhanced our understanding of cardiovascular developmental biology. This knowledge may help implement novel differentiation or gene editing strategies that could overcome these limitations. PSC-CMs are an exciting therapeutic prospect. Despite substantial recent advances, limitations of the technology remain. However, with our continued and increasing biological understanding, these issues are addressable, with several worldwide clinical trials anticipated in the coming years.
Collapse
|
106
|
Jiang B, Ou W, Shamul JG, Chen H, Van Belleghem S, Stewart S, Liu Z, Fisher JP, He X. Rock inhibitor may compromise human induced pluripotent stem cells for cardiac differentiation in 3D. Bioact Mater 2022; 9:508-522. [PMID: 34786523 PMCID: PMC8581226 DOI: 10.1016/j.bioactmat.2021.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 11/22/2022] Open
Abstract
Cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) are valuable for the understanding/treatment of the deadly heart diseases and their drug screening. However, the very much needed homogeneous 3D cardiac differentiation of human iPSCs is still challenging. Here, it is discovered surprisingly that Rock inhibitor (RI), used ubiquitously to improve the survival/yield of human iPSCs, induces early gastrulation-like change to human iPSCs in 3D culture and may cause their heterogeneous differentiation into all the three germ layers (i.e., ectoderm, mesoderm, and endoderm) at the commonly used concentration (10 μM). This greatly compromises the capacity of human iPSCs for homogeneous 3D cardiac differentiation. By reducing the RI to 1 μM for 3D culture, the human iPSCs retain high pluripotency/quality in inner cell mass-like solid 3D spheroids. Consequently, the beating efficiency of 3D cardiac differentiation can be improved to more than 95 % in ~7 days (compared to less than ~50 % in 14 days for the 10 μM RI condition). Furthermore, the outset beating time (OBT) of all resultant cardiac spheroids (CSs) is synchronized within only 1 day and they form a synchronously beating 3D construct after 5-day culture in gelatin methacrylol (GelMA) hydrogel, showing high homogeneity (in terms of the OBT) in functional maturity of the CSs. Moreover, the resultant cardiomyocytes are of high quality with key functional ultrastructures and highly responsive to cardiac drugs. These discoveries may greatly facilitate the utilization of human iPSCs for understanding and treating heart diseases.
Collapse
Affiliation(s)
- Bin Jiang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - James G. Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - Hao Chen
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - Sarah Van Belleghem
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, 65212, United States
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, 21201, United States
| |
Collapse
|
107
|
Kim H, Song BW, Park SJ, Choi SW, Moon H, Hwang KC, Kang SW, Moon SH, Yang Y, Kwon IC, Kim SH. Ultraefficient extracellular vesicle-guided direct reprogramming of fibroblasts into functional cardiomyocytes. SCIENCE ADVANCES 2022; 8:eabj6621. [PMID: 35213232 PMCID: PMC8880777 DOI: 10.1126/sciadv.abj6621] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 12/31/2021] [Indexed: 05/26/2023]
Abstract
Direct lineage conversion holds great promise in the regenerative medicine field for restoring damaged tissues using functionally engineered counterparts. However, current methods of direct lineage conversion, even those using virus-mediated transgenic expression of tumorigenic factors, are extremely inefficient (~25%). Thus, advanced methodologies capable of revolutionizing efficiency and addressing safety concerns are key to clinical translation of these technologies. Here, we propose an extracellular vesicle (EV)-guided, nonviral, direct lineage conversion strategy to enhance transdifferentiation of fibroblasts to induced cardiomyocyte-like cells (iCMs). The resulting iCMs have typical cardiac Ca2+ transients and electrophysiological features and exhibit global gene expression profiles similar to those of cardiomyocytes. This is the first demonstration of the use of EVs derived from embryonic stem cells undergoing cardiac differentiation as biomimetic tools to induce cardiac reprogramming with extremely high efficiency (>60%), establishing a general, more readily accessible platform for generating a variety of specialized somatic cells through direct lineage conversion.
Collapse
Affiliation(s)
- Hyosuk Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Byeong-Wook Song
- College of Medicine, Institute for Bio-Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Soon-Jung Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Seong Woo Choi
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hanbyeol Moon
- College of Medicine, Institute for Bio-Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Ki-Chul Hwang
- College of Medicine, Institute for Bio-Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Sun-Woong Kang
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Sung-Hwan Moon
- College of Medicine, Institute for Bio-Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- KIST-DFCI ON-SITE-LAB, Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
108
|
Ramirez-Calderon G, Colombo G, Hernandez-Bautista CA, Astro V, Adamo A. Heart in a Dish: From Traditional 2D Differentiation Protocols to Cardiac Organoids. Front Cell Dev Biol 2022; 10:855966. [PMID: 35252213 PMCID: PMC8893312 DOI: 10.3389/fcell.2022.855966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) constitute a valuable model to study the complexity of early human cardiac development and investigate the molecular mechanisms involved in heart diseases. The differentiation of hPSCs into cardiac lineages in vitro can be achieved by traditional two-dimensional (2D) monolayer approaches or by adopting innovative three-dimensional (3D) cardiac organoid protocols. Human cardiac organoids (hCOs) are complex multicellular aggregates that faithfully recapitulate the cardiac tissue’s transcriptional, functional, and morphological features. In recent years, significant advances in the field have dramatically improved the robustness and efficiency of hCOs derivation and have promoted the application of hCOs for drug screening and heart disease modeling. This review surveys the current differentiation protocols, focusing on the most advanced 3D methods for deriving hCOs from hPSCs. Furthermore, we describe the potential applications of hCOs in the pharmaceutical and tissue bioengineering fields, including their usage to investigate the consequences of Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV2) infection in the heart.
Collapse
|
109
|
The role of metabolism in directed differentiation versus trans-differentiation of cardiomyocytes. Semin Cell Dev Biol 2022; 122:56-65. [PMID: 34074592 PMCID: PMC8725317 DOI: 10.1016/j.semcdb.2021.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
The advent of induced pluripotent stem cells (iPSCs) and identification of transcription factors for cardiac reprogramming have raised hope to cure heart disease, the leading cause of death in the world. Our knowledge in heart development and molecular barriers of cardiac reprogramming is advancing, but many hurdles are yet to be overcome for clinical translation. Importantly, we lack a full understanding of molecular mechanisms governing cell fate conversion toward cardiomyocytes. In this review, we will discuss the role of metabolism in directed differentiation versus trans-differentiation of cardiomyocytes. Cardiomyocytes exhibit a unique metabolic feature distinct from PSCs and cardiac fibroblasts, and there are multiple overlapping molecular mechanisms underlying metabolic reprogramming during cardiomyogenesis. We will discuss key metabolic changes occurring during cardiomyocytes differentiation from PSCs and cardiac fibroblasts, and the potential role of metabolic reprogramming in the enhancement strategies for cardiomyogenesis. Only when such details are discovered will more effective strategies to enhance the de novo production of cardiomyocytes be possible.
Collapse
|
110
|
Hota SK, Rao KS, Blair AP, Khalilimeybodi A, Hu KM, Thomas R, So K, Kameswaran V, Xu J, Polacco BJ, Desai RV, Chatterjee N, Hsu A, Muncie JM, Blotnick AM, Winchester SAB, Weinberger LS, Hüttenhain R, Kathiriya IS, Krogan NJ, Saucerman JJ, Bruneau BG. Brahma safeguards canalization of cardiac mesoderm differentiation. Nature 2022; 602:129-134. [PMID: 35082446 PMCID: PMC9196993 DOI: 10.1038/s41586-021-04336-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/08/2021] [Indexed: 12/26/2022]
Abstract
Differentiation proceeds along a continuum of increasingly fate-restricted intermediates, referred to as canalization1,2. Canalization is essential for stabilizing cell fate, but the mechanisms that underlie robust canalization are unclear. Here we show that the BRG1/BRM-associated factor (BAF) chromatin-remodelling complex ATPase gene Brm safeguards cell identity during directed cardiogenesis of mouse embryonic stem cells. Despite the establishment of a well-differentiated precardiac mesoderm, Brm-/- cells predominantly became neural precursors, violating germ layer assignment. Trajectory inference showed a sudden acquisition of a non-mesodermal identity in Brm-/- cells. Mechanistically, the loss of Brm prevented de novo accessibility of primed cardiac enhancers while increasing the expression of neurogenic factor POU3F1, preventing the binding of the neural suppressor REST and shifting the composition of BRG1 complexes. The identity switch caused by the Brm mutation was overcome by increasing BMP4 levels during mesoderm induction. Mathematical modelling supports these observations and demonstrates that Brm deletion affects cell fate trajectory by modifying saddle-node bifurcations2. In the mouse embryo, Brm deletion exacerbated mesoderm-deleted Brg1-mutant phenotypes, severely compromising cardiogenesis, and reveals an in vivo role for Brm. Our results show that Brm is a compensable safeguard of the fidelity of mesoderm chromatin states, and support a model in which developmental canalization is not a rigid irreversible path, but a highly plastic trajectory.
Collapse
Affiliation(s)
- Swetansu K Hota
- Gladstone Institutes, San Francisco, CA, USA.
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA.
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
| | - Kavitha S Rao
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | | | - Ali Khalilimeybodi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kevin M Hu
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | | | - Kevin So
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Vasumathi Kameswaran
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Jiewei Xu
- Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | - Benjamin J Polacco
- Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | | | | | - Austin Hsu
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | | | - Aaron M Blotnick
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Sarah A B Winchester
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
| | - Leor S Weinberger
- Gladstone Institutes, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Ruth Hüttenhain
- Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | - Irfan S Kathiriya
- Gladstone Institutes, San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA, USA.
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA.
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
111
|
Dhahri W, Sadikov Valdman T, Wilkinson D, Pereira E, Ceylan E, Andharia N, Qiang B, Masoudpour H, Wulkan F, Quesnel E, Jiang W, Funakoshi S, Mazine A, Gomez-Garcia MJ, Latifi N, Jiang Y, Huszti E, Simmons CA, Keller G, Laflamme MA. In Vitro Matured Human Pluripotent Stem Cell-derived Cardiomyocytes Form Grafts With Enhanced Structure and Function in Injured Hearts. Circulation 2022; 145:1412-1426. [PMID: 35089805 DOI: 10.1161/circulationaha.121.053563] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have tremendous promise for application in cardiac regeneration, but their translational potential is limited by an immature phenotype. We hypothesized that large-scale manufacturing of mature hPSC-CMs could be achieved via culture on polydimethylsiloxane (PDMS) lined roller bottles and that the transplantation of these cells would mediate better structural and functional outcomes than with conventional immature hPSC-CM populations. METHODS We comprehensively phenotyped hPSC-CMs after in vitro maturation for 20 and 40 days on either PDMS or standard tissue culture plastic (TCP) substrates. All hPSC-CMs were generated using a transgenic hPSC line that stably expressed a voltage-sensitive fluorescent reporter to facilitate in vitro and in vivo electrophysiological studies, and cardiomyocyte populations were also analyzed in vitro by immunocytochemistry, ultrastructure and fluorescent calcium imaging, as well as bulk and single-cell transcriptomics. We next compared outcomes after the transplantation of these populations into a guinea pig model of myocardial infarction (MI) using endpoints including histology, optical mapping of graft- and host-derived action potentials, echocardiography, and telemetric electrocardiographic (ECG) monitoring. RESULTS We demonstrated the economic generation of >1x108 mature hPSC-CMs per PDMS-lined roller bottle. Compared to their counterparts generated on TCP substrates, PDMS-matured hPSC-CMs exhibited increased cardiac gene expression and more mature structural and functional properties in vitro. More importantly, intra-cardiac grafts formed with PDMS-matured myocytes showed greatly enhanced structure and alignment, better host-graft electromechanical integration, less pro-arrhythmic behavior, and greater beneficial effects on contractile function. CONCLUSIONS In summary, we describe practical methods for the scaled generation of mature hPSC-CMs and provide the first evidence that the transplantation of more mature cardiomyocytes yields better outcomes in vivo.
Collapse
Affiliation(s)
- Wahiba Dhahri
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | | | | | | | - Eylül Ceylan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Naaz Andharia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Beiping Qiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Hassan Masoudpour
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Fanny Wulkan
- 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
| | - Shunsuke Funakoshi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Amine Mazine
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - M Juliana Gomez-Garcia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Neda Latifi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Yidi Jiang
- Biostatistics Research Unit, University Health Network, Toronto, ON, Canada
| | - Ella Huszti
- Biostatistics Research Unit, University Health Network, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Gordon Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, 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 and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
112
|
Harnessing the Power of Stem Cell Models to Study Shared Genetic Variants in Congenital Heart Diseases and Neurodevelopmental Disorders. Cells 2022; 11:cells11030460. [PMID: 35159270 PMCID: PMC8833927 DOI: 10.3390/cells11030460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Advances in human pluripotent stem cell (hPSC) technology allow one to deconstruct the human body into specific disease-relevant cell types or create functional units representing various organs. hPSC-based models present a unique opportunity for the study of co-occurring disorders where “cause and effect” can be addressed. Poor neurodevelopmental outcomes have been reported in children with congenital heart diseases (CHD). Intuitively, abnormal cardiac function or surgical intervention may stunt the developing brain, leading to neurodevelopmental disorders (NDD). However, recent work has uncovered several genetic variants within genes associated with the development of both the heart and brain that could also explain this co-occurrence. Given the scalability of hPSCs, straightforward genetic modification, and established differentiation strategies, it is now possible to investigate both CHD and NDD as independent events. We will first overview the potential for shared genetics in both heart and brain development. We will then summarize methods to differentiate both cardiac & neural cells and organoids from hPSCs that represent the developmental process of the heart and forebrain. Finally, we will highlight strategies to rapidly screen several genetic variants together to uncover potential phenotypes and how therapeutic advances could be achieved by hPSC-based models.
Collapse
|
113
|
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
|
114
|
Ng WH, Johnston EK, Tan JJ, Bliley JM, Feinberg AW, Stolz DB, Sun M, Wijesekara P, Hawkins F, Kotton DN, Ren X. Recapitulating human cardio-pulmonary co-development using simultaneous multilineage differentiation of pluripotent stem cells. eLife 2022; 11:67872. [PMID: 35018887 PMCID: PMC8846595 DOI: 10.7554/elife.67872] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
The extensive crosstalk between the developing heart and lung is critical to their proper morphogenesis and maturation. However, there remains a lack of models that investigate the critical cardio-pulmonary mutual interaction during human embryogenesis. Here, we reported a novel stepwise strategy for directing the simultaneous induction of both mesoderm-derived cardiac and endoderm-derived lung epithelial lineages within a single differentiation of human-induced pluripotent stem cells (hiPSCs) via temporal specific tuning of WNT and nodal signaling in the absence of exogenous growth factors. Using 3D suspension culture, we established concentric cardio-pulmonary micro-Tissues (μTs), and expedited alveolar maturation in the presence of cardiac accompaniment. Upon withdrawal of WNT agonist, the cardiac and pulmonary components within each dual-lineage μT effectively segregated from each other with concurrent initiation of cardiac contraction. We expect that our multilineage differentiation model will offer an experimentally tractable system for investigating human cardio-pulmonary interaction and tissue boundary formation during embryogenesis. Organs begin developing during the first few months of pregnancy, while the baby is still an embryo. These early stages of development are known as embryogenesis – a tightly organized process, during which the embryo forms different layers of stem cells. These cells can be activated to turn into a particular type of cell, such as a heart or a lung cell. The heart and lungs develop from different layers within the embryo, which must communicate with each other for the organs to form correctly. For example, chemical signals can be released from and travel between layers of the embryo, activating processes inside cells located in the different areas. In mouse models, chemical signals and cells travel between developing heart and lung, which helps both organs to form into the correct structure. But it is unclear how well the observations from mouse models translate to heart and lung development in humans. To find out more, Ng et al. developed a human model of heart and lung co-development during embryogenesis using human pluripotent stem cells. The laboratory-grown stem cells were treated with chemical signals, causing them to form different layers that developed into early forms of heart and lung cells. The cells were then transferred into a specific growing condition, where they arranged into three-dimensional structures termed microtissues. Ng et al. found that lung cells developed faster when grown in microtissues with accompanying developing heart cells compared to microtissues containing only developing lung cells. In addition, Ng et al. revealed that the co-developing heart and lung tissues automatically separate from each other during later stage, without the need for chemical signals. This human cell-based model of early forms of co-developing heart and lung cells may help provide researchers with new strategies to probe the underlying mechanisms of human heart and lung interaction during embryogenesis.
Collapse
Affiliation(s)
- Wai Hoe Ng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, United States
| | - Elizabeth K Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, United States
| | - Jun Jie Tan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Jacqueline M Bliley
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, United States
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, United States
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, United States
| | - Ming Sun
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, United States
| | - Piyumi Wijesekara
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, United States
| | - Finn Hawkins
- Center for Regenerative Medicine, Boston University, Boston, United States
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University, Boston, MA, United States
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, United States
| |
Collapse
|
115
|
Thomas D, Cunningham NJ, Shenoy S, Wu JC. Human-induced pluripotent stem cells in cardiovascular research: current approaches in cardiac differentiation, maturation strategies, and scalable production. Cardiovasc Res 2022; 118:20-36. [PMID: 33757124 PMCID: PMC8932155 DOI: 10.1093/cvr/cvab115] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Manifestations of cardiovascular diseases (CVDs) in a patient or a population differ based on inherent biological makeup, lifestyle, and exposure to environmental risk factors. These variables mean that therapeutic interventions may not provide the same benefit to every patient. In the context of CVDs, human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) offer an opportunity to model CVDs in a patient-specific manner. From a pharmacological perspective, iPSC-CM models can serve as go/no-go tests to evaluate drug safety. To develop personalized therapies for early diagnosis and treatment, human-relevant disease models are essential. Hence, to implement and leverage the utility of iPSC-CMs for large-scale treatment or drug discovery, it is critical to (i) carefully evaluate the relevant limitations of iPSC-CM differentiations, (ii) establish quality standards for defining the state of cell maturity, and (iii) employ techniques that allow scalability and throughput with minimal batch-to-batch variability. In this review, we briefly describe progress made with iPSC-CMs in disease modelling and pharmacological testing, as well as current iPSC-CM maturation techniques. Finally, we discuss current platforms for large-scale manufacturing of iPSC-CMs that will enable high-throughput drug screening applications.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| | - Nathan J Cunningham
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| | - Sushma Shenoy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| |
Collapse
|
116
|
OUP accepted manuscript. Stem Cells 2022; 40:260-272. [DOI: 10.1093/stmcls/sxab017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/04/2021] [Indexed: 11/12/2022]
|
117
|
Simmons DW, Huebsch N. iPSC-Derived Micro-Heart Muscle for Medium-Throughput Pharmacology and Pharmacogenomic Studies. Methods Mol Biol 2022; 2485:111-131. [PMID: 35618902 DOI: 10.1007/978-1-0716-2261-2_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Micro-heart muscle arrays enable medium-throughput experiments to model the cardiac response to a variety of environmental and pharmaceutical effects. Here, we describe stem cell culture maintenance, methods for successful cardiac differentiation, and formation of micro-heart muscle arrays for electrophysiology and molecular biology assays.
Collapse
Affiliation(s)
- Daniel W Simmons
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO, USA.
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
118
|
Zhou Y, Zhang J. Remuscularization of Ventricular Infarcts Using the Existing Cardiac Cells. ADVANCED TECHNOLOGIES IN CARDIOVASCULAR BIOENGINEERING 2022:51-78. [DOI: 10.1007/978-3-030-86140-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
119
|
Lee CS, Kim J, Cho HJ, Kim HS. Cardiovascular Regeneration via Stem Cells and Direct Reprogramming: A Review. Korean Circ J 2022; 52:341-353. [PMID: 35502566 PMCID: PMC9064703 DOI: 10.4070/kcj.2022.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Despite recent advancements in treatment strategies, cardiovascular disease such as heart failure remains a significant source of global mortality. Stem cell technology and cellular reprogramming are rapidly growing fields that will continue to prove useful in cardiac regenerative therapeutics. This review provides information on the role of human pluripotent stem cells (hPSCs) in cardiac regeneration and discusses the practical applications of hPSC-derived cardiomyocytes (CMCs). Moreover, we discuss the practical applications of hPSC-derived CMCs while outlining the relevance of directly-reprogrammed CMCs in regenerative medicine. This review critically summarizes the most recent advances in the field will help to guide future research in this developing area. Cardiovascular disease (CVD) is the leading causes of morbidity and death globally. In particular, a heart failure remains a major problem that contributes to global mortality. Considerable advancements have been made in conventional pharmacological therapies and coronary intervention surgery for cardiac disorder treatment. However, more than 15% of patients continuously progress to end-stage heart failure and eventually require heart transplantation. Over the past year, numerous numbers of protocols to generate cardiomyocytes (CMCs) from human pluripotent stem cells (hPSCs) have been developed and applied in clinical settings. Number of studies have described the therapeutic effects of hPSCs in animal models and revealed the underlying repair mechanisms of cardiac regeneration. In addition, biomedical engineering technologies have improved the therapeutic potential of hPSC-derived CMCs in vivo. Recently substantial progress has been made in driving the direct differentiation of somatic cells into mature CMCs, wherein an intermediate cellular reprogramming stage can be bypassed. This review provides information on the role of hPSCs in cardiac regeneration and discusses the practical applications of hPSC-derived CMCs; furthermore, it outlines the relevance of directly reprogrammed CMCs in regenerative medicine.
Collapse
Affiliation(s)
- Choon-Soo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
120
|
Pezhouman A, Nguyen NB, Shevtsov A, Qiao R, Ardehali R. In Vitro Generation of Heart Field Specific Cardiomyocytes. Methods Mol Biol 2022; 2429:257-267. [PMID: 35507167 DOI: 10.1007/978-1-0716-1979-7_17] [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] [Indexed: 05/26/2023]
Abstract
Myocardial infarction (MI) can lead to irreversible loss of cardiomyocytes (CMs), primarily localized to the left ventricle (LV) of the heart. The CMs of the LV are predominantly derived from first heart field (FHF) progenitors, whereas the majority of CMs within the right ventricle originate from the second heart field (SHF) during early cardiogenesis. Human embryonic stem cells (hESCs) serve as a valuable source of CMs for understanding early cardiac development and lineage commitment of CMs within these two heart fields that ultimately enable the development of more effective candidates for cell therapy. An ideal candidate may be FHF CMs that share the same ontogeny with the LV CMs that die after MI. We previously generated a double reporter hESC line that utilizes two important cardiac transcription factors, TBX5 and NKX2-5. TBX5 marks FHF progenitors and CMs, while NKX2-5 is expressed in nearly all myocytes of the developing heart. Here, we describe a step-by-step approach to efficiently generate FHF and SHF CMs using this double reporter hESC line. In addition, this approach can be applied to any non-genetically modified hESC lines to enrich FHF and SHF CMs. Obtaining enriched populations of these two CM subtypes provides a platform for downstream comparative analyses and in vitro studies to facilitate a deeper understanding of cardiovascular lineage commitment and the development of more effective candidates for cell therapy to treat diseases or defects that affect specific regions of the heart.
Collapse
Affiliation(s)
- Arash Pezhouman
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ngoc B Nguyen
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Allison Shevtsov
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rong Qiao
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
121
|
From Spheroids to Organoids: The Next Generation of Model Systems of Human Cardiac Regeneration in a Dish. Int J Mol Sci 2021; 22:ijms222413180. [PMID: 34947977 PMCID: PMC8708686 DOI: 10.3390/ijms222413180] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids are tiny, self-organized, three-dimensional tissue cultures that are derived from the differentiation of stem cells. The growing interest in the use of organoids arises from their ability to mimic the biology and physiology of specific tissue structures in vitro. Organoids indeed represent promising systems for the in vitro modeling of tissue morphogenesis and organogenesis, regenerative medicine and tissue engineering, drug therapy testing, toxicology screening, and disease modeling. Although 2D cell cultures have been used for more than 50 years, even for their simplicity and low-cost maintenance, recent years have witnessed a steep rise in the availability of organoid model systems. Exploiting the ability of cells to re-aggregate and reconstruct the original architecture of an organ makes it possible to overcome many limitations of 2D cell culture systems. In vitro replication of the cellular micro-environment of a specific tissue leads to reproducing the molecular, biochemical, and biomechanical mechanisms that directly influence cell behavior and fate within that specific tissue. Lineage-specific self-organizing organoids have now been generated for many organs. Currently, growing cardiac organoid (cardioids) from pluripotent stem cells and cardiac stem/progenitor cells remains an open challenge due to the complexity of the spreading, differentiation, and migration of cardiac muscle and vascular layers. Here, we summarize the evolution of biological model systems from the generation of 2D spheroids to 3D organoids by focusing on the generation of cardioids based on the currently available laboratory technologies and outline their high potential for cardiovascular research.
Collapse
|
122
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
123
|
Engineering the niche to differentiate and deploy cardiovascular cells. Curr Opin Biotechnol 2021; 74:122-128. [PMID: 34861477 DOI: 10.1016/j.copbio.2021.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/23/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022]
Abstract
Applications for stem cells have ranged from therapeutic interventions to more conventional screening and in vitro modeling, but significant limitations to each is due to the lack of maturity from decades old monolayer protocols. While those methods remain the 'gold standard,' newer three-dimensional methods, when combined with engineered niche, stand to significantly improve cell maturity and enable new applications. Here in three parts, we first discuss past methods, and where and why we believe those methods produced suboptimal myocytes. Second, we note how newer methods are moving the field into an era of cell mechanical, electrical, and biological maturity. Finally, we highlight how these improvements will solve issues of scale and engraftment to yield clinical success. It is our conclusion that only through a combination of diverse cell populations and engineered niche will we create an engineered heart tissue with the maturity and vasculature to integrate successfully into a host.
Collapse
|
124
|
Tani H, Tohyama S, Kishino Y, Kanazawa H, Fukuda K. Production of functional cardiomyocytes and cardiac tissue from human induced pluripotent stem cells for regenerative therapy. J Mol Cell Cardiol 2021; 164:83-91. [PMID: 34822838 DOI: 10.1016/j.yjmcc.2021.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/02/2021] [Accepted: 11/17/2021] [Indexed: 12/28/2022]
Abstract
The emergence of human induced pluripotent stem cells (hiPSCs) has revealed the potential for curing end-stage heart failure. Indeed, transplantation of hiPSC-derived cardiomyocytes (hiPSC-CMs) may have applications as a replacement for heart transplantation and conventional regenerative therapies. However, there are several challenges that still must be overcome for clinical applications, including large-scale production of hiPSCs and hiPSC-CMs, elimination of residual hiPSCs, purification of hiPSC-CMs, maturation of hiPSC-CMs, efficient engraftment of transplanted hiPSC-CMs, development of an injection device, and avoidance of post-transplant arrhythmia and immunological rejection. Thus, we developed several technologies based on understanding of the metabolic profiles of hiPSCs and hiPSC derivatives. In this review, we outline how to overcome these hurdles to realize the transplantation of hiPSC-CMs in patients with heart failure and introduce cutting-edge findings and perspectives for future regenerative therapy.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
125
|
Salerno AG, Wanschel ACBA, Dulce RA, Hatzistergos KE, Balkan W, Hare JM. S-nitrosoglutathione reductase (GSNOR) deficiency accelerates cardiomyocyte differentiation of induced pluripotent stem cells. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1. [PMID: 34790975 DOI: 10.20517/jca.2021.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Introduction Induced pluripotent stem cells (iPSCs) provide a model of cardiomyocyte (CM) maturation. Nitric oxide signaling promotes CM differentiation and maturation, although the mechanisms remain controversial. Aim The study tested the hypothesis that in the absence of S-nitrosoglutathione reductase (GSNOR), a denitrosylase regulating protein S-nitrosylation, the resultant increased S-nitrosylation accelerates the differentiation and maturation of iPSC-derived cardiomyocytes (CMs). Methods and Results iPSCs derived from mice lacking GSNOR (iPSCGSNOR-/-) matured faster than wildtype iPSCs (iPSCWT) and demonstrated transient increases in expression of murine Snail Family Transcriptional Repressor 1 gene (Snail), murine Snail Family Transcriptional Repressor 2 gene (Slug) and murine Twist Family BHLH Transcription Factor 1 gene (Twist), transcription factors that promote epithelial-to-mesenchymal transition (EMT) and that are regulated by Glycogen Synthase Kinase 3 Beta (GSK3β). Murine Glycogen Synthase Kinase 3 Beta (Gsk3β) gene exhibited much greater S-nitrosylation, but lower expression in iPSCGSNOR-/-. S-nitrosoglutathione (GSNO)-treated iPSCWT and human (h)iPSCs also demonstrated reduced expression of GSK3β. Nkx2.5 expression, a CM marker, was increased in iPSCGSNOR-/- upon directed differentiation toward CMs on Day 4, whereas murine Brachyury (t), Isl1, and GATA Binding Protein (Gata4) mRNA were decreased, compared to iPSCWT, suggesting that GSNOR deficiency promotes CM differentiation beginning immediately following cell adherence to the culture dish-transitioning from mesoderm to cardiac progenitor. Conclusion Together these findings suggest that increased S-nitrosylation of Gsk3β promotes CM differentiation and maturation from iPSCs. Manipulating the post-translational modification of GSK3β may provide an important translational target and offers new insight into understanding of CM differentiation from pluripotent stem cells. One sentence summary Deficiency of GSNOR or addition of GSNO accelerates early differentiation and maturation of iPSC-cardiomyocytes.
Collapse
Affiliation(s)
- Alessandro G Salerno
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Amarylis C B A Wanschel
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Raul A Dulce
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Konstantinos E Hatzistergos
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
126
|
Silver SE, Barrs RW, Mei Y. Transplantation of Human Pluripotent Stem Cell-Derived Cardiomyocytes for Cardiac Regenerative Therapy. Front Cardiovasc Med 2021; 8:707890. [PMID: 34820426 PMCID: PMC8606657 DOI: 10.3389/fcvm.2021.707890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/20/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and bears an immense economic burden. Late-stage heart failure often requires total heart transplantation; however, due to donor shortages and lifelong immunosuppression, alternative cardiac regenerative therapies are in high demand. Human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells, have emerged as a viable source of human cardiomyocytes for transplantation. Recent developments in several mammalian models of cardiac injury have provided strong evidence of the therapeutic potential of hPSC-derived cardiomyocytes (hPSC-CM), showing their ability to electromechanically integrate with host cardiac tissue and promote functional recovery. In this review, we will discuss recent developments in hPSC-CM differentiation and transplantation strategies for delivery to the heart. We will highlight the mechanisms through which hPSC-CMs contribute to heart repair, review major challenges in successful transplantation of hPSC-CMs, and present solutions that are being explored to address these limitations. We end with a discussion of the clinical use of hPSC-CMs, including hurdles to clinical translation, current clinical trials, and future perspectives on hPSC-CM transplantation.
Collapse
Affiliation(s)
- Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
127
|
Raad FS, Khan TA, Esser TU, Hudson JE, Seth BI, Fujita B, Gandamala R, Tietze LF, Zimmermann WH. Chalcone-Supported Cardiac Mesoderm Induction in Human Pluripotent Stem Cells for Heart Muscle Engineering. ChemMedChem 2021; 16:3300-3305. [PMID: 34309224 PMCID: PMC8597156 DOI: 10.1002/cmdc.202100222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/14/2021] [Indexed: 11/12/2022]
Abstract
Human pluripotent stem cells (hPSCs) hold great promise for applications in cell therapy and drug screening in the cardiovascular field. Bone morphogenetic protein 4 (BMP4) is key for early cardiac mesoderm induction in hPSC and subsequent cardiomyocyte derivation. Small-molecular BMP4 mimetics may help to standardize cardiomyocyte derivation from hPSCs. Based on observations that chalcones can stimulate BMP4 signaling pathways, we hypothesized their utility in cardiac mesoderm induction. To test this, we set up a two-tiered screening strategy, (1) for directed differentiation of hPSCs with commercially available chalcones (4'-hydroxychalcone [4'HC] and Isoliquiritigen) and 24 newly synthesized chalcone derivatives, and (2) a functional screen to assess the propensity of the obtained cardiomyocytes to self-organize into contractile engineered human myocardium (EHM). We identified 4'HC, 4-fluoro-4'-methoxychalcone, and 4-fluoro-4'-hydroxychalcone as similarly effective in cardiac mesoderm induction, but only 4'HC as an effective replacement for BMP4 in the derivation of contractile EHM-forming cardiomyocytes.
Collapse
Affiliation(s)
- Farah S. Raad
- Institute of Pharmacology and ToxicologyUniversity Medical CenterGeorg-August-UniversityGöttingenGermany
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
| | - Taukeer A. Khan
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
- Institute of Organic and Biomolecular ChemistryGeorg-August-UniversityGöttingenGermany
| | - Tilman U. Esser
- Institute of Pharmacology and ToxicologyUniversity Medical CenterGeorg-August-UniversityGöttingenGermany
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
| | - James E. Hudson
- Institute of Pharmacology and ToxicologyUniversity Medical CenterGeorg-August-UniversityGöttingenGermany
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
| | - Bhakti Irene Seth
- Institute of Pharmacology and ToxicologyUniversity Medical CenterGeorg-August-UniversityGöttingenGermany
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
| | - Buntaro Fujita
- Institute of Pharmacology and ToxicologyUniversity Medical CenterGeorg-August-UniversityGöttingenGermany
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
| | - Ravi Gandamala
- Institute of Organic and Biomolecular ChemistryGeorg-August-UniversityGöttingenGermany
| | - Lutz F. Tietze
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
- Institute of Organic and Biomolecular ChemistryGeorg-August-UniversityGöttingenGermany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and ToxicologyUniversity Medical CenterGeorg-August-UniversityGöttingenGermany
- DZHK (German Center for Cardiovascular Research) – Partner site GöttingenGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)Georg-August-UniversityGöttingenGermany
- DZNE (German Center for Neurodegenerative Diseases) – Partner site GöttingenGöttingenGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP)GöttingenGermany
| |
Collapse
|
128
|
Miwa T, Idiris A, Kumagai H. High-throughput 3D Spheroid Formation and Effective Cardiomyocyte Differentiation from Human iPS Cells Using the Microfabric Vessels EZSPHERE TM. Bio Protoc 2021; 11:e4203. [PMID: 34859118 DOI: 10.21769/bioprotoc.4203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 11/02/2022] Open
Abstract
High-throughput 3D spheroid formation from human induced pluripotent stem cells (hiPSCs) can be easily performed using the unique microfabric vessels EZSPHERE, resulting in effective and large scale generation of differentiated cells such as cardiomyocytes or neurons. Such hiPSC-derived cardiomyocytes (hiPSC-CMs) or neurons are very useful in the fields of regenerative medicine or cell-based drug safety tests. Previous studies indicated that 3D spheroids arising from hiPSCs are effectively differentiated into high quality hiPSC-CMs by controlling Wnt signals through utilization of the microfabric vessels EZSPHERE. Here, we describe a simple and highly efficient protocol for generating a large number of uniformly sized hiPSC spheroids and inducing them for cardiac differentiation using the EZSPHERE. This method comprises the collection and dissociation of the spheroids from cardiac differentiation medium, in the middle stage of the whole cardiac differentiation process, and re-seeding the obtained single cells into the EZSPHERE to re-aggregate them into uniform hiPSC-CM spheroids with controlled size. This re-aggregation process promotes non-canonical Wnt signal-related cardiac development and improves the purity and maturity of the hiPSC-CMs generated. Graphic abstract: Overview of cardiac differentiation from iPSCs by spheroid formation and reaggregation using EZSPHERE.
Collapse
Affiliation(s)
- Tatsuaki Miwa
- Consumer Production Division, AGC Techno Glass Co., Ltd., Haibara-gun, Shizuoka, Japan
| | - Alimjan Idiris
- Material Integration Laboratories, Technology General Division, AGC Inc., Yokohama-shi, Kanagawa, Japan
| | | |
Collapse
|
129
|
Murphy SA, Chen EZ, Tung L, Boheler KR, Kwon C. Maturing heart muscle cells: Mechanisms and transcriptomic insights. Semin Cell Dev Biol 2021; 119:49-60. [PMID: 33952430 PMCID: PMC8653577 DOI: 10.1016/j.semcdb.2021.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
Cardiomyocyte (CM) maturation is the transformation of differentiated fetal CMs into adult CMs that involves changes in morphology, cell function and metabolism, and the transcriptome. This process is, however, incomplete and ultimately arrested in pluripotent stem cell-derived CMs (PSC-CMs) in culture, which hinders their broad biomedical application. For this reason, enormous efforts are currently being made with the goal of generating mature PSC-CMs. In this review, we summarize key aspects of maturation observed in native CMs and discuss recent findings on the factors and mechanisms that regulate the process. Particular emphasis is put on transcriptional regulation and single-cell RNA-sequencing analysis that has emerged as a key tool to study time-series gene regulation and to determine the maturation state. We then discuss different biomimetic strategies to enhance PSC-CM maturation and discuss their effects at the single cell transcriptomic and functional levels.
Collapse
Affiliation(s)
- Sean A Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elaine Zhelan Chen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Kenneth R Boheler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute of Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
130
|
Schaniel C, Dhanan P, Hu B, Xiong Y, Raghunandan T, Gonzalez DM, Dariolli R, D'Souza SL, Yadaw AS, Hansen J, Jayaraman G, Mathew B, Machado M, Berger SI, Tripodig J, Najfeld V, Garg J, Miller M, Surlyn CS, Michelis KC, Tangirala NC, Weerahandi H, Thomas DC, Beaumont KG, Sebra R, Mahajan M, Schadt E, Vidovic D, Schürer SC, Goldfarb J, Azeloglu EU, Birtwistle MR, Sobie EA, Kovacic JC, Dubois NC, Iyengar R. A library of induced pluripotent stem cells from clinically well-characterized, diverse healthy human individuals. Stem Cell Reports 2021; 16:3036-3049. [PMID: 34739849 PMCID: PMC8693622 DOI: 10.1016/j.stemcr.2021.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
A library of well-characterized human induced pluripotent stem cell (hiPSC) lines from clinically healthy human subjects could serve as a useful resource of normal controls for in vitro human development, disease modeling, genotype-phenotype association studies, and drug response evaluation. We report generation and extensive characterization of a gender-balanced, racially/ethnically diverse library of hiPSC lines from 40 clinically healthy human individuals who range in age from 22 to 61 years. The hiPSCs match the karyotype and short tandem repeat identities of their parental fibroblasts, and have a transcription profile characteristic of pluripotent stem cells. We provide whole-genome sequencing data for one hiPSC clone from each individual, genomic ancestry determination, and analysis of mendelian disease genes and risks. We document similar transcriptomic profiles, single-cell RNA-sequencing-derived cell clusters, and physiology of cardiomyocytes differentiated from multiple independent hiPSC lines. This extensive characterization makes this hiPSC library a valuable resource for many studies on human biology. A library of induced pluripotent stem cells from 40 healthy human subjects Racially/ethnically diverse subjects of clinically well-characterized health Whole-genome sequencing identifies variants of mild common phenotypes or incomplete penetrance Similar physiology of cardiomyocytes from independent hiPSC clones and individuals
Collapse
Affiliation(s)
- Christoph Schaniel
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Priyanka Dhanan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Bin Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuguang Xiong
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teeya Raghunandan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David M Gonzalez
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rafael Dariolli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sunita L D'Souza
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; St. Jude's Children's Research Hospital, Memphis, TN, USA
| | - Arjun S Yadaw
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jens Hansen
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gomathi Jayaraman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Seth I Berger
- Center for Genetic Medicine Research & Rare Disease Institute, Children's National Hospital, Washington, DC, USA
| | - Joseph Tripodig
- Sema4, Stamford, CT, USA; Department of Pathology, Tumor Cytogenomics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vesna Najfeld
- Department of Pathology, Tumor Cytogenomics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jalaj Garg
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, and The Mount Sinai Hospital, New York, NY, USA; Division of Cardiology, Cardiac Arrhythmia Service, Loma Linda University Health, Loma Linda, CA, USA
| | - Marc Miller
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, and The Mount Sinai Hospital, New York, NY, USA
| | - Colleen S Surlyn
- Department of Medicine, Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA; Southeast Health Center, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Katherine C Michelis
- Department of Medicine, Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA; Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, USA
| | - Neelima C Tangirala
- Department of Medicine, Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA
| | - Himali Weerahandi
- Department of Medicine, Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA; Department of Medicine, Division of General Internal Medicine and Clinical Innovation, NYU Grossman School of Medicine, New York, NY, USA
| | - David C Thomas
- Department of Medicine, Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Hospital, New York, NY, USA
| | - Kristin G Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Milind Mahajan
- St. Jude's Children's Research Hospital, Memphis, TN, USA
| | - Eric Schadt
- St. Jude's Children's Research Hospital, Memphis, TN, USA
| | - Dusica Vidovic
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL, USA
| | - Stephan C Schürer
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL, USA
| | - Joseph Goldfarb
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marc R Birtwistle
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Eric A Sobie
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jason C Kovacic
- Center for Genetic Medicine Research & Rare Disease Institute, Children's National Hospital, Washington, DC, USA; Department of Pathology, Tumor Cytogenomics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Nicole C Dubois
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Ravi Iyengar
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
131
|
Eschenhagen T, Ridders K, Weinberger F. How to repair a broken heart with pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol 2021; 163:106-117. [PMID: 34687723 DOI: 10.1016/j.yjmcc.2021.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 01/14/2023]
Abstract
Heart regeneration addresses a central problem in cardiology, the irreversibility of the loss of myocardium that eventually leads to heart failure. True restoration of heart function can only be achieved by remuscularization, i.e. replacement of lost myocardium by new, force-developing heart muscle. With the availability of principally unlimited human cardiomyocytes from pluripotent stem cells, one option to remuscularize the injured heart is to produce large numbers of cardiomyocytes plus/minus other cardiovascular cell types or progenitors ex vivo and apply them to the heart, either by injection or application as a patch. Exciting progress over the past decade has led to the first clinical applications, but important questions remain. Academic and increasingly corporate activity is ongoing to answer them and optimize the approach to finally develop a true regenerative therapy of heart failure.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | | | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
132
|
Hamad S, Derichsweiler D, Hescheler J, Pfannkuche K. Scalable Generation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2454:145-161. [PMID: 34664217 DOI: 10.1007/7651_2021_395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) can be expanded at limitless scale in vitro and give rise to various organotypic cells, cardiomyocytes (CMs) among them. Advanced protocols shape the differentiation process of pluripotent stem cells by controlled growth factor application. Modulating the Wnt signaling pathway is effective to direct hiPSCs to CMs (hiPSC-CMs) and native growth factors were replaced by small chemical compounds. Here, we describe a refined protocol for scalable generation of hiPSC-CMs that manipulates porcupine and tankyrase sub-pathways of Wnt signaling for tight inhibition of non-canonical Wnt signaling. The approach results in a differentiation efficiency toward hiPSC-CMs of 87 ± 0.9% in stirred bioreactor cultures and yields about 70 million hiPSC-CMs per 100 mL serum free cardiac differentiation medium. The differentiation protocol is easily adapted from 3D to 2D culture and vice versa and has been demonstrated to work with different hiPSC lines.
Collapse
Affiliation(s)
- Sarkawt Hamad
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany.,Biology Department, Faculty of Science, Soran University, Soran, Kurdistan Region, Iraq
| | - Daniel Derichsweiler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Kurt Pfannkuche
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany. .,Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany. .,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany. .,Marga-and-Walter-Boll Laboratory for Cardiac Tissue Engineering, University of Cologne, Cologne, Germany.
| |
Collapse
|
133
|
Abstract
It has been nearly 15 years since the discovery of human-induced pluripotent stem cells (iPSCs). During this time, differentiation methods to targeted cells have dramatically improved, and many types of cells in the human body can be currently generated at high efficiency. In the cardiovascular field, the ability to generate human cardiomyocytes in vitro with the same genetic background as patients has provided a great opportunity to investigate human cardiovascular diseases at the cellular level to clarify the molecular mechanisms underlying the diseases and discover potential therapeutics. Additionally, iPSC-derived cardiomyocytes have provided a powerful platform to study drug-induced cardiotoxicity and identify patients at high risk for the cardiotoxicity; thus, accelerating personalized precision medicine. Moreover, iPSC-derived cardiomyocytes can be sources for cardiac cell therapy. Here, we review these achievements and discuss potential improvements for the future application of iPSC technology in cardiovascular diseases.
Collapse
|
134
|
Rankin SA, Steimle JD, Yang XH, Rydeen AB, Agarwal K, Chaturvedi P, Ikegami K, Herriges MJ, Moskowitz IP, Zorn AM. Tbx5 drives Aldh1a2 expression to regulate a RA-Hedgehog-Wnt gene regulatory network coordinating cardiopulmonary development. eLife 2021; 10:69288. [PMID: 34643182 PMCID: PMC8555986 DOI: 10.7554/elife.69288] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 09/23/2021] [Indexed: 12/14/2022] Open
Abstract
The gene regulatory networks that coordinate the development of the cardiac and pulmonary systems are essential for terrestrial life but poorly understood. The T-box transcription factor Tbx5 is critical for both pulmonary specification and heart development, but how these activities are mechanistically integrated remains unclear. Here using Xenopus and mouse embryos, we establish molecular links between Tbx5 and retinoic acid (RA) signaling in the mesoderm and between RA signaling and sonic hedgehog expression in the endoderm to unveil a conserved RA-Hedgehog-Wnt signaling cascade coordinating cardiopulmonary (CP) development. We demonstrate that Tbx5 directly maintains expression of aldh1a2, the RA-synthesizing enzyme, in the foregut lateral plate mesoderm via an evolutionarily conserved intronic enhancer. Tbx5 promotes posterior second heart field identity in a positive feedback loop with RA, antagonizing a Fgf8-Cyp regulatory module to restrict FGF activity to the anterior. We find that Tbx5/Aldh1a2-dependent RA signaling directly activates shh transcription in the adjacent foregut endoderm through a conserved MACS1 enhancer. Hedgehog signaling coordinates with Tbx5 in the mesoderm to activate expression of wnt2/2b, which induces pulmonary fate in the foregut endoderm. These results provide mechanistic insight into the interrelationship between heart and lung development informing CP evolution and birth defects.
Collapse
Affiliation(s)
- Scott A Rankin
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Jeffrey D Steimle
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Xinan H Yang
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Ariel B Rydeen
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Kunal Agarwal
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Praneet Chaturvedi
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Kohta Ikegami
- Department of Pediatrics, University of Chicago, Chicago, United States
| | | | - Ivan P Moskowitz
- Department of Pediatrics, University of Chicago, Chicago, United States.,Department of Pathology, University of Chicago, Chicago, United States.,Department of Human Genetics, University of Chicago, Chicago, United States
| | - Aaron M Zorn
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,University of Cincinnati, College of Medicine, Department of Pediatrics, Chicago, United States
| |
Collapse
|
135
|
Miyamoto M, Nam L, Kannan S, Kwon C. Heart organoids and tissue models for modeling development and disease. Semin Cell Dev Biol 2021; 118:119-128. [PMID: 33775518 PMCID: PMC8513373 DOI: 10.1016/j.semcdb.2021.03.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
Organoids, or miniaturized organs formed in vitro, hold potential to revolutionize how researchers approach and answer fundamental biological and pathological questions. In the context of cardiac biology, development of a bona fide cardiac organoid enables study of heart development, function, and pathogenesis in a dish, providing insight into the nature of congenital heart disease and offering the opportunity for high-throughput probing of adult heart disease and drug discovery. Recently, multiple groups have reported novel methods for generating in vitro models of the heart; however, there are substantial conceptual and methodological differences. In this review we will evaluate recent cardiac organoid studies through the lens of the core principles of organoid technology: patterned self-organization of multiple cell types resembling the in vivo organ. Based on this, we will classify systems into the following related types of tissues: developmental cardiac organoids, chamber cardiac organoids, microtissues, and engineered heart tissues. Furthermore, we highlight the interventions which allow for organoid formation, such as modulation of highly conserved cardiogenic signaling pathways mediated by developmental morphogens. We expect that consolidation and categorization of existing organoid models will help eliminate confusion in the field and facilitate progress towards creation of an ideal cardiac organoid.
Collapse
Affiliation(s)
- Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States; Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Lucy Nam
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States; Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States; Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
136
|
Rowton M, Guzzetta A, Rydeen AB, Moskowitz IP. Control of cardiomyocyte differentiation timing by intercellular signaling pathways. Semin Cell Dev Biol 2021; 118:94-106. [PMID: 34144893 PMCID: PMC8968240 DOI: 10.1016/j.semcdb.2021.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023]
Abstract
Congenital Heart Disease (CHD), malformations of the heart present at birth, is the most common class of life-threatening birth defect (Hoffman (1995) [1], Gelb (2004) [2], Gelb (2014) [3]). A major research challenge is to elucidate the genetic determinants of CHD and mechanistically link CHD ontogeny to a molecular understanding of heart development. Although the embryonic origins of CHD are unclear in most cases, dysregulation of cardiovascular lineage specification, patterning, proliferation, migration or differentiation have been described (Olson (2004) [4], Olson (2006) [5], Srivastava (2006) [6], Dunwoodie (2007) [7], Bruneau (2008) [8]). Cardiac differentiation is the process whereby cells become progressively more dedicated in a trajectory through the cardiac lineage towards mature cardiomyocytes. Defects in cardiac differentiation have been linked to CHD, although how the complex control of cardiac differentiation prevents CHD is just beginning to be understood. The stages of cardiac differentiation are highly stereotyped and have been well-characterized (Kattman et al. (2011) [9], Wamstad et al. (2012) [10], Luna-Zurita et al. (2016) [11], Loh et al. (2016) [12], DeLaughter et al. (2016) [13]); however, the developmental and molecular mechanisms that promote or delay the transition of a cell through these stages have not been as deeply investigated. Tight temporal control of progenitor differentiation is critically important for normal organ size, spatial organization, and cellular physiology and homeostasis of all organ systems (Raff et al. (1985) [14], Amthor et al. (1998) [15], Kopan et al. (2014) [16]). This review will focus on the action of signaling pathways in the control of cardiomyocyte differentiation timing. Numerous signaling pathways, including the Wnt, Fibroblast Growth Factor, Hedgehog, Bone Morphogenetic Protein, Insulin-like Growth Factor, Thyroid Hormone and Hippo pathways, have all been implicated in promoting or inhibiting transitions along the cardiac differentiation trajectory. Gaining a deeper understanding of the mechanisms controlling cardiac differentiation timing promises to yield insights into the etiology of CHD and to inform approaches to restore function to damaged hearts.
Collapse
|
137
|
Cho SW, Kim HK, Sung JH, Han J. Stage specific transcriptome profiles at cardiac lineage commitment during cardiomyocyte differentiation from mouse and human pluripotent stem cells. BMB Rep 2021. [PMID: 34120677 PMCID: PMC8505231 DOI: 10.5483/bmbrep.2021.54.9.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cardiomyocyte differentiation occurs through complex and finely regulated processes including cardiac lineage commitment and maturation from pluripotent stem cells (PSCs). To gain some insight into the genome-wide characteristics of cardiac lineage commitment, we performed transcriptome analysis on both mouse embryonic stem cells (mESCs) and human induced PSCs (hiPSCs) at specific stages of cardiomyocyte differentiation. Specifically, the gene expression profiles and the protein–protein interaction networks of the mESC-derived platelet-derived growth factor receptor-alpha (PDGFRα)+ cardiac lineage-committed cells (CLCs) and hiPSC-derived kinase insert domain receptor (KDR)+ and PDGFRα+ cardiac progenitor cells (CPCs) at cardiac lineage commitment were compared with those of mesodermal cells and differentiated cardiomyocytes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that the genes significantly upregulated at cardiac lineage commitment were associated with responses to organic substances and external stimuli, extracellular and myocardial contractile components, receptor binding, gated channel activity, PI3K‑AKT signaling, and cardiac hypertrophy and dilation pathways. Protein–protein interaction network analysis revealed that the expression levels of genes that regulate cardiac maturation, heart contraction, and calcium handling showed a consistent increase during cardiac differentiation; however, the expression levels of genes that regulate cell differentiation and multicellular organism development decreased at the cardiac maturation stage following lineage commitment. Additionally, we identified for the first time the protein–protein interaction network connecting cardiac development, the immune system, and metabolism during cardiac lineage commitment in both mESC-derived PDGFRα+ CLCs and hiPSC-derived KDR+PDGFRα+ CPCs. These findings shed light on the regulation of cardiac lineage commitment and the pathogenesis of cardiometabolic diseases.
Collapse
Affiliation(s)
- Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University College of Medicine, Ilsan Paik Hospital Vision 21 Cardiac & Vascular Center, Goyang 10380, Korea
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Ji Hee Sung
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| |
Collapse
|
138
|
Li X, Gao F, Wang X, Liang Q, Bai A, Liu Z, Chen X, Li E, Chen S, Lu C, Qian R, Sun N, Liang P, Xu C. E2A ablation enhances proportion of nodal-like cardiomyocytes in cardiac-specific differentiation of human embryonic stem cells. EBioMedicine 2021; 71:103575. [PMID: 34488017 PMCID: PMC8426208 DOI: 10.1016/j.ebiom.2021.103575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Human sinoatrial cardiomyocytes are essential building blocks for cell therapies of conduction system disorders. However, current differentiation protocols for deriving nodal cardiomyocytes from human pluripotent stem cells (hPSCs) are very inefficient. METHODS By employing the hPSCs to cardiomyocyte (CM) in vitro differentiation system and generating E2A-knockout hESCs using CRISPR/Cas9 gene editing technology, we analyze the functions of E2A in CM differentiation. FINDINGS We found that knockout of the transcription factor E2A substantially increased the proportion of nodal-like cells in hESC-derived CMs. The E2A ablated CMs displayed smaller cell size, increased beating rates, weaker contractile force, and other functional characteristics similar to sinoatrial node (SAN) cells. Transcriptomic analyses indicated that ion channel-encoding genes were up-regulated in E2A ablated CMs. E2A directly bounded to the promoters of genes key to SAN development via conserved E-box motif, and promoted their expression. Unexpect enhanced activity of NOTCH pathway after E2A ablation could also facilate to induct ventricle workingtype CMs reprogramming into SAN-like cells. INTERPRETATION Our study revealed a new role for E2A during directed cardiac differentiation of hESCs and may provide new clues for enhancing induction efficiency of SAN-like cardiomyocytes from hPSCs in the future. FUNDING This work was supported by the NSFC (No.82070391, N.S.; No.81870175 and 81922006, P.L.), the National Key R&D Program of China (2018YFC2000202, N.S.; 2017YFA0103700, P.L.), the Haiju program of National Children's Medical Center EK1125180102, and Innovative research team of high-level local universities in Shanghai and a key laboratory program of the Education Commission of Shanghai Municipality (ZDSYS14005).
Collapse
Affiliation(s)
- Xiuya Li
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, School of Life Sciences and Technology, Tongji University,Shanghai 200092, China
| | - Fei Gao
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaochen Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Qianqian Liang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Aobing Bai
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhuo Liu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xinyun Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ermin Li
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Shanghai Key Lab of Birth Defect, Children's Hospital of Fudan University, Shanghai, 201102, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Fudan University, Shanghai 200032, China.
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China.
| | - Chen Xu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
139
|
Cheng L, Xie M, Qiao W, Song Y, Zhang Y, Geng Y, Xu W, Wang L, Wang Z, Huang K, Dong N, Sun Y. Generation and characterization of cardiac valve endothelial-like cells from human pluripotent stem cells. Commun Biol 2021; 4:1039. [PMID: 34489520 PMCID: PMC8421482 DOI: 10.1038/s42003-021-02571-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 08/18/2021] [Indexed: 12/31/2022] Open
Abstract
The cardiac valvular endothelial cells (VECs) are an ideal cell source that could be used for making the valve organoids. However, few studies have been focused on the derivation of this important cell type. Here we describe a two-step chemically defined xeno-free method for generating VEC-like cells from human pluripotent stem cells (hPSCs). HPSCs were specified to KDR+/ISL1+ multipotent cardiac progenitors (CPCs), followed by differentiation into valve endothelial-like cells (VELs) via an intermediate endocardial cushion cell (ECC) type. Mechanistically, administration of TGFb1 and BMP4 may specify VEC fate by activating the NOTCH/WNT signaling pathways and previously unidentified targets such as ATF3 and KLF family of transcription factors. When seeded onto the surface of the de-cellularized porcine aortic valve (DCV) matrix scaffolds, hPSC-derived VELs exhibit superior proliferative and clonogenic potential than the primary VECs and human aortic endothelial cells (HAEC). Our results show that hPSC-derived valvular cells could be efficiently generated from hPSCs, which might be used as seed cells for construction of valve organoids or next generation tissue engineered heart valves. Cheng et al. provide a detailed characterization of the differentiation of human pluripotent stem cells to valve endothelial cells and their function. Their results show that the valve endothelial-like cells express key markers for valve endothelial cells, exhibiting proliferative and clonogenic potential.
Collapse
Affiliation(s)
- LinXi Cheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - MingHui Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - WeiHua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Song
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - YanYong Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - YingChao Geng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - WeiLin Xu
- Wuhan Textile University, Wuhan, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Department of Cardiovascular Internal Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - NianGuo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - YuHua Sun
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China. .,University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
140
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
141
|
Das S, Nam H, Jang J. 3D bioprinting of stem cell-laden cardiac patch: A promising alternative for myocardial repair. APL Bioeng 2021; 5:031508. [PMID: 34368602 PMCID: PMC8318604 DOI: 10.1063/5.0030353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Stem cell-laden three-dimensional (3D) bioprinted cardiac patches offer an alternative and promising therapeutic and regenerative approach for ischemic cardiomyopathy by reversing scar formation and promoting myocardial regeneration. Numerous studies have reported using either multipotent or pluripotent stem cells or their combination for 3D bioprinting of a cardiac patch with the sole aim of restoring cardiac function by faithfully rejuvenating the cardiomyocytes and associated vasculatures that are lost to myocardial infarction. While many studies have demonstrated success in mimicking cardiomyocytes' behavior, improving cardiac function and providing new hope for regenerating heart post-myocardial infarction, some others have reported contradicting data in apparent ways. Nonetheless, all investigators in the field are speed racing toward determining a potential strategy to effectively treat losses due to myocardial infarction. This review discusses various types of candidate stem cells that possess cardiac regenerative potential, elucidating their applications and limitations. We also brief the challenges of and an update on the implementation of the state-of-the-art 3D bioprinting approach to fabricate cardiac patches and highlight different strategies to implement vascularization and augment cardiac functional properties with respect to electrophysiological similarities to native tissue.
Collapse
Affiliation(s)
- Sanskrita Das
- Department of Convergence IT Engineering, POSTECH, 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Hyoryung Nam
- Department of Convergence IT Engineering, POSTECH, 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Jinah Jang
- Author to whom correspondence should be addressed:
| |
Collapse
|
142
|
Tsuruda M, Morino-Koga S, Ogawa M. Bone morphogenetic protein 4 differently promotes distinct VE-cadherin + precursor stages during the definitive hematopoietic development from embryonic stem cell-derived mesodermal cells. Exp Hematol 2021; 103:40-51.e7. [PMID: 34464660 DOI: 10.1016/j.exphem.2021.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/04/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022]
Abstract
Definitive hematopoietic cells develop from fetal liver kinase 1 (Flk1)+ mesodermal cells during the in vitro differentiation of mouse embryonic stem cells (ESCs). VE-cadherin+CD41-CD45-(V+41-45-) hemogenic endothelial cells (HECs) and VE-cadherin+CD41+CD45- (V+41+45-) cells mediate the definitive hematopoietic development from Flk1+ cells. Bone morphogenetic protein 4 (BMP4) is known to be essential for the formation of mesoderm. However, the role of BMP4 in differentiation of the VE-cadherin+ definitive hematopoietic precursors from the mesoderm has been elusive. We addressed this issue using a co-aggregation culture of ESC-derived Flk1+ cells with OP9 stromal cells. This culture method induced V+41-45- cells, V+41+45- cells, and CD45+ cells from Flk1+ cells. V+41+45- cells possessed potential for erythromyeloid and T-lymphoid differentiation. When Flk1+ cells were cultured in the presence of a high concentration of BMP4, the generation of V+41-45- cells was enhanced. The increase in V+41-45- cells led to the subsequent increase in V+41+45- and CD45+ cells. The addition of BMP4 also increased hematopoietic colony-forming cells of various lineages. Furthermore, BMP4 promoted the expansion of V+41+45- cells independently of the preceding V+41-45- cell stage. These results suggest that BMP4 has promotive effects on the differentiation of V+41-45- HECs from Flk1+ mesodermal cells and the subsequent proliferation of V+41+45- hematopoietic precursors. These findings may provide insights for establishing a culture system to induce definitive hematopoietic stem cells from ESCs.
Collapse
Affiliation(s)
- Mariko Tsuruda
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Saori Morino-Koga
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Minetaro Ogawa
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Chuo-ku, Kumamoto, Japan.
| |
Collapse
|
143
|
Wang W, Lu G, Liu H, Xiong Z, Leung H, Cao R, Pang AL, Su X, Law PWN, Zhao Z, Chen Z, Chan W. Pten Regulates Cardiomyocyte Differentiation by Modulating Non-CG Methylation via Dnmt3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100849. [PMID: 34247447 PMCID: PMC8425920 DOI: 10.1002/advs.202100849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/04/2021] [Indexed: 06/13/2023]
Abstract
The regulation of cardiomyocyte differentiation is a fundamental aspect of cardiac development and regenerative medicine. PTEN plays important roles during embryonic development. However, its role in cardiomyocyte differentiation remains unknown. In this study, a low-cost protocol for cardiomyocyte differentiation from mouse embryonic stem cells (ESCs) is presented and it is shown that Pten deletion potently suppresses cardiomyocyte differentiation. Transcriptome analysis shows that the expression of a series of cardiomyocyte marker genes is downregulated in Pten-/- cardiomyocytes. Pten ablation induces Dnmt3b expression via the AKT/FoxO3a pathway and regulates the expression of a series of imprinted genes, including Igf2. Double knockout of Dnmt3l and Dnmt3b rescues the deficiency of cardiomyocyte differentiation of Pten-/- ESCs. The DNA methylomes from wild-type and Pten-/- embryoid bodies and cardiomyocytes are analyzed by whole-genome bisulfite sequencing. Pten deletion significantly promotes the non-CG (CHG and CHH) methylation levels of genomic DNA during cardiomyocyte differentiation, and the non-CG methylation levels of cardiomyocyte genes and Igf2 are increased in Pten-/- cardiomyocytes. Igf2 or Igf1r deletion also suppresses cardiomyocyte differentiation through the MAPK/ERK signaling pathway, and IGF2 supplementation partially rescues the cardiomyocyte differentiation. Finally, Pten conditional knockout mice are generated and the role of PTEN in cardiomyocyte differentiation is verified in vivo.
Collapse
Affiliation(s)
- Wuming Wang
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Gang Lu
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Hong‐Bin Liu
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Zhiqiang Xiong
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Ho‐Duen Leung
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Ruican Cao
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Alan Lap‐Yin Pang
- R&D DivisionTGD Life Company Limited15W, Hong Kong Science ParkShatinHong KongChina
| | - Xianwei Su
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Patrick Wai Nok Law
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Zhiju Zhao
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Zi‐Jiang Chen
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Wai‐Yee Chan
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| |
Collapse
|
144
|
Wang YF, Liu C, Xu PF. Deciphering and reconstitution of positional information in the human brain development. ACTA ACUST UNITED AC 2021; 10:29. [PMID: 34467458 PMCID: PMC8408296 DOI: 10.1186/s13619-021-00091-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022]
Abstract
Organoid has become a novel in vitro model to research human development and relevant disorders in recent years. With many improvements on the culture protocols, current brain organoids could self-organize into a complicated three-dimensional organization that mimics most of the features of the real human brain at the molecular, cellular, and further physiological level. However, lacking positional information, an important characteristic conveyed by gradients of signaling molecules called morphogens, leads to the deficiency of spatiotemporally regulated cell arrangements and cell–cell interactions in the brain organoid development. In this review, we will overview the role of morphogen both in the vertebrate neural development in vivo as well as the brain organoid culture in vitro, the strategies to apply morphogen concentration gradients in the organoid system and future perspectives of the brain organoid technology.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Zhejiang University and University of Edinburgh, Jiaxing, Zhejiang, China.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Dr, Singapore, 117599, Singapore
| | - Cong Liu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng-Fei Xu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
145
|
Testa G, Di Benedetto G, Passaro F. Advanced Technologies to Target Cardiac Cell Fate Plasticity for Heart Regeneration. Int J Mol Sci 2021; 22:ijms22179517. [PMID: 34502423 PMCID: PMC8431232 DOI: 10.3390/ijms22179517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
The adult human heart can only adapt to heart diseases by starting a myocardial remodeling process to compensate for the loss of functional cardiomyocytes, which ultimately develop into heart failure. In recent decades, the evolution of new strategies to regenerate the injured myocardium based on cellular reprogramming represents a revolutionary new paradigm for cardiac repair by targeting some key signaling molecules governing cardiac cell fate plasticity. While the indirect reprogramming routes require an in vitro engineered 3D tissue to be transplanted in vivo, the direct cardiac reprogramming would allow the administration of reprogramming factors directly in situ, thus holding great potential as in vivo treatment for clinical applications. In this framework, cellular reprogramming in partnership with nanotechnologies and bioengineering will offer new perspectives in the field of cardiovascular research for disease modeling, drug screening, and tissue engineering applications. In this review, we will summarize the recent progress in developing innovative therapeutic strategies based on manipulating cardiac cell fate plasticity in combination with bioengineering and nanotechnology-based approaches for targeting the failing heart.
Collapse
Affiliation(s)
- Gianluca Testa
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
- Interdepartmental Center for Nanotechnology Research—NanoBem, University of Molise, 86100 Campobasso, Italy
| | - Giorgia Di Benedetto
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 80138 Naples, Italy;
| | - Fabiana Passaro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 80138 Naples, Italy;
- Correspondence:
| |
Collapse
|
146
|
Lewis-Israeli YR, Wasserman AH, Gabalski MA, Volmert BD, Ming Y, Ball KA, Yang W, Zou J, Ni G, Pajares N, Chatzistavrou X, Li W, Zhou C, Aguirre A. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat Commun 2021; 12:5142. [PMID: 34446706 PMCID: PMC8390749 DOI: 10.1038/s41467-021-25329-5] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Congenital heart defects constitute the most common human birth defect, however understanding of how these disorders originate is limited by our ability to model the human heart accurately in vitro. Here we report a method to generate developmentally relevant human heart organoids by self-assembly using human pluripotent stem cells. Our procedure is fully defined, efficient, reproducible, and compatible with high-content approaches. Organoids are generated through a three-step Wnt signaling modulation strategy using chemical inhibitors and growth factors. Heart organoids are comparable to age-matched human fetal cardiac tissues at the transcriptomic, structural, and cellular level. They develop sophisticated internal chambers with well-organized multi-lineage cardiac cell types, recapitulate heart field formation and atrioventricular specification, develop a complex vasculature, and exhibit robust functional activity. We also show that our organoid platform can recreate complex metabolic disorders associated with congenital heart defects, as demonstrated by an in vitro model of pregestational diabetes-induced congenital heart defects.
Collapse
Affiliation(s)
- Yonatan R Lewis-Israeli
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Aaron H Wasserman
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Mitchell A Gabalski
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Brett D Volmert
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Yixuan Ming
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Kristen A Ball
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Weiyang Yang
- Division of Biomedical Devices, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Jinyun Zou
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Guangming Ni
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Natalia Pajares
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Wen Li
- Division of Biomedical Devices, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Chao Zhou
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Aitor Aguirre
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
147
|
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
|
148
|
Abstract
Cardiac congenital disabilities are the most common organ malformations, but we still do not understand how they arise in the human embryo. Moreover, although cardiovascular disease is the most common cause of death globally, the development of new therapies is lagging compared with other fields. One major bottleneck hindering progress is the lack of self-organizing human cardiac models that recapitulate key aspects of human heart development, physiology and disease. Current in vitro cardiac three-dimensional systems are either engineered constructs or spherical aggregates of cardiomyocytes and other cell types. Although tissue engineering enables the modeling of some electro-mechanical properties, it falls short of mimicking heart development, morphogenetic defects and many clinically relevant aspects of cardiomyopathies. Here, we review different approaches and recent efforts to overcome these challenges in the field using a new generation of self-organizing embryonic and cardiac organoids.
Collapse
Affiliation(s)
- Pablo Hofbauer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Stefan M Jahnel
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Sasha Mendjan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| |
Collapse
|
149
|
Isolation of Cardiomyocytes Derived from Human Pluripotent Stem Cells Using miRNA Switches. Methods Mol Biol 2021. [PMID: 34302646 DOI: 10.1007/978-1-0716-1484-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The most common method for isolating cells of interest is an antibody method that recognizes cell surface antigens. However, specific surface antigens for many cell types have not been identified. We have developed the microRNA (miRNA)-responsive synthetic mRNA systems (miRNA switches), which isolate target cells based on endogenous miRNA activity. In this chapter, we describe protocols for isolating human pluripotent stem cell (hPSC)-derived cardiomyocytes using miRNA switches with or without cell sorting.
Collapse
|
150
|
Kim YJ, Tamadon A, Kim YY, Kang BC, Ku SY. Epigenetic Regulation of Cardiomyocyte Differentiation from Embryonic and Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:8599. [PMID: 34445302 PMCID: PMC8395249 DOI: 10.3390/ijms22168599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/17/2022] Open
Abstract
With the intent to achieve the best modalities for myocardial cell therapy, different cell types are being evaluated as potent sources for differentiation into cardiomyocytes. Embryonic stem cells and induced pluripotent stem cells have great potential for future progress in the treatment of myocardial diseases. We reviewed aspects of epigenetic mechanisms that play a role in the differentiation of these cells into cardiomyocytes. Cardiomyocytes proliferate during fetal life, and after birth, they undergo permanent terminal differentiation. Upregulation of cardiac-specific genes in adults induces hypertrophy due to terminal differentiation. The repression or expression of these genes is controlled by chromatin structural and epigenetic changes. However, few studies have reviewed and analyzed the epigenetic aspects of the differentiation of embryonic stem cells and induced pluripotent stem cells into cardiac lineage cells. In this review, we focus on the current knowledge of epigenetic regulation of cardiomyocyte proliferation and differentiation from embryonic and induced pluripotent stem cells through histone modification and microRNAs, the maintenance of pluripotency, and its alteration during cardiac lineage differentiation.
Collapse
Affiliation(s)
- Yong-Jin Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul 08308, Korea;
| | - Amin Tamadon
- Department of Marine Stem Cell and Tissue Engineering, Bushehr University of Medical Sciences, Bushehr 14174, Iran;
| | - Yoon-Young Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea;
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea;
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Byeong-Cheol Kang
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea;
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea;
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Korea
| |
Collapse
|