1
|
Finsberg H, Charwat V, Healy KE, Wall ST. Automatic motion estimation with applications to hiPSC-CMs. Biomed Phys Eng Express 2024; 10:065004. [PMID: 39173648 DOI: 10.1088/2057-1976/ad7268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are an effective tool for studying cardiac function and disease, and hold promise for screening drug effects on human tissue. Understanding alterations in motion patterns within these cells is crucial for comprehending how the administration of a drug or the onset of a disease can impact the rhythm of the human heart. However, quantifying motion accurately and efficiently from optical measurements using microscopy is currently time consuming. In this work, we present a unified framework for performing motion analysis on a sequence of microscopically obtained images of tissues consisting of hiPSC-CMs. We provide validation of our developed software using a synthetic test case and show how it can be used to extract displacements and velocities in hiPSC-CM microtissues. Finally, we show how to apply the framework to quantify the effect of an inotropic compound. The described software system is distributed as a python package that is easy to install, well tested and can be integrated into any python workflow.
Collapse
Affiliation(s)
| | | | - Kevin E Healy
- Department of Material Science and Engineering, University of California, Berkeley, CA, United States of America
- Department of Bioengineering, University of California, Berkeley, CA, United States of America
| | - Samuel T Wall
- Simula Research Laboratory, Norway
- Organos, Inc, Berkeley, CA, United States of America
| |
Collapse
|
2
|
van Doorn ECH, Amesz JH, Sadeghi AH, de Groot NMS, Manintveld OC, Taverne YJHJ. Preclinical Models of Cardiac Disease: A Comprehensive Overview for Clinical Scientists. Cardiovasc Eng Technol 2024; 15:232-249. [PMID: 38228811 PMCID: PMC11116217 DOI: 10.1007/s13239-023-00707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
For recent decades, cardiac diseases have been the leading cause of death and morbidity worldwide. Despite significant achievements in their management, profound understanding of disease progression is limited. The lack of biologically relevant and robust preclinical disease models that truly grasp the molecular underpinnings of cardiac disease and its pathophysiology attributes to this stagnation, as well as the insufficiency of platforms that effectively explore novel therapeutic avenues. The area of fundamental and translational cardiac research has therefore gained wide interest of scientists in the clinical field, while the landscape has rapidly evolved towards an elaborate array of research modalities, characterized by diverse and distinctive traits. As a consequence, current literature lacks an intelligible and complete overview aimed at clinical scientists that focuses on selecting the optimal platform for translational research questions. In this review, we present an elaborate overview of current in vitro, ex vivo, in vivo and in silico platforms that model cardiac health and disease, delineating their main benefits and drawbacks, innovative prospects, and foremost fields of application in the scope of clinical research incentives.
Collapse
Affiliation(s)
- Elisa C H van Doorn
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jorik H Amesz
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Amir H Sadeghi
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Natasja M S de Groot
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Yannick J H J Taverne
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Kafili G, Kabir H, Jalali Kandeloos A, Golafshan E, Ghasemi S, Mashayekhan S, Taebnia N. Recent advances in soluble decellularized extracellular matrix for heart tissue engineering and organ modeling. J Biomater Appl 2023; 38:577-604. [PMID: 38006224 PMCID: PMC10676626 DOI: 10.1177/08853282231207216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Abstract
Despite the advent of tissue engineering (TE) for the remodeling, restoring, and replacing damaged cardiovascular tissues, the progress is hindered by the optimal mechanical and chemical properties required to induce cardiac tissue-specific cellular behaviors including migration, adhesion, proliferation, and differentiation. Cardiac extracellular matrix (ECM) consists of numerous structural and functional molecules and tissue-specific cells, therefore it plays an important role in stimulating cell proliferation and differentiation, guiding cell migration, and activating regulatory signaling pathways. With the improvement and modification of cell removal methods, decellularized ECM (dECM) preserves biochemical complexity, and bio-inductive properties of the native matrix and improves the process of generating functional tissue. In this review, we first provide an overview of the latest advancements in the utilization of dECM in in vitro model systems for disease and tissue modeling, as well as drug screening. Then, we explore the role of dECM-based biomaterials in cardiovascular regenerative medicine (RM), including both invasive and non-invasive methods. In the next step, we elucidate the engineering and material considerations in the preparation of dECM-based biomaterials, namely various decellularization techniques, dECM sources, modulation, characterizations, and fabrication approaches. Finally, we discuss the limitations and future directions in fabrication of dECM-based biomaterials for cardiovascular modeling, RM, and clinical translation.
Collapse
Affiliation(s)
- Golara Kafili
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
| | - Hannaneh Kabir
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, USA
| | | | - Elham Golafshan
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
| | - Sara Ghasemi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
4
|
Tang X, Xu R, Wang Y, Chen K, Cui S. TERC haploid cell reprogramming: a novel therapeutic strategy for aplastic anemia. Mol Med 2023; 29:94. [PMID: 37424004 DOI: 10.1186/s10020-023-00691-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023] Open
Abstract
The telomerase RNA component (TERC) gene plays an important role in telomerase-dependent extension and maintenance of the telomeres. In the event of TERC haploinsufficiency, telomere length is often affected; this, in turn, can result in the development of progeria-related diseases such as aplastic anemia (AA) and congenital keratosis. Cell reprogramming can reverse the differentiation process and can, therefore, transform cells into pluripotent stem cells with stronger differentiation and self-renewal abilities; further, cell reprograming can also extend the telomere length of these cells, which may be crucial in the diagnosis and treatment of telomere depletion diseases such as AA. In this study, we summarized the effects of TERC haploid cell reprogramming on telomere length and the correlation between this alteration and the pathogenesis of AA; by investigating the role of cell reprogramming in AA, we aimed to identify novel diagnostic indicators and therapeutic strategies for patients with AA.
Collapse
Affiliation(s)
- Xinyu Tang
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ruirong Xu
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Yan Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Kaiqing Chen
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Siyuan Cui
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
5
|
Meyer KM, Malhotra N, Kwak JS, El Refaey M. Relevance of KCNJ5 in Pathologies of Heart Disease. Int J Mol Sci 2023; 24:10849. [PMID: 37446026 PMCID: PMC10341679 DOI: 10.3390/ijms241310849] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Abnormalities in G-protein-gated inwardly rectifying potassium (GIRK) channels have been implicated in diseased states of the cardiovascular system; however, the role of GIRK4 (Kir3.4) in cardiac physiology and pathophysiology has yet to be completely understood. Within the heart, the KACh channel, consisting of two GIRK1 and two GIRK4 subunits, plays a major role in modulating the parasympathetic nervous system's influence on cardiac physiology. Being that GIRK4 is necessary for the functional KACh channel, KCNJ5, which encodes GIRK4, it presents as a therapeutic target for cardiovascular pathology. Human variants in KCNJ5 have been identified in familial hyperaldosteronism type III, long QT syndrome, atrial fibrillation, and sinus node dysfunction. Here, we explore the relevance of KCNJ5 in each of these diseases. Further, we address the limitations and complexities of discerning the role of KCNJ5 in cardiovascular pathophysiology, as identical human variants of KCNJ5 have been identified in several diseases with overlapping pathophysiology.
Collapse
Affiliation(s)
- Karisa M. Meyer
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nipun Malhotra
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jung seo Kwak
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Mona El Refaey
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Wang Y, Yu M, Hao K, Lei W, Tang M, Hu S. Cardiomyocyte Maturation-the Road is not Obstructed. Stem Cell Rev Rep 2022; 18:2966-2981. [PMID: 35788883 DOI: 10.1007/s12015-022-10407-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 12/29/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) represent one of the most promising ways to treat cardiovascular diseases. High-purity cardiomyocytes (CM) from different cell sources could be obtained at present. However, the immature nature of these cardiomyocytes hinders its further clinical application. From immature to mature state, it involves structural, functional, and metabolic changes in cardiomyocytes. Generally, two types of culturing (2D and 3D) systems have been reported to induce cardiomyocyte maturation. 2D culture mainly achieves the maturation of cardiomyocytes through long-term culture, co-culture, supplementation of small molecule compounds, and the application of biophysical cues. The combined use of biomaterial's surface topography and biophysical cues also facilitates the maturation of cardiomyocytes. Cardiomyocyte maturation is a complex process involving many signaling pathways, and current methods fail to fully reproduce this process. Therefore, analyzing the signaling pathway network related to the maturation and producing hPSC-CMs with adult-like phenotype is a challenge. In this review, we summarized the structural and functional differences between hPSC-CMs and mature cardiomyocytes, and introduced various methods to induce cardiomyocyte maturation.
Collapse
Affiliation(s)
- Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Kaili Hao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Mingliang Tang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
7
|
Dou W, Malhi M, Zhao Q, Wang L, Huang Z, Law J, Liu N, Simmons CA, Maynes JT, Sun Y. Microengineered platforms for characterizing the contractile function of in vitro cardiac models. MICROSYSTEMS & NANOENGINEERING 2022; 8:26. [PMID: 35299653 PMCID: PMC8882466 DOI: 10.1038/s41378-021-00344-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 05/08/2023]
Abstract
Emerging heart-on-a-chip platforms are promising approaches to establish cardiac cell/tissue models in vitro for research on cardiac physiology, disease modeling and drug cardiotoxicity as well as for therapeutic discovery. Challenges still exist in obtaining the complete capability of in situ sensing to fully evaluate the complex functional properties of cardiac cell/tissue models. Changes to contractile strength (contractility) and beating regularity (rhythm) are particularly important to generate accurate, predictive models. Developing new platforms and technologies to assess the contractile functions of in vitro cardiac models is essential to provide information on cell/tissue physiologies, drug-induced inotropic responses, and the mechanisms of cardiac diseases. In this review, we discuss recent advances in biosensing platforms for the measurement of contractile functions of in vitro cardiac models, including single cardiomyocytes, 2D monolayers of cardiomyocytes, and 3D cardiac tissues. The characteristics and performance of current platforms are reviewed in terms of sensing principles, measured parameters, performance, cell sources, cell/tissue model configurations, advantages, and limitations. In addition, we highlight applications of these platforms and relevant discoveries in fundamental investigations, drug testing, and disease modeling. Furthermore, challenges and future outlooks of heart-on-a-chip platforms for in vitro measurement of cardiac functional properties are discussed.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Qili Zhao
- Institute of Robotics and Automatic Information System and the Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin, 300350 China
| | - Li Wang
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353 China
| | - Zongjie Huang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Na Liu
- School of Mechatronics Engineering and Automation, Shanghai University, Shanghai, 200444 China
| | - Craig A. Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9 Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1 Canada
| | - Jason T. Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8 Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9 Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 3G4 Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5T 3A1 Canada
| |
Collapse
|
8
|
Ye L, Yu Y, Zhao ZA, Zhao D, Ni X, Wang Y, Fang X, Yu M, Wang Y, Tang JM, Chen Y, Shen Z, Lei W, Hu S. Patient-specific iPSC-derived cardiomyocytes reveal abnormal regulation of FGF16 in a familial atrial septal defect. Cardiovasc Res 2022; 118:859-871. [PMID: 33956078 DOI: 10.1093/cvr/cvab154] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Congenital heart disease (CHD) frequently occurs in newborns due to abnormal formation of the heart or major blood vessels. Mutations in the GATA4 gene, which encodes GATA binding protein 4, are responsible for atrial septal defect (ASD), a common CHD. This study aims to gain insights into the molecular mechanisms of CHD using human-induced pluripotent stem cells (iPSCs) from a family cohort with ASD. METHODS AND RESULTS Patient-specific iPSCs possess the same genetic information as the donor and can differentiate into various cell types from all three germ layers in vitro, thus presenting a promising approach for disease modelling and molecular mechanism research. Here, we generated a patient-specific iPSC line (iPSC-G4T280M) from a family cohort carrying a hereditary ASD mutation in GATA4 gene (T280M), as well as a human embryonic stem cell line (ESC-G4T280M) carrying the isogenic T280M mutation using the CRISPR/Cas9 genome editing method. The GATA4-mutant iPSCs and ESCs were then differentiated into cardiomyocytes (CMs) to model GATA4 mutation-associated ASD. We observed an obvious defect in cell proliferation in cardiomyocytes derived from both GATA4T280M-mutant iPSCs (iPSC-G4T280M-CMs) and ESCs (ESC-G4T280M-CMs), while the impaired proliferation ability of iPSC-G4T280M-CMs could be restored by gene correction. Integrated analysis of RNA-Seq and ChIP-Seq data indicated that FGF16 is a direct target of wild-type GATA4. However, the T280M mutation obstructed GATA4 occupancy at the FGF16 promoter region, leading to impaired activation of FGF16 transcription. Overexpression of FGF16 in GATA4-mutant cardiomyocytes rescued the cell proliferation defect. The direct relationship between GATA4T280M and ASD was demonstrated in a human iPSC model for the first time. CONCLUSIONS In summary, our study revealed the molecular mechanism of the GATA4T280M mutation in ASD. Understanding the roles of the GATA4-FGF16 axis in iPSC-CMs will shed light on heart development and provide novel insights for the treatment of ASD and other CHD disorders.
Collapse
Affiliation(s)
- Lingqun Ye
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - You Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Zhen-Ao Zhao
- Institute of Microcirculation & Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou 075000, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou 075000, China
| | - Dandan Zhao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Xuan Ni
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Yong Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Xing Fang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Yongming Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200432, China
| | - Jun-Ming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, Shiyan 442000, China
| | - Ying Chen
- Central Lab, the Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215000, China
| |
Collapse
|
9
|
Liu C, Feng X, Li G, Gokulnath P, Xiao J. Generating 3D human cardiac constructs from pluripotent stem cells. EBioMedicine 2022; 76:103813. [PMID: 35093634 PMCID: PMC8804169 DOI: 10.1016/j.ebiom.2022.103813] [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: 10/20/2021] [Revised: 12/11/2021] [Accepted: 12/29/2021] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cell (hPSC) technology has offered nearly infinite opportunities to model all kinds of human diseases in vitro. Cardiomyocytes derived from hPSCs have proved to be efficient tools for cardiac disease modeling, drug screening and pathological mechanism studies. In this review, we discuss the advantages and limitations of 2D hPSC-cardiomyocyte (hPSC-CM) system, and introduce the recent development of three-dimensional (3D) culture platforms derived from hPSCs. Although the development of bioengineering technologies has greatly improved 3D platform construction, there are certainly challenges and room for development for further in-depth research.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xing Feng
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
10
|
Dessouki FBA, Singal PK, Singla DK. Rat-Induced Pluripotent Stem Cells-Derived Cardiac Myocytes in a Cell Culture Dish. Methods Mol Biol 2022; 2520:37-51. [PMID: 34128207 PMCID: PMC10716860 DOI: 10.1007/7651_2021_406] [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: 01/24/2023]
Abstract
Induced pluripotent stem (iPS) cells are genetically reprogrammed somatic cells that exhibit embryonic stem cell-like characteristics such as self-renewal and pluripotency. These cells have broad differentiation capability to convert into diverse cell types that make up the primary germ layers during embryonic development. iPS cells can spontaneously differentiate and form cell aggregates termed embryoid bodies (EBs) in the absence of differentiation inhibitory factors. Unlike other methods used to generate EBs, "the hanging drop" method offers reproducibility and homogeneity from a set number of iPS cells. As such, we describe the differentiation of rat-induced pluripotent stem cells into cardiac myocytes in vitro using the hanging drop method. Both the confirmation and identification of the cardiac myocytes are done using immunocytochemistry, RT-PCR, Western Blot, and Flow Cytometry. Briefly, a specific number of iPS cells are placed in droplets on the lid of culture dishes and incubated for 2 days, yielding embryoid bodies, which are suspended and plated. Spontaneous beating of cardiomyocytes can be seen 7-14 days after the plating of EBs and specific cardiac markers can be observed through identification assays.
Collapse
Affiliation(s)
- Fatima Bianca A Dessouki
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
11
|
Aboul-Soud MAM, Alzahrani AJ, Mahmoud A. Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies, Disease Modelling and Drug Screening. Cells 2021; 10:cells10092319. [PMID: 34571968 PMCID: PMC8467501 DOI: 10.3390/cells10092319] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has made an invaluable contribution to the field of regenerative medicine, paving way for identifying the true potential of human embryonic stem cells (ESCs). Since the controversy around ethicality of ESCs continue to be debated, iPSCs have been used to circumvent the process around destruction of the human embryo. The use of iPSCs have transformed biological research, wherein increasing number of studies are documenting nuclear reprogramming strategies to make them beneficial models for drug screening as well as disease modelling. The flexibility around the use of iPSCs include compatibility to non-invasive harvesting, and ability to source from patients with rare diseases. iPSCs have been widely used in cardiac disease modelling, studying inherited arrhythmias, neural disorders including Alzheimer’s disease, liver disease, and spinal cord injury. Extensive research around identifying factors that are involved in maintaining the identity of ESCs during induction of pluripotency in somatic cells is undertaken. The focus of the current review is to detail all the clinical translation research around iPSCs and the strength of its ever-growing potential in the clinical space.
Collapse
Affiliation(s)
- Mourad A. M. Aboul-Soud
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
- Correspondence:
| | - Alhusain J. Alzahrani
- Department of Clinical Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39524, Saudi Arabia;
| | - Amer Mahmoud
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| |
Collapse
|
12
|
Wang Y, Lei W, Yang J, Ni X, Ye L, Shen Z, Hu S. The updated view on induced pluripotent stem cells for cardiovascular precision medicine. Pflugers Arch 2021; 473:1137-1149. [DOI: 10.1007/s00424-021-02530-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
|
13
|
Jæger KH, Wall S, Tveito A. Computational prediction of drug response in short QT syndrome type 1 based on measurements of compound effect in stem cell-derived cardiomyocytes. PLoS Comput Biol 2021; 17:e1008089. [PMID: 33591962 PMCID: PMC7909705 DOI: 10.1371/journal.pcbi.1008089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/26/2021] [Accepted: 12/20/2020] [Indexed: 12/20/2022] Open
Abstract
Short QT (SQT) syndrome is a genetic cardiac disorder characterized by an abbreviated QT interval of the patient's electrocardiogram. The syndrome is associated with increased risk of arrhythmia and sudden cardiac death and can arise from a number of ion channel mutations. Cardiomyocytes derived from induced pluripotent stem cells generated from SQT patients (SQT hiPSC-CMs) provide promising platforms for testing pharmacological treatments directly in human cardiac cells exhibiting mutations specific for the syndrome. However, a difficulty is posed by the relative immaturity of hiPSC-CMs, with the possibility that drug effects observed in SQT hiPSC-CMs could be very different from the corresponding drug effect in vivo. In this paper, we apply a multistep computational procedure for translating measured drug effects from these cells to human QT response. This process first detects drug effects on individual ion channels based on measurements of SQT hiPSC-CMs and then uses these results to estimate the drug effects on ventricular action potentials and QT intervals of adult SQT patients. We find that the procedure is able to identify IC50 values in line with measured values for the four drugs quinidine, ivabradine, ajmaline and mexiletine. In addition, the predicted effect of quinidine on the adult QT interval is in good agreement with measured effects of quinidine for adult patients. Consequently, the computational procedure appears to be a useful tool for helping predicting adult drug responses from pure in vitro measurements of patient derived cell lines.
Collapse
MESH Headings
- Action Potentials/drug effects
- Adult
- Ajmaline/pharmacology
- Algorithms
- Anti-Arrhythmia Agents/pharmacology
- Arrhythmias, Cardiac/drug therapy
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Cell Line
- Computational Biology
- Drug Evaluation, Preclinical/methods
- Drug Evaluation, Preclinical/statistics & numerical data
- ERG1 Potassium Channel/genetics
- Electrocardiography
- Heart Conduction System/abnormalities
- Heart Conduction System/physiopathology
- Heart Defects, Congenital/drug therapy
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/physiopathology
- Humans
- In Vitro Techniques
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/physiology
- Ivabradine/pharmacology
- Mexiletine/pharmacology
- Models, Cardiovascular
- Mutation
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/physiology
- Quinidine/pharmacology
- Translational Research, Biomedical
Collapse
Affiliation(s)
| | | | - Aslak Tveito
- Simula Research Laboratory, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Miao S, Zhao D, Wang X, Ni X, Fang X, Yu M, Ye L, Yang J, Wu H, Han X, Qu L, Li L, Lan F, Shen Z, Lei W, Zhao ZA, Hu S. Retinoic acid promotes metabolic maturation of human Embryonic Stem Cell-derived Cardiomyocytes. Theranostics 2020; 10:9686-9701. [PMID: 32863954 PMCID: PMC7449904 DOI: 10.7150/thno.44146] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiomyocytes differentiated from human embryonic stem cells (hESCs) represent a promising cell source for heart repair, disease modeling and drug testing. However, improving the differentiation efficiency and maturation of hESC-derived cardiomyocytes (hESC-CMs) is still a major concern. Retinoic acid (RA) signaling plays multiple roles in heart development. However, the effects of RA on cardiomyocyte differentiation efficiency and maturation are still unknown. Methods: RA was added at different time intervals to identify the best treatment windows for cardiomyocyte differentiation and maturation. The efficiency of cardiomyocyte differentiation was detected by quantitative real-time PCR and flow cytometry. Cardiomyocytes maturation was detected by immunofluorescence staining, metabolic assays and patch clamp to verify structural, metabolic and electrophysiological maturation, respectively. RNA sequencing was used for splicing analysis. Results: We found that RA treatment at the lateral mesoderm stage (days 2-4) significantly improved cardiomyocyte differentiation, as evidenced by the upregulation of TNNT2, NKX2.5 and MYH6 on day 10 of differentiation. In addition, flow cytometry showed that the proportion of differentiated cardiomyocytes in the RA-treated group was significantly higher than that in control group. RA treatment on days 15-20 increased cardiomyocyte area, sarcomere length, multinucleation and mitochondrial copy number. RNA sequencing revealed RA promoted RNA isoform switch to the maturation-related form. Meanwhile, RA promoted electrophysiological maturation and calcium handling of hESC-CMs. Importantly, RA-treated cardiomyocytes showed decreased glycolysis and enhanced mitochondrial oxidative phosphorylation, with the increased utilization of fatty acid and exogenous pyruvate but not glutamine. Conclusion: Our data indicated that RA treatment at an early time window (days 2-4) promotes the efficiency of cardiomyocyte differentiation and that RA treatment post beating (days 15-20) promotes cardiomyocyte maturation. The biphasic effects of RA provide new insights for improving cardiomyocyte differentiation and quality.
Collapse
|
15
|
Gong Y, Chen Z, Yang L, Ai X, Yan B, Wang H, Qiu L, Tan Y, Witman N, Wang W, Zhao Y, Fu W. Intrinsic Color Sensing System Allows for Real-Time Observable Functional Changes on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. ACS NANO 2020; 14:8232-8246. [PMID: 32609489 DOI: 10.1021/acsnano.0c01745] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stem-cell based in vitro differentiation for disease modeling offers great value to explore the molecular and functional underpinnings driving many types of cardiomyopathy and congenital heart diseases. Nevertheless, one major caveat in the application of in vitro differentiation of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (hiPSC-CMs) involves the immature phenotype of the CMs. Most of the existing methods need complex apparatus and require laborious procedures in order to monitor the cardiac differentiation/maturation process and often result in cell death. Here we developed an intrinsic color sensing system utilizing a microgroove structural color methacrylated gelatin film, which allows us to monitor the cardiac differentiation process of hiPSC-derived cardiac progenitor cells in real time. Subsequently this system can be employed as an assay system to live monitor induced functional changes on hiPSC-CMs stemming from drug treatment, the effects of which are simply revealed through color diversity. Our research shows that early intervention of cardiac differentiation through simple physical cues can enhance cardiac differentiation and maturation to some extent. Our system also simplifies the previous complex experimental processes for evaluating the physiological effects of successful differentiation and drug treatment and lays a solid foundation for future transformational applications.
Collapse
Affiliation(s)
- Yiqi Gong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Zhuoyue Chen
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Li Yang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai 200032, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Bingqian Yan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Liya Qiu
- Shanghai Institute of Technical Physics of the Chinese Academy of Sciences, Shanghai 200083, China
| | - Yao Tan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Nevin Witman
- Department of Medicine and Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Wei Wang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| |
Collapse
|
16
|
Studying ALS: Current Approaches, Effect on Potential Treatment Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1241:195-217. [PMID: 32383122 DOI: 10.1007/978-3-030-41283-8_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is one of the most common neurodegenerative diseases, characterized by inevitable progressive paralysis. To date, only two disease modifying therapeutic options are available for the patients with ALS, although they show very modest effect on disease course. The main reason of failure in the field of pharmacological correction of ALS is inability to untangle complex relationships taking place during ALS initiation and progression. Traditional methods of research, based on morphology or transgenic animal models studying provided lots of information about ALS throughout the years. However, translation of these results to humans was unsuccessful due to incomplete recapitulation of molecular pathology and overall inadequacy of the models used in the research.In this review we summarize current knowledge regarding ALS molecular pathology with depiction of novel methods applied recently for the studies. Furthermore we describe present and potential treatment strategies that are based on the recent findings in ALS disease mechanisms.
Collapse
|
17
|
Computational translation of drug effects from animal experiments to human ventricular myocytes. Sci Rep 2020; 10:10537. [PMID: 32601303 PMCID: PMC7324560 DOI: 10.1038/s41598-020-66910-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 12/20/2022] Open
Abstract
Using animal cells and tissues as precise measuring devices for developing new drugs presents a long-standing challenge for the pharmaceutical industry. Despite the very significant resources that continue to be dedicated to animal testing of new compounds, only qualitative results can be obtained. This often results in both false positives and false negatives. Here, we show how the effect of drugs applied to animal ventricular myocytes can be translated, quantitatively, to estimate a number of different effects of the same drug on human cardiomyocytes. We illustrate and validate our methodology by translating, from animal to human, the effect of dofetilide applied to dog cardiomyocytes, the effect of E-4031 applied to zebrafish cardiomyocytes, and, finally, the effect of sotalol applied to rabbit cardiomyocytes. In all cases, the accuracy of our quantitative estimates are demonstrated. Our computations reveal that, in principle, electrophysiological data from testing using animal ventricular myocytes, can give precise, quantitative estimates of the effect of new compounds on human cardiomyocytes.
Collapse
|
18
|
Jæger KH, Charwat V, Charrez B, Finsberg H, Maleckar MM, Wall S, Healy KE, Tveito A. Improved Computational Identification of Drug Response Using Optical Measurements of Human Stem Cell Derived Cardiomyocytes in Microphysiological Systems. Front Pharmacol 2020; 10:1648. [PMID: 32116671 PMCID: PMC7029356 DOI: 10.3389/fphar.2019.01648] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/16/2019] [Indexed: 11/13/2022] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) hold great potential for drug screening applications. However, their usefulness is limited by the relative immaturity of the cells' electrophysiological properties as compared to native cardiomyocytes in the adult human heart. In this work, we extend and improve on methodology to address this limitation, building on previously introduced computational procedures which predict drug effects for adult cells based on changes in optical measurements of action potentials and Ca2+ transients made in stem cell derived cardiac microtissues. This methodology quantifies ion channel changes through the inversion of data into a mathematical model, and maps this response to an adult phenotype through the assumption of functional invariance of fundamental intracellular and membrane channels during maturation. Here, we utilize an updated action potential model to represent both hiPSC-CMs and adult cardiomyocytes, apply an IC50-based model of dose-dependent drug effects, and introduce a continuation-based optimization algorithm for analysis of dose escalation measurements using five drugs with known effects. The improved methodology can identify drug induced changes more efficiently, and quantitate important metrics such as IC50 in line with published values. Consequently, the updated methodology is a step towards employing computational procedures to elucidate drug effects in adult cardiomyocytes for new drugs using stem cell-derived experimental tissues.
Collapse
Affiliation(s)
| | - Verena Charwat
- Department of Bioengineering, College of Engineering, University of California, Berkeley, CA, United States
| | - Bérénice Charrez
- Department of Bioengineering, College of Engineering, University of California, Berkeley, CA, United States
| | - Henrik Finsberg
- Department of Scientific Computing, Simula Research Laboratory, Oslo, Norway
| | - Mary M. Maleckar
- Department of Scientific Computing, Simula Research Laboratory, Oslo, Norway
| | - Samuel Wall
- Department of Scientific Computing, Simula Research Laboratory, Oslo, Norway
| | - Kevin E. Healy
- Department of Bioengineering, College of Engineering, University of California, Berkeley, CA, United States
- Department of Materials Science and Engineering, College of Engineering, University of California, Berkeley, CA, United States
| | - Aslak Tveito
- Department of Scientific Computing, Simula Research Laboratory, Oslo, Norway
| |
Collapse
|
19
|
Padrón-Barthe L, Villalba-Orero M, Gómez-Salinero JM, Domínguez F, Román M, Larrasa-Alonso J, Ortiz-Sánchez P, Martínez F, López-Olañeta M, Bonzón-Kulichenko E, Vázquez J, Martí-Gómez C, Santiago DJ, Prados B, Giovinazzo G, Gómez-Gaviro MV, Priori S, Garcia-Pavia P, Lara-Pezzi E. Severe Cardiac Dysfunction and Death Caused by Arrhythmogenic Right Ventricular Cardiomyopathy Type 5 Are Improved by Inhibition of Glycogen Synthase Kinase-3β. Circulation 2019; 140:1188-1204. [PMID: 31567019 PMCID: PMC6784777 DOI: 10.1161/circulationaha.119.040366] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy/arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disease characterized by fibrofatty replacement of the myocardium, resulting in heart failure and sudden cardiac death. The most aggressive arrhythmogenic cardiomyopathy/ARVC subtype is ARVC type 5 (ARVC5), caused by a p.S358L mutation in TMEM43 (transmembrane protein 43). The function and localization of TMEM43 are unknown, as is the mechanism by which the p.S358L mutation causes the disease. Here, we report the characterization of the first transgenic mouse model of ARVC5. METHODS We generated transgenic mice overexpressing TMEM43 in either its wild-type or p.S358L mutant (TMEM43-S358L) form in postnatal cardiomyocytes under the control of the α-myosin heavy chain promoter. RESULTS We found that mice expressing TMEM43-S358L recapitulate the human disease and die at a young age. Mutant TMEM43 causes cardiomyocyte death and severe fibrofatty replacement. We also demonstrate that TMEM43 localizes at the nuclear membrane and interacts with emerin and β-actin. TMEM43-S358L shows partial delocalization to the cytoplasm, reduced interaction with emerin and β-actin, and activation of glycogen synthase kinase-3β (GSK3β). Furthermore, we show that targeting cardiac fibrosis has no beneficial effect, whereas overexpression of the calcineurin splice variant calcineurin Aβ1 results in GSK3β inhibition and improved cardiac function and survival. Similarly, treatment of TMEM43 mutant mice with a GSK3β inhibitor improves cardiac function. Finally, human induced pluripotent stem cells bearing the p.S358L mutation also showed contractile dysfunction that was partially restored after GSK3β inhibition. CONCLUSIONS Our data provide evidence that TMEM43-S358L leads to sustained cardiomyocyte death and fibrofatty replacement. Overexpression of calcineurin Aβ1 in TMEM43 mutant mice or chemical GSK3β inhibition improves cardiac function and increases mice life span. Our results pave the way toward new therapeutic approaches for ARVC5.
Collapse
Affiliation(s)
- Laura Padrón-Barthe
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - María Villalba-Orero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Jesús M Gómez-Salinero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Fernando Domínguez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.)
| | - Marta Román
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.)
| | - Javier Larrasa-Alonso
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Paula Ortiz-Sánchez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Fernando Martínez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Marina López-Olañeta
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Elena Bonzón-Kulichenko
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Carlos Martí-Gómez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Demetrio J Santiago
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Belén Prados
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Giovanna Giovinazzo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - María Victoria Gómez-Gaviro
- Departamento de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain (M.V.G.-G.).,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain (M.V.G.-G.)
| | - Silvia Priori
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.).,Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy (S.P.)
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.).,Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain (P.G.-P.).,Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain (P.G.-P.)
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,Faculty of Medicine, National Heart & Lung Institute, Imperial College London, UK (E.L.-P.)
| |
Collapse
|
20
|
Schneider O, Zeifang L, Fuchs S, Sailer C, Loskill P. User-Friendly and Parallelized Generation of Human Induced Pluripotent Stem Cell-Derived Microtissues in a Centrifugal Heart-on-a-Chip. Tissue Eng Part A 2019; 25:786-798. [PMID: 30968738 PMCID: PMC6535963 DOI: 10.1089/ten.tea.2019.0002] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/25/2019] [Indexed: 01/11/2023] Open
Abstract
IMPACT STATEMENT With the ultimate goal in tissue engineering of approaching in vivo functionality as closely as possible, organ-on-a-chip (OoC) systems provide unprecedented game-changing opportunities by enabling creation of perfused three-dimensional tissues. Most of the recently developed OoC systems, however, require complex handling steps. Hence, a large gap still exists between technology development and collection of valuable biological data in a standardized medium- or high-throughput manner. The system presented here bridges this gap by providing a user-friendly framework for the parallelized creation of multiple physiologically relevant tissues, which could be applicable in every laboratory without additional equipment.
Collapse
Affiliation(s)
- Oliver Schneider
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Lisa Zeifang
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Stefanie Fuchs
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Carla Sailer
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Peter Loskill
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Faculty of Medicine, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|