1
|
Botti S, Bartolucci C, Altomare C, Paci M, Barile L, Krause R, Pavarino LF, Severi S. A novel ionic model for matured and paced atrial-like human iPSC-CMs integrating I Kur and I KCa currents. Comput Biol Med 2024; 180:108899. [PMID: 39106668 DOI: 10.1016/j.compbiomed.2024.108899] [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: 05/03/2024] [Revised: 06/25/2024] [Accepted: 07/14/2024] [Indexed: 08/09/2024]
Abstract
This work introduces the first atrial-specific in-silico human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) model, based on a set of phenotype-specific IKur,IKCa and IK1 membrane currents. This model is built on novel in-vitro experimental data recently published by some of the co-authors to simulate the paced action potential of matured atrial-like hiPSC-CMs. The model consists of a system of stiff ordinary differential equations depending on several parameters, which have been tuned by automatic optimization techniques to closely match selected experimental biomarkers. The new model effectively simulates the electronic in-vitro hiPSC-CMs maturation process, transitioning from an unstable depolarized membrane diastolic potential to a stable hyperpolarized resting potential, and exhibits spontaneous firing activity in unpaced conditions. Moreover, our model accurately reflects the experimental rate dependence data at different cycle length and demonstrates the expected response to a specific current blocker. This atrial-specific in-silico model provides a novel computational tool for electrophysiological studies of cardiac stem cells and their applications to drug evaluation and atrial fibrillation treatment.
Collapse
Affiliation(s)
- Sofia Botti
- Euler Institute, Faculty of Informatics, Università della Svizzera Italiana, Lugano, 6900, Switzerland; Department of Mathematics "Felice Casorati", University of Pavia, Pavia, 27100, Italy.
| | - Chiara Bartolucci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, 47521, Italy
| | - Claudia Altomare
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, 6900, Switzerland; Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, 6500, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, 6900, Switzerland
| | - Michelangelo Paci
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, 47521, Italy
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale, Lugano, 6900, Switzerland; Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, 6500, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, 6900, Switzerland
| | - Rolf Krause
- Euler Institute, Faculty of Informatics, Università della Svizzera Italiana, Lugano, 6900, Switzerland; Faculty of Mathematics and Informatics, UniDistance, Brig, 3900, Switzerland
| | - Luca Franco Pavarino
- Department of Mathematics "Felice Casorati", University of Pavia, Pavia, 27100, Italy
| | - Stefano Severi
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, 47521, Italy
| |
Collapse
|
2
|
Farboud SP, Fathi E, Valipour B, Farahzadi R. Toward the latest advancements in cardiac regeneration using induced pluripotent stem cells (iPSCs) technology: approaches and challenges. J Transl Med 2024; 22:783. [PMID: 39175068 PMCID: PMC11342568 DOI: 10.1186/s12967-024-05499-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
A novel approach to treating heart failures was developed with the introduction of iPSC technology. Knowledge in regenerative medicine, developmental biology, and the identification of illnesses at the cellular level has exploded since the discovery of iPSCs. One of the most frequent causes of mortality associated with cardiovascular disease is the loss of cardiomyocytes (CMs), followed by heart failure. A possible treatment for heart failure involves restoring cardiac function and replacing damaged tissue with healthy, regenerated CMs. Significant strides in stem cell biology during the last ten years have transformed the in vitro study of human illness and enhanced our knowledge of the molecular pathways underlying human disease, regenerative medicine, and drug development. We seek to examine iPSC advancements in disease modeling, drug discovery, iPSC-Based cell treatments, and purification methods in this article.
Collapse
Affiliation(s)
- Seyedeh Parya Farboud
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Gao H, Wang Z, Yang F, Wang X, Wang S, Zhang Q, Liu X, Sun Y, Kong J, Yao J. Graphene-integrated mesh electronics with converged multifunctionality for tracking multimodal excitation-contraction dynamics in cardiac microtissues. Nat Commun 2024; 15:2321. [PMID: 38485708 PMCID: PMC10940632 DOI: 10.1038/s41467-024-46636-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024] Open
Abstract
Cardiac microtissues provide a promising platform for disease modeling and developmental studies, which require the close monitoring of the multimodal excitation-contraction dynamics. However, no existing assessing tool can track these multimodal dynamics across the live tissue. We develop a tissue-like mesh bioelectronic system to track these multimodal dynamics. The mesh system has tissue-level softness and cell-level dimensions to enable stable embedment in the tissue. It is integrated with an array of graphene sensors, which uniquely converges both bioelectrical and biomechanical sensing functionalities in one device. The system achieves stable tracking of the excitation-contraction dynamics across the tissue and throughout the developmental process, offering comprehensive assessments for tissue maturation, drug effects, and disease modeling. It holds the promise to provide more accurate quantification of the functional, developmental, and pathophysiological states in cardiac tissues, creating an instrumental tool for improving tissue engineering and studies.
Collapse
Affiliation(s)
- Hongyan Gao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Zhien Wang
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Xiaoyu Wang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Siqi Wang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Quan Zhang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Xiaomeng Liu
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Jing Kong
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jun Yao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
4
|
Zhang Z, Qiu S, Wang Z, Hu Y. Vitamin D levels and five cardiovascular diseases: A Mendelian randomization study. Heliyon 2024; 10:e23674. [PMID: 38187309 PMCID: PMC10767153 DOI: 10.1016/j.heliyon.2023.e23674] [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: 04/01/2023] [Revised: 11/24/2023] [Accepted: 12/09/2023] [Indexed: 01/09/2024] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide, whilst vitamin D levels have been found to be associated with cardiovascular disease. To investigate the causal relationship between vitamin D levels and five cardiovascular diseases, a genome-wide association study (GWAS) was carried out using data on vitamin D levels (sample size = 79366), angina pectoris (18168 cases and 187840 controls), coronary heart disease (21012 cases and 197780 controls), lacunar stroke (6030 cases and 248929 controls), heart attack (10693 cases and 451187 controls), and hypertension (55917 cases and 162837 controls), with a Mendelian randomization (MR) analysis being subsequently performed. Six single nucleotide polymorphisms were used as instrumental variables (IVs). In addition, sensitivity analysis was performed to verify the reliability of the MR results here. The results showed a causal relationship between vitamin D levels and angina pectoris (OR = 0.51, 95 % CI: 0.28-0.93, P = 0.03), coronary heart disease (OR = 0.53, 95 % CI: 0.34-0.81, P = 0.004), and lacunar stroke (OR = 0.41, 95 % CI: 0.20-0.86, P = 0.02), but no causal relationship with heart attacks (OR = 1.00, 95 % CI: 0.99-1.01, P = 0.76) or hypertension (OR = 0.99, 95 % CI: 0.73-1.34, P = 0.94). Additionally, our IVs data showed no heterogeneity or pleiotropy, whilst the results of the MR analysis were reliable. This study contributes to the prevention and treatment of these five cardiovascular diseases.
Collapse
Affiliation(s)
- Zhishuai Zhang
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjang Production & Construction Group, Tarim University, Alaer, China
| | - Shizheng Qiu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhaoqing Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yang Hu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
5
|
Yang H, Yang Y, Kiskin FN, Shen M, Zhang JZ. Recent advances in regulating the proliferation or maturation of human-induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2023; 14:228. [PMID: 37649113 PMCID: PMC10469435 DOI: 10.1186/s13287-023-03470-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
In the last decade, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM)-based cell therapy has drawn broad attention as a potential therapy for treating injured hearts. However, mass production of hiPSC-CMs remains challenging, limiting their translational potential in regenerative medicine. Therefore, multiple strategies including cell cycle regulators, small molecules, co-culture systems, and epigenetic modifiers have been used to improve the proliferation of hiPSC-CMs. On the other hand, the immaturity of these proliferative hiPSC-CMs could lead to lethal arrhythmias due to their limited ability to functionally couple with resident cardiomyocytes. To achieve functional maturity, numerous methods such as prolonged culture, biochemical or biophysical stimulation, in vivo transplantation, and 3D culture approaches have been employed. In this review, we summarize recent approaches used to promote hiPSC-CM proliferation, and thoroughly review recent advances in promoting hiPSC-CM maturation, which will serve as the foundation for large-scale production of mature hiPSC-CMs for future clinical applications.
Collapse
Affiliation(s)
- Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Mengcheng Shen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
6
|
Genome Editing and Pathological Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:87-101. [DOI: 10.1007/978-981-19-5642-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
7
|
Zhu Y, Jackson D, Hunter B, Beattie L, Turner L, Hambly BD, Jeremy RW, Malecki C, Robertson EN, Li A, Remedios C, Richmond D, Semsarian C, O'Sullivan JF, Bannon PG, Lal S. Models of cardiovascular surgery biobanking to facilitate translational research and precision medicine. ESC Heart Fail 2021; 9:21-30. [PMID: 34931483 PMCID: PMC8787984 DOI: 10.1002/ehf2.13768] [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: 05/01/2021] [Revised: 11/07/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
Biobanking in health care has evolved over the last few decades from simple biological sample repositories to complex and dynamic units with multi‐organizational infrastructure networks and has become an essential tool for modern medical research. Cardiovascular tissue biobanking provides a unique opportunity to utilize cardiac and vascular samples for translational research into heart failure and other related pathologies. Current techniques for diagnosis, classification, and treatment monitoring of cardiac disease relies primarily on interpretation of clinical signs, imaging, and blood biomarkers. Further research at the disease source (i.e. myocardium and blood vessels) has been limited by a relative lack of access to quality human cardiac tissue and the inherent shortcomings of most animal models of heart disease. In this review, we describe a model for cardiovascular tissue biobanking and databasing, and its potential to facilitate basic and translational research. We share techniques to procure endocardial samples from patients with hypertrophic cardiomyopathy, heart failure with reduced ejection fraction, and heart failure with preserved ejection fraction, in addition to aortic disease samples. We discuss some of the issues with respect to data collection, privacy, biobank consent, and the governance of tissue biobanking. The development of tissue biobanks as described here has significant scope to improve and facilitate translational research in multi‐omic fields such as genomics, transcriptomics, proteomics, and metabolomics. This research heralds an era of precision medicine, in which patients with cardiovascular pathology can be provided with optimized and personalized medical care for the treatment of their individual phenotype.
Collapse
Affiliation(s)
- YingYan Zhu
- Department of Cardiothoracic Surgery Royal Prince Alfred Hospital Sydney New South Wales Australia
| | - Dan Jackson
- Department of Cardiothoracic Surgery Royal Prince Alfred Hospital Sydney New South Wales Australia
| | - Benjamin Hunter
- Cardiovascular Precision Laboratory The University of Sydney Sydney New South Wales 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Lorna Beattie
- Department of Cardiothoracic Surgery Royal Prince Alfred Hospital Sydney New South Wales Australia
- The Baird Institute for Applied Heart and Lung Surgical Research Sydney New South Wales Australia
| | - Lisa Turner
- Department of Cardiothoracic Surgery Royal Prince Alfred Hospital Sydney New South Wales Australia
- The Baird Institute for Applied Heart and Lung Surgical Research Sydney New South Wales Australia
| | - Brett D. Hambly
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Richmond W. Jeremy
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- The Baird Institute for Applied Heart and Lung Surgical Research Sydney New South Wales Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney New South Wales Australia
| | - Cassandra Malecki
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Elizabeth N. Robertson
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney New South Wales Australia
| | - Amy Li
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Department of Pharmacy & Biomedical Sciences La Trobe University Melbourne Victoria Australia
| | - Cris Remedios
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - David Richmond
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney New South Wales Australia
| | - Christopher Semsarian
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney New South Wales Australia
- Agnes Ginges Centre for Molecular Cardiology Centenary Institute Sydney New South Wales Australia
| | - John F. O'Sullivan
- Cardiovascular Precision Laboratory The University of Sydney Sydney New South Wales 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney New South Wales Australia
- Heart Research Institute The University of Sydney Sydney New South Wales Australia
| | - Paul G. Bannon
- Department of Cardiothoracic Surgery Royal Prince Alfred Hospital Sydney New South Wales Australia
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- The Baird Institute for Applied Heart and Lung Surgical Research Sydney New South Wales Australia
- RPA Institute of Academic Surgery (IAS) Royal Prince Alfred Hospital and the University of Sydney Sydney New South Wales Australia
| | - Sean Lal
- Cardiovascular Precision Laboratory The University of Sydney Sydney New South Wales 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
- Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- The Baird Institute for Applied Heart and Lung Surgical Research Sydney New South Wales Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney New South Wales Australia
| |
Collapse
|
8
|
Tafaleng EN, Malizio MR, Fox IJ, Soto-Gutierrez A. Synthetic human livers for modeling metabolic diseases. Curr Opin Gastroenterol 2021; 37:224-230. [PMID: 33769378 PMCID: PMC8223234 DOI: 10.1097/mog.0000000000000726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW In this review, we will explore recent advances in human induced pluripotent stem cell (iPSC)-based modeling of metabolic liver disease and biofabrication of synthetic human liver tissue while also discussing the emerging concept of synthetic biology to generate more physiologically relevant liver disease models. RECENT FINDING iPSC-based platforms have facilitated the study of underlying cellular mechanisms and potential therapeutic strategies for a number of metabolic liver diseases. Concurrently, rapid progress in biofabrication and gene editing technologies have led to the generation of human hepatic tissue that more closely mimic the complexity of the liver. SUMMARY iPSC-based liver tissue is rapidly becoming available for modeling liver physiology due to its ability to recapitulate the complex three-dimensional architecture of the liver and recapitulate interactions between the different cell types and their surroundings. These mini livers have also been used to recapitulate liver disease pathways using the tools of synthetic biology, such as gene editing, to control gene circuits. Further development in this field will undoubtedly bolster future investigations not only in disease modeling and basic research, but also in personalized medicine and autologous transplantation.
Collapse
Affiliation(s)
- Edgar N. Tafaleng
- Department of Surgery, University of Pittsburgh School of Medicine, Pennsylvania, USA
| | - Michelle R. Malizio
- Department of Pathology, University of Pittsburgh School of Medicine, Pennsylvania, USA
| | - Ira J. Fox
- Department of Surgery, University of Pittsburgh School of Medicine, Pennsylvania, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh School of Medicine, Pennsylvania, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
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]
|
10
|
Meki MH, Miller JM, Mohamed TMA. Heart Slices to Model Cardiac Physiology. Front Pharmacol 2021; 12:617922. [PMID: 33613292 PMCID: PMC7890402 DOI: 10.3389/fphar.2021.617922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/05/2021] [Indexed: 12/02/2022] Open
Abstract
Translational research in the cardiovascular field is hampered by the unavailability of cardiac models that can recapitulate organ-level physiology of the myocardium. Outside the body, cardiac tissue undergoes rapid dedifferentiation and maladaptation in culture. There is an ever-growing demand for preclinical platforms that allow for accurate, standardized, long-term, and rapid drug testing. Heart slices is an emerging technology that solves many of the problems with conventional myocardial culture systems. Heart slices are thin (<400 µm) slices of heart tissue from the adult ventricle. Several recent studies using heart slices have shown their ability to maintain the adult phenotype for prolonged periods in a multi cell-type environment. Here, we review the current status of cardiac culture systems and highlight the unique advantages offered by heart slices in the light of recent efforts in developing physiologically relevant heart slice culture systems.
Collapse
Affiliation(s)
- Moustafa H. Meki
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
| | - Jessica M. Miller
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
| | - Tamer M. A. Mohamed
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, United States
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
11
|
van Gorp PRR, Trines SA, Pijnappels DA, de Vries AAF. Multicellular In vitro Models of Cardiac Arrhythmias: Focus on Atrial Fibrillation. Front Cardiovasc Med 2020; 7:43. [PMID: 32296716 PMCID: PMC7138102 DOI: 10.3389/fcvm.2020.00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia in clinical practice with a large socioeconomic impact due to its associated morbidity, mortality, reduction in quality of life and health care costs. Currently, antiarrhythmic drug therapy is the first line of treatment for most symptomatic AF patients, despite its limited efficacy, the risk of inducing potentially life-threating ventricular tachyarrhythmias as well as other side effects. Alternative, in-hospital treatment modalities consisting of electrical cardioversion and invasive catheter ablation improve patients' symptoms, but often have to be repeated and are still associated with serious complications and only suitable for specific subgroups of AF patients. The development and progression of AF generally results from the interplay of multiple disease pathways and is accompanied by structural and functional (e.g., electrical) tissue remodeling. Rational development of novel treatment modalities for AF, with its many different etiologies, requires a comprehensive insight into the complex pathophysiological mechanisms. Monolayers of atrial cells represent a simplified surrogate of atrial tissue well-suited to investigate atrial arrhythmia mechanisms, since they can easily be used in a standardized, systematic and controllable manner to study the role of specific pathways and processes in the genesis, perpetuation and termination of atrial arrhythmias. In this review, we provide an overview of the currently available two- and three-dimensional multicellular in vitro systems for investigating the initiation, maintenance and termination of atrial arrhythmias and AF. This encompasses cultures of primary (animal-derived) atrial cardiomyocytes (CMs), pluripotent stem cell-derived atrial-like CMs and (conditionally) immortalized atrial CMs. The strengths and weaknesses of each of these model systems for studying atrial arrhythmias will be discussed as well as their implications for future studies.
Collapse
Affiliation(s)
| | | | | | - Antoine A. F. de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
12
|
Ahmed RE, Anzai T, Chanthra N, Uosaki H. A Brief Review of Current Maturation Methods for Human Induced Pluripotent Stem Cells-Derived Cardiomyocytes. Front Cell Dev Biol 2020; 8:178. [PMID: 32266260 PMCID: PMC7096382 DOI: 10.3389/fcell.2020.00178] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/03/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Therefore, the discovery of induced pluripotent stem cells (iPSCs) and the subsequent generation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) was a pivotal point in regenerative medicine and cardiovascular research. They constituted an appealing tool for replacing dead and dysfunctional cardiac tissue, screening cardiac drugs and toxins, and studying inherited cardiac diseases. The problem is that these cells remain largely immature, and in order to utilize them, they must reach a functional degree of maturity. To attempt to mimic in vivo environment, various methods including prolonging culture time, co-culture and modulations of chemical, electrical, mechanical culture conditions have been tried. In addition to that, changing the topology of the culture made huge progress with the introduction of the 3D culture that closely resembles the in vivo cardiac topology and overcomes many of the limitations of the conventionally used 2D models. Nonetheless, 3D culture alone is not enough, and using a combination of these methods is being explored. In this review, we summarize the main differences between immature, fetal-like hiPSC-CMs and adult cardiomyocytes, then glance at the current approaches used to promote hiPSC-CMs maturation. In the second part, we focus on the evolving 3D culture model - it's structure, the effect on hiPSC-CMs maturation, incorporation with different maturation methods, limitations and future prospects.
Collapse
Affiliation(s)
- Razan Elfadil Ahmed
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
13
|
Zhao Y, Rafatian N, Wang EY, Wu Q, Lai BFL, Lu RX, Savoji H, Radisic M. Towards chamber specific heart-on-a-chip for drug testing applications. Adv Drug Deliv Rev 2020; 165-166:60-76. [PMID: 31917972 PMCID: PMC7338250 DOI: 10.1016/j.addr.2019.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
Modeling of human organs has long been a task for scientists in order to lower the costs of therapeutic development and understand the pathological onset of human disease. For decades, despite marked differences in genetics and etiology, animal models remained the norm for drug discovery and disease modeling. Innovative biofabrication techniques have facilitated the development of organ-on-a-chip technology that has great potential to complement conventional animal models. However, human organ as a whole, more specifically the human heart, is difficult to regenerate in vitro, in terms of its chamber specific orientation and its electrical functional complexity. Recent progress with the development of induced pluripotent stem cell differentiation protocols, made recapitulating the complexity of the human heart possible through the generation of cells representative of atrial & ventricular tissue, the sinoatrial node, atrioventricular node and Purkinje fibers. Current heart-on-a-chip approaches incorporate biological, electrical, mechanical, and topographical cues to facilitate tissue maturation, therefore improving the predictive power for the chamber-specific therapeutic effects targeting adult human. In this review, we will give a summary of current advances in heart-on-a-chip technology and provide a comprehensive outlook on the challenges involved in the development of human physiologically relevant heart-on-a-chip.
Collapse
Affiliation(s)
- Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Naimeh Rafatian
- Division of Cardiology and Peter Munk Cardiac Center, University of Health Network, Toronto, Ontario M5G 2N2, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Benjamin F L Lai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xingze Lu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Toronto General Research Institute, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
14
|
Mesquita FCP, Arantes PC, Kasai-Brunswick TH, Araujo DS, Gubert F, Monnerat G, Silva Dos Santos D, Neiman G, Leitão IC, Barbosa RAQ, Coutinho JL, Vaz IM, Dos Santos MN, Borgonovo T, Cruz FES, Miriuka S, Medei EH, Campos de Carvalho AC, Carvalho AB. R534C mutation in hERG causes a trafficking defect in iPSC-derived cardiomyocytes from patients with type 2 long QT syndrome. Sci Rep 2019; 9:19203. [PMID: 31844156 PMCID: PMC6915575 DOI: 10.1038/s41598-019-55837-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
Patient-specific cardiomyocytes obtained from induced pluripotent stem cells (CM-iPSC) offer unprecedented mechanistic insights in the study of inherited cardiac diseases. The objective of this work was to study a type 2 long QT syndrome (LQTS2)-associated mutation (c.1600C > T in KCNH2, p.R534C in hERG) in CM-iPSC. Peripheral blood mononuclear cells were isolated from two patients with the R534C mutation and iPSCs were generated. In addition, the same mutation was inserted in a control iPSC line by genome editing using CRISPR/Cas9. Cells expressed pluripotency markers and showed spontaneous differentiation into the three embryonic germ layers. Electrophysiology demonstrated that action potential duration (APD) of LQTS2 CM-iPSC was significantly longer than that of the control line, as well as the triangulation of the action potentials (AP), implying a longer duration of phase 3. Treatment with the IKr inhibitor E4031 only caused APD prolongation in the control line. Patch clamp showed a reduction of IKr on LQTS2 CM-iPSC compared to control, but channel activation was not significantly affected. Immunofluorescence for hERG demonstrated perinuclear staining in LQTS2 CM-iPSC. In conclusion, CM-iPSC recapitulated the LQTS2 phenotype and our findings suggest that the R534C mutation in KCNH2 leads to a channel trafficking defect to the plasma membrane.
Collapse
Affiliation(s)
- Fernanda C P Mesquita
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Paulo C Arantes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Tais H Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Dayana S Araujo
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Fernanda Gubert
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco F, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Gustavo Monnerat
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Danúbia Silva Dos Santos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Gabriel Neiman
- FLENI Foundation, Sede Escobar. Ruta 9, Km 53, Belen de Escobar, BA, B1625, Argentina
| | - Isabela C Leitão
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Raiana A Q Barbosa
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Jorge L Coutinho
- National Institute of Cardiology, Rua das Laranjeiras 374, Rio de Janeiro, RJ, 22240-006, Brazil
| | - Isadora M Vaz
- Pontifical Catholic University of Parana. Rua Imaculada Conceição 1155, Curitiba, PR, 80215-901, Brazil
| | - Marcus N Dos Santos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Tamara Borgonovo
- Pontifical Catholic University of Parana. Rua Imaculada Conceição 1155, Curitiba, PR, 80215-901, Brazil
| | - Fernando E S Cruz
- National Institute of Cardiology, Rua das Laranjeiras 374, Rio de Janeiro, RJ, 22240-006, Brazil
| | - Santiago Miriuka
- FLENI Foundation, Sede Escobar. Ruta 9, Km 53, Belen de Escobar, BA, B1625, Argentina
| | - Emiliano H Medei
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Antonio C Campos de Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Institute of Cardiology, Rua das Laranjeiras 374, Rio de Janeiro, RJ, 22240-006, Brazil.
- National Institute for Science and Technology in Regenerative Medicine. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Adriana B Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco G, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
- National Institute for Science and Technology in Regenerative Medicine. Avenida Carlos Chagas Filho 373, Bloco M, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
15
|
Ghazizadeh Z, Kiviniemi T, Olafsson S, Plotnick D, Beerens ME, Zhang K, Gillon L, Steinbaugh MJ, Barrera V, Sui SH, Werdich AA, Kapur S, Eranti A, Gunn J, Jalkanen J, Airaksinen J, Kleber AG, Hollmén M, MacRae CA. Metastable Atrial State Underlies the Primary Genetic Substrate for MYL4 Mutation-Associated Atrial Fibrillation. Circulation 2019; 141:301-312. [PMID: 31735076 DOI: 10.1161/circulationaha.119.044268] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common clinical arrhythmia and is associated with heart failure, stroke, and increased mortality. The myocardial substrate for AF is poorly understood because of limited access to primary human tissue and mechanistic questions around existing in vitro or in vivo models. METHODS Using an MYH6:mCherry knock-in reporter line, we developed a protocol to generate and highly purify human pluripotent stem cell-derived cardiomyocytes displaying physiological and molecular characteristics of atrial cells. We modeled human MYL4 mutants, one of the few definitive genetic causes of AF. To explore non-cell-autonomous components of AF substrate, we also created a zebrafish Myl4 knockout model, which exhibited molecular, cellular, and physiologic abnormalities that parallel those in humans bearing the cognate mutations. RESULTS There was evidence of increased retinoic acid signaling in both human embryonic stem cells and zebrafish mutant models, as well as abnormal expression and localization of cytoskeletal proteins, and loss of intracellular nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide + hydrogen. To identify potentially druggable proximate mechanisms, we performed a chemical suppressor screen integrating multiple human cellular and zebrafish in vivo endpoints. This screen identified Cx43 (connexin 43) hemichannel blockade as a robust suppressor of the abnormal phenotypes in both models of MYL4 (myosin light chain 4)-related atrial cardiomyopathy. Immunofluorescence and coimmunoprecipitation studies revealed an interaction between MYL4 and Cx43 with altered localization of Cx43 hemichannels to the lateral membrane in MYL4 mutants, as well as in atrial biopsies from unselected forms of human AF. The membrane fraction from MYL4-/- human embryonic stem cell derived atrial cells demonstrated increased phospho-Cx43, which was further accentuated by retinoic acid treatment and by the presence of risk alleles at the Pitx2 locus. PKC (protein kinase C) was induced by retinoic acid, and PKC inhibition also rescued the abnormal phenotypes in the atrial cardiomyopathy models. CONCLUSIONS These data establish a mechanistic link between the transcriptional, metabolic and electrical pathways previously implicated in AF substrate and suggest novel avenues for the prevention or therapy of this common arrhythmia.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tuomas Kiviniemi
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Sigurast Olafsson
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David Plotnick
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Manu E Beerens
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kun Zhang
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Leah Gillon
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Victor Barrera
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Shannan Ho Sui
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Andreas A Werdich
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sunil Kapur
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Antti Eranti
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Jarmo Gunn
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Juho Jalkanen
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Juhani Airaksinen
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Andre G Kleber
- Department of Pathology, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, MA (A.G.K.)
| | - Maija Hollmén
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Calum A MacRae
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Genetics and Network Medicine Divisions (C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Boston, MA (C.A.M.)
| |
Collapse
|
16
|
Fu H, Miao C, Rui Y, Hu F, Shen M, Xu H, Zhang C, Dong Y, Wang W, Gu H, Duan Y. Strategy to prevent cardiac toxicity induced by polyacrylic acid decorated iron MRI contrast agent and investigation of its mechanism. Biomaterials 2019; 222:119442. [PMID: 31491561 DOI: 10.1016/j.biomaterials.2019.119442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/30/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022]
Abstract
Polyelectrolyte modified iron oxide nanoparticles have great potential applications for clinical magnetic resonance imaging (MRI) and anemia treatments, however, possible associated heart toxicity is rarely reported. Here, polyacrylic acid (PAA)-coated Fe3O4 nanoparticles (PION) were synthesized and lethal reactions appeared when it was applied in vivo. The investigation of underlying mechanism showed that PION could break electrolyte balance and further resulted in serious heart failure, which was observed under color doppler ultrasound and dynamic vector blood flow technique. The results demonstrated that PION had a strong absorption tendency for divalent ions and the maximum tolerated dose (MTD) was lower than 100 mg/kg. From electrocardiography (ECG), PION presented an obvious impact on CaV1.2 ion channel, which leading to fatal arrhythmia. An appropriate solution for preventing this deadly effect was pre-chelation Ca2+ (n (Ca): n (COOH) = 3: 8) to PION (PION-Ca), which displayed much higher cardiac and electrophysiological safety when sealing the binding point of divalent cation ions with PAA. The injection in Beagle dogs further confirmed the safety of PION-Ca. This study explored the mechanism and offered a solution for cardiac toxicity induced by PAA-coated nanoparticles, which guides for enhancing the safety of such polyelectrolyte decorated nanoparticles and provides assurance for clinical applications.
Collapse
Affiliation(s)
- Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chongchong Miao
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanpeng Rui
- Department of Radiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenglin Hu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Shen
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chunfu Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongchen Gu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
17
|
Jimenez-Tellez N, Greenway SC. Cellular models for human cardiomyopathy: What is the best option? World J Cardiol 2019; 11:221-235. [PMID: 31754410 PMCID: PMC6859298 DOI: 10.4330/wjc.v11.i10.221] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/17/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
Abstract
The genetic cardiomyopathies are a group of disorders related by abnormal myocardial structure and function. Although individually rare, these diseases collectively represent a significant health burden since they usually develop early in life and are a major cause of morbidity and mortality amongst affected children. The heterogeneity and rarity of these disorders requires the use of an appropriate model system in order to characterize the mechanism of disease and develop useful therapeutics since standard drug trials are infeasible. A common approach to study human disease involves the use of animal models, especially rodents, but due to important biological and physiological differences, this model system may not recapitulate human disease. An alternative approach for studying the metabolic cardiomyopathies relies on the use of cellular models which have most frequently been immortalized cell lines or patient-derived fibroblasts. However, the recent introduction of induced pluripotent stem cells (iPSCs), which have the ability to differentiate into any cell type in the body, is of great interest and has the potential to revolutionize the study of rare diseases. In this paper we review the advantages and disadvantages of each model system by comparing their utility for the study of mitochondrial cardiomyopathy with a particular focus on the use of iPSCs in cardiovascular biology for the modeling of rare genetic or metabolic diseases.
Collapse
Affiliation(s)
- Nerea Jimenez-Tellez
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Steven C Greenway
- Departments of Pediatrics, Cardiac Sciences, Biochemistry & Molecular Biology, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
18
|
Burnett SD, Blanchette AD, Grimm FA, House JS, Reif DM, Wright FA, Chiu WA, Rusyn I. Population-based toxicity screening in human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2019; 381:114711. [PMID: 31425687 PMCID: PMC6745256 DOI: 10.1016/j.taap.2019.114711] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/06/2019] [Accepted: 08/14/2019] [Indexed: 11/23/2022]
Abstract
The potential for cardiotoxicity is carefully evaluated for pharmaceuticals, as it is a major safety liability. However, environmental chemicals are seldom tested for their cardiotoxic potential. Moreover, there is a large variability in both baseline and drug-induced cardiovascular risk in humans, but data are lacking on the degree to which susceptibility to chemically-induced cardiotoxicity may also vary. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes have become an important in vitro model for drug screening. Thus, we hypothesized that a population-based model of iPSC-derived cardiomyocytes from a diverse set of individuals can be used to assess potential hazard and inter-individual variability in chemical effects on these cells. We conducted concentration-response screening of 134 chemicals (pharmaceuticals, industrial and environmental chemicals and food constituents) in iPSC-derived cardiomyocytes from 43 individuals, comprising both sexes and diverse ancestry. We measured kinetic calcium flux and conducted high-content imaging following chemical exposure, and utilized a panel of functional and cytotoxicity parameters in concentration-response for each chemical and donor. We show reproducible inter-individual variability in both baseline and chemical-induced effects on iPSC-derived cardiomyocytes. Further, chemical-specific variability in potency and degree of population variability were quantified. This study shows the feasibility of using an organotypic population-based human in vitro model to quantitatively assess chemicals for which little cardiotoxicity information is available. Ultimately, these results advance in vitro toxicity testing methodologies by providing an innovative tool for population-based cardiotoxicity screening, contributing to the paradigm shift from traditional animal models of toxicity to in vitro toxicity testing methods.
Collapse
Affiliation(s)
- Sarah D Burnett
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA.
| | - Alexander D Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA.
| | - Fabian A Grimm
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA.
| | - John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA.
| | - David M Reif
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Department of Statistics, North Carolina State University, Raleigh, NC 27695, USA
| | - Fred A Wright
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Department of Statistics, North Carolina State University, Raleigh, NC 27695, USA.
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA.
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA.
| |
Collapse
|
19
|
Abstract
Systems medicine is a holistic approach to deciphering the complexity of human physiology in health and disease. In essence, a living body is constituted of networks of dynamically interacting units (molecules, cells, organs, etc) that underlie its collective functions. Declining resilience because of aging and other chronic environmental exposures drives the system to transition from a health state to a disease state; these transitions, triggered by acute perturbations or chronic disturbance, manifest as qualitative shifts in the interactions and dynamics of the disease-perturbed networks. Understanding health-to-disease transitions poses a high-dimensional nonlinear reconstruction problem that requires deep understanding of biology and innovation in study design, technology, and data analysis. With a focus on the principles of systems medicine, this Review discusses approaches for deciphering this biological complexity from a novel perspective, namely, understanding how disease-perturbed networks function; their study provides insights into fundamental disease mechanisms. The immediate goals for systems medicine are to identify early transitions to cardiovascular (and other chronic) diseases and to accelerate the translation of new preventive, diagnostic, or therapeutic targets into clinical practice, a critical step in the development of personalized, predictive, preventive, and participatory (P4) medicine.
Collapse
Affiliation(s)
- Kalliopi Trachana
- From the Institute for Systems Biology, Seattle, WA (K.T., R.B., G.G., N.D.P., S.H., L.E.H.)
| | - Rhishikesh Bargaje
- From the Institute for Systems Biology, Seattle, WA (K.T., R.B., G.G., N.D.P., S.H., L.E.H.)
| | - Gustavo Glusman
- From the Institute for Systems Biology, Seattle, WA (K.T., R.B., G.G., N.D.P., S.H., L.E.H.)
| | - Nathan D Price
- From the Institute for Systems Biology, Seattle, WA (K.T., R.B., G.G., N.D.P., S.H., L.E.H.)
| | - Sui Huang
- From the Institute for Systems Biology, Seattle, WA (K.T., R.B., G.G., N.D.P., S.H., L.E.H.).,Department of Biological Sciences, University of Calgary, Alberta, Canada (S.H.)
| | - Leroy E Hood
- From the Institute for Systems Biology, Seattle, WA (K.T., R.B., G.G., N.D.P., S.H., L.E.H.)
| |
Collapse
|
20
|
Olivaes J, Bonamino MH, Markoski MM. CRISPR/Cas 9 system for the treatment of dilated cardiomyopathy: A hypothesis related to function of a MAP kinase. Med Hypotheses 2019; 128:91-93. [DOI: 10.1016/j.mehy.2019.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/23/2019] [Accepted: 05/12/2019] [Indexed: 10/26/2022]
|
21
|
Blanchette AD, Grimm FA, Dalaijamts C, Hsieh NH, Ferguson K, Luo YS, Rusyn I, Chiu WA. Thorough QT/QTc in a Dish: An In Vitro Human Model That Accurately Predicts Clinical Concentration-QTc Relationships. Clin Pharmacol Ther 2019; 105:1175-1186. [PMID: 30346629 PMCID: PMC6465173 DOI: 10.1002/cpt.1259] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022]
Abstract
"Thorough QT/corrected QT (QTc)" (TQT) studies are cornerstones of clinical cardiovascular safety assessment. However, TQT studies are resource intensive, and preclinical models predictive of the threshold of regulatory concern are lacking. We hypothesized that an in vitro model using induced pluripotent stem cell (iPSC)-derived cardiomyocytes from a diverse sample of human subjects can serve as a "TQT study in a dish." For 10 positive and 3 negative control drugs, in vitro concentration-QTc, computed using a population Bayesian model, accurately predicted known in vivo concentration-QTc. Moreover, predictions of the percent confidence that the regulatory threshold of 10 ms QTc prolongation would be breached were also consistent with in vivo evidence. This "TQT study in a dish," consisting of a population-based iPSC-derived cardiomyocyte model and Bayesian concentration-QTc modeling, has several advantages over existing in vitro platforms, including higher throughput, lower cost, and the ability to accurately predict the in vivo concentration range below the threshold of regulatory concern.
Collapse
Affiliation(s)
| | - Fabian A. Grimm
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| | - Chimedullam Dalaijamts
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| | - Nan-Hung Hsieh
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| | - Kyle Ferguson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| | - Yu-Syuan Luo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| | - Weihsueh A. Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX
| |
Collapse
|
22
|
Shafaattalab S, Lin E, Christidi E, Huang H, Nartiss Y, Garcia A, Lee J, Protze S, Keller G, Brunham L, Tibbits GF, Laksman Z. Ibrutinib Displays Atrial-Specific Toxicity in Human Stem Cell-Derived Cardiomyocytes. Stem Cell Reports 2019; 12:996-1006. [PMID: 31031187 PMCID: PMC6524928 DOI: 10.1016/j.stemcr.2019.03.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 01/08/2023] Open
Abstract
Ibrutinib (IB) is an oral Bruton's tyrosine kinase (BTK) inhibitor that has demonstrated benefit in B cell cancers, but is associated with a dramatic increase in atrial fibrillation (AF). We employed cell-specific differentiation protocols and optical mapping to investigate the effects of IB and other tyrosine kinase inhibitors (TKIs) on the voltage and calcium transients of atrial and ventricular human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). IB demonstrated direct cell-specific effects on atrial hPSC-CMs that would be predicted to predispose to AF. Second-generation BTK inhibitors did not have the same effect. Furthermore, IB exposure was associated with differential chamber-specific regulation of a number of regulatory pathways including the receptor tyrosine kinase pathway, which may be implicated in the pathogenesis of AF. Our study is the first to demonstrate cell-type-specific toxicity in hPSC-derived atrial and ventricular cardiomyocytes, which reliably reproduces the clinical cardiotoxicity observed.
hPSCs can be differentiated into atrial and ventricular cardiomyocytes (CMs) Drug effects can be measured using optical mapping of voltage and calcium transients Ibrutinib demonstrates cell-specific toxicity on atrial hPSC-CMs Ibrutinib exposure is associated with chamber-specific effects on regulatory pathways
Collapse
Affiliation(s)
- Sanam Shafaattalab
- Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1A6, Canada; British Columbia Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Eric Lin
- Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1A6, Canada
| | - Effimia Christidi
- University of British Columbia, 170-6371 Crescent Road, Vancouver, BC V6T 1Z2, Canada
| | - Haojun Huang
- University of British Columbia, 170-6371 Crescent Road, Vancouver, BC V6T 1Z2, Canada
| | - Yulia Nartiss
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Analucia Garcia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Jeehon Lee
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stephanie Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gordon Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Liam Brunham
- University of British Columbia, 170-6371 Crescent Road, Vancouver, BC V6T 1Z2, Canada
| | - Glen F Tibbits
- Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1A6, Canada; British Columbia Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Zachary Laksman
- Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1A6, Canada; University of British Columbia, 170-6371 Crescent Road, Vancouver, BC V6T 1Z2, Canada.
| |
Collapse
|
23
|
Chen Z, Xian W, Bellin M, Dorn T, Tian Q, Goedel A, Dreizehnter L, Schneider CM, Ward-van Oostwaard D, Ng JKM, Hinkel R, Pane LS, Mummery CL, Lipp P, Moretti A, Laugwitz KL, Sinnecker D. Subtype-specific promoter-driven action potential imaging for precise disease modelling and drug testing in hiPSC-derived cardiomyocytes. Eur Heart J 2019; 38:292-301. [PMID: 28182242 PMCID: PMC5381588 DOI: 10.1093/eurheartj/ehw189] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 03/18/2016] [Accepted: 04/19/2016] [Indexed: 12/30/2022] Open
Affiliation(s)
- Zhifen Chen
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Wenying Xian
- Institute for Molecular Cell Biology, Medical Faculty, University Homburg/Saar, Universität des Saarlandes, Homburg/Saar 66421, Germany
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Tatjana Dorn
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Qinghai Tian
- Institute for Molecular Cell Biology, Medical Faculty, University Homburg/Saar, Universität des Saarlandes, Homburg/Saar 66421, Germany
| | - Alexander Goedel
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Lisa Dreizehnter
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Christine M Schneider
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Dorien Ward-van Oostwaard
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Judy King Man Ng
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | | | - Luna Simona Pane
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Peter Lipp
- Institute for Molecular Cell Biology, Medical Faculty, University Homburg/Saar, Universität des Saarlandes, Homburg/Saar 66421, Germany
| | | | | | - Daniel Sinnecker
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technische Universität München, Munich 81675, Germany
| |
Collapse
|
24
|
Tsifaki M, Kelaini S, Caines R, Yang C, Margariti A. Regenerating the Cardiovascular System Through Cell Reprogramming; Current Approaches and a Look Into the Future. Front Cardiovasc Med 2018; 5:109. [PMID: 30177971 PMCID: PMC6109758 DOI: 10.3389/fcvm.2018.00109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD), despite the advances of the medical field, remains one of the leading causes of mortality worldwide. Discovering novel treatments based on cell therapy or drugs is critical, and induced pluripotent stem cells (iPS Cells) technology has made it possible to design extensive disease-specific in vitro models. Elucidating the differentiation process challenged our previous knowledge of cell plasticity and capabilities and allows the concept of cell reprogramming technology to be established, which has inspired the creation of both in vitro and in vivo techniques. Patient-specific cell lines provide the opportunity of studying their pathophysiology in vitro, which can lead to novel drug development. At the same time, in vivo models have been designed where in situ transdifferentiation of cell populations into cardiomyocytes or endothelial cells (ECs) give hope toward effective cell therapies. Unfortunately, the efficiency as well as the concerns about the safety of all these methods make it exceedingly difficult to pass to the clinical trial phase. It is our opinion that creating an ex vivo model out of patient-specific cells will be one of the most important goals in the future to help surpass all these hindrances. Thus, in this review we aim to present the current state of research in reprogramming toward the cardiovascular system's regeneration, and showcase how the development and study of a multicellular 3D ex vivo model will improve our fighting chances.
Collapse
Affiliation(s)
- Marianna Tsifaki
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sophia Kelaini
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
25
|
Kalra S, Montanaro F, Denning C. Can Human Pluripotent Stem Cell-Derived Cardiomyocytes Advance Understanding of Muscular Dystrophies? J Neuromuscul Dis 2018; 3:309-332. [PMID: 27854224 PMCID: PMC5123622 DOI: 10.3233/jnd-150133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Muscular dystrophies (MDs) are clinically and molecularly a highly heterogeneous group of single-gene disorders that primarily affect striated muscles. Cardiac disease is present in several MDs where it is an important contributor to morbidity and mortality. Careful monitoring of cardiac issues is necessary but current management of cardiac involvement does not effectively protect from disease progression and cardiac failure. There is a critical need to gain new knowledge on the diverse molecular underpinnings of cardiac disease in MDs in order to guide cardiac treatment development and assist in reaching a clearer consensus on cardiac disease management in the clinic. Animal models are available for the majority of MDs and have been invaluable tools in probing disease mechanisms and in pre-clinical screens. However, there are recognized genetic, physiological, and structural differences between human and animal hearts that impact disease progression, manifestation, and response to pharmacological interventions. Therefore, there is a need to develop parallel human systems to model cardiac disease in MDs. This review discusses the current status of cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSC) to model cardiac disease, with a focus on Duchenne muscular dystrophy (DMD) and myotonic dystrophy (DM1). We seek to provide a balanced view of opportunities and limitations offered by this system in elucidating disease mechanisms pertinent to human cardiac physiology and as a platform for treatment development or refinement.
Collapse
Affiliation(s)
- Spandan Kalra
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, UK
| | - Federica Montanaro
- Dubowitz Neuromuscular Centre, Department of Molecular Neurosciences, University College London - Institute of Child Health, London, UK
| | - Chris Denning
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, UK
| |
Collapse
|
26
|
Kumar S, Blangero J, Curran JE. Induced Pluripotent Stem Cells in Disease Modeling and Gene Identification. Methods Mol Biol 2018; 1706:17-38. [PMID: 29423791 DOI: 10.1007/978-1-4939-7471-9_2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Experimental modeling of human inherited disorders provides insight into the cellular and molecular mechanisms involved, and the underlying genetic component influencing, the disease phenotype. The breakthrough development of induced pluripotent stem cell (iPSC) technology represents a quantum leap in experimental modeling of human diseases, providing investigators with a self-renewing and, thus, unlimited source of pluripotent cells for targeted differentiation. In principle, the entire range of cell types found in the human body can be interrogated using an iPSC approach. Therefore, iPSC technology, and the increasingly refined abilities to differentiate iPSCs into disease-relevant target cells, has far-reaching implications for understanding disease pathophysiology, identifying disease-causing genes, and developing more precise therapeutics, including advances in regenerative medicine. In this chapter, we discuss the technological perspectives and recent developments in the application of patient-derived iPSC lines for human disease modeling and disease gene identification.
Collapse
Affiliation(s)
- Satish Kumar
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, School of Medicine, 1214 W Schunior St, Edinburg, TX, 78541, USA.
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, School of Medicine, 1214 W Schunior St, Edinburg, TX, 78541, USA
| | - Joanne E Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, School of Medicine, 1214 W Schunior St, Edinburg, TX, 78541, USA
| |
Collapse
|
27
|
The Impact of CRISPR/Cas9 Technology on Cardiac Research: From Disease Modelling to Therapeutic Approaches. Stem Cells Int 2017; 2017:8960236. [PMID: 29434642 PMCID: PMC5757142 DOI: 10.1155/2017/8960236] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022] Open
Abstract
Genome-editing technology has emerged as a powerful method that enables the generation of genetically modified cells and organisms necessary to elucidate gene function and mechanisms of human diseases. The clustered regularly interspaced short palindromic repeats- (CRISPR-) associated 9 (Cas9) system has rapidly become one of the most popular approaches for genome editing in basic biomedical research over recent years because of its simplicity and adaptability. CRISPR/Cas9 genome editing has been used to correct DNA mutations ranging from a single base pair to large deletions in both in vitro and in vivo model systems. CRISPR/Cas9 has been used to increase the understanding of many aspects of cardiovascular disorders, including lipid metabolism, electrophysiology and genetic inheritance. The CRISPR/Cas9 technology has been proven to be effective in creating gene knockout (KO) or knockin in human cells and is particularly useful for editing induced pluripotent stem cells (iPSCs). Despite these progresses, some biological, technical, and ethical issues are limiting the therapeutic potential of genome editing in cardiovascular diseases. This review will focus on various applications of CRISPR/Cas9 genome editing in the cardiovascular field, for both disease research and the prospect of in vivo genome-editing therapies in the future.
Collapse
|
28
|
Modeling Atrial Fibrillation using Human Embryonic Stem Cell-Derived Atrial Tissue. Sci Rep 2017; 7:5268. [PMID: 28706272 PMCID: PMC5509676 DOI: 10.1038/s41598-017-05652-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022] Open
Abstract
Since current experimental models of Atrial Fibrillation (AF) have significant limitations, we used human embryonic stem cells (hESCs) to generate an atrial-specific tissue model of AF for pharmacologic testing. We generated atrial-like cardiomyocytes (CMs) from hESCs which preferentially expressed atrial-specific genes, and had shorter action potential (AP) durations compared to ventricular-like CMs. We then generated confluent atrial-like CM sheets and interrogated them using optical mapping techniques. Atrial-like CM sheets (~1 cm in diameter) showed uniform AP propagation, and rapid re-entrant rotor patterns, as seen in AF could be induced. Anti-arrhythmic drugs were tested on single atrial-like CMs and cell sheets. Flecainide profoundly slowed upstroke velocity without affecting AP duration, leading to reduced conduction velocities (CVs), curvatures and cycle lengths of rotors, consistent with increased rotor organization and expansion. By contrast, consistent with block of rapid delayed rectifier K+ currents (Ikr) and AP prolongation in isolated atrial-like CMs, dofetilide prolonged APs and reduced cycle lengths of rotors in cell sheets without affecting CV. In conclusion, using our hESC-derived atrial CM preparations, we demonstrate that flecainide and dofetilide modulate reentrant arrhythmogenic rotor activation patterns in a manner that helps explain their efficacy in treating and preventing AF.
Collapse
|
29
|
Hříbková H, Zelinková J, Sun YM. Progress in human pluripotent stem cell-based modeling systems for neurological diseases. NEUROGENESIS 2017. [DOI: 10.1080/23262133.2017.1324258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hana Hříbková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Zelinková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Yuh-Man Sun
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
30
|
Integration concepts for multi-organ chips: how to maintain flexibility?! Future Sci OA 2017; 3:FSO180. [PMID: 28670472 PMCID: PMC5481865 DOI: 10.4155/fsoa-2016-0092] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/01/2017] [Indexed: 12/28/2022] Open
Abstract
Multi-organ platforms have an enormous potential to lead to a paradigm shift in a multitude of research domains including drug development, toxicological screening, personalized medicine as well as disease modeling. Integrating multiple organ–tissues into one microfluidic circulation merges the advantages of cell lines (human genetic background) and animal models (complex physiology) and enables the creation of more in vivo-like in vitro models. In recent years, a variety of design concepts for multi-organ platforms have been introduced, categorizable into static, semistatic and flexible systems. The most promising approach seems to be flexible interconnection of single-organ platforms to application-specific multi-organ systems. This perspective elucidates the concept of ‘mix-and-match’ toolboxes and discusses the numerous advantages compared with static/semistatic platforms as well as remaining challenges. ‘Organs-on-a-chip’ are platforms accommodating organ-specific human tissues in microscale 3D chambers with physiologically relevant structure. Broken down to the basic building blocks but simultaneously mimicking essential organ functions, these sophisticated biochips can help reduce the need for animal models in drug development, toxicity screening and basic research. However, to simulate a drug's journey through the human body, it is necessary to consider how a combination of organs responds to a given drug. In this perspective, concepts of realizing such ‘multi-organ platforms’ and the need for ‘mix-and-match’ toolboxes, which contain a range of single-organ units interconnected in individual, application-specific configurations, are discussed.
Collapse
|
31
|
Narmada BC, Goh YT, Li H, Sinha S, Yu H, Cheung C. Human Stem Cell-Derived Endothelial-Hepatic Platform for Efficacy Testing of Vascular-Protective Metabolites from Nutraceuticals. Stem Cells Transl Med 2017; 6:851-863. [PMID: 28297582 PMCID: PMC5442778 DOI: 10.5966/sctm.2016-0129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/09/2016] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis underlies many cardiovascular and cerebrovascular diseases. Nutraceuticals are emerging as a therapeutic moiety for restoring vascular health. Unlike small-molecule drugs, the complexity of ingredients in nutraceuticals often confounds evaluation of their efficacy in preclinical evaluation. It is recognized that the liver is a vital organ in processing complex compounds into bioactive metabolites. In this work, we developed a coculture system of human pluripotent stem cell-derived endothelial cells (hPSC-ECs) and human pluripotent stem cell-derived hepatocytes (hPSC-HEPs) for predicting vascular-protective effects of nutraceuticals. To validate our model, two compounds (quercetin and genistein), known to have anti-inflammatory effects on vasculatures, were selected. We found that both quercetin and genistein were ineffective at suppressing inflammatory activation by interleukin-1β owing to limited metabolic activity of hPSC-ECs. Conversely, hPSC-HEPs demonstrated metabolic capacity to break down both nutraceuticals into primary and secondary metabolites. When hPSC-HEPs were cocultured with hPSC-ECs to permit paracrine interactions, the continuous turnover of metabolites mitigated interleukin-1β stimulation on hPSC-ECs. We observed significant reductions in inflammatory gene expressions, nuclear translocation of nuclear factor κB, and interleukin-8 production. Thus, integration of hPSC-HEPs could accurately reproduce systemic effects involved in drug metabolism in vivo to unravel beneficial constituents in nutraceuticals. This physiologically relevant endothelial-hepatic platform would be a great resource in predicting the efficacy of complex nutraceuticals and mechanistic interrogation of vascular-targeting candidate compounds. Stem Cells Translational Medicine 2017;6:851-863.
Collapse
Affiliation(s)
| | - Yeek Teck Goh
- Institute of Molecular and Cell Biology, Proteos, Singapore
| | - Huan Li
- Institute of Bioengineering and Nanotechnology, Nanos, Singapore
| | - Sanjay Sinha
- The Anne McLaren Laboratory of Regenerative Medicine, Wellcome Trust‐Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Division of Cardiovascular Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, Nanos, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Mechanobiology Institute, Singapore
- Singapore‐MIT Alliance for Research and Technology, BioSyM, Singapore
| | - Christine Cheung
- Institute of Molecular and Cell Biology, Proteos, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
32
|
Zhang Q, Chen W, Tan S, Lin T. Stem Cells for Modeling and Therapy of Parkinson's Disease. Hum Gene Ther 2016; 28:85-98. [PMID: 27762639 DOI: 10.1089/hum.2016.116] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disease after Alzheimer's disease, which is characterized by a low level of dopamine being expressing in the striatum and a deterioration of dopaminergic neurons (DAn) in the substantia nigra pars compacta. Generation of PD-derived DAn, including differentiation of human embryonic stem cells, human neural stem cells, human-induced pluripotent stem cells, and direct reprogramming, provides an ideal tool to model PD, creating the possibility of mimicking key essential pathological processes and charactering single-cell changes in vitro. Furthermore, thanks to the understanding of molecular neuropathogenesis of PD and new advances in stem-cell technology, it is anticipated that optimal functionally transplanted DAn with targeted correction and transgene-free insertion will be generated for use in cell transplantation. This review elucidates stem-cell technology for modeling PD and offering desired safe cell resources for cell transplantation therapy.
Collapse
Affiliation(s)
- Qingxi Zhang
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Wanling Chen
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Sheng Tan
- 2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Tongxiang Lin
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,3 Stem Cell Research Center, Fujian Agriculture and Forestry University , Fuzhou, China
| |
Collapse
|
33
|
Modeling psychiatric disorders: from genomic findings to cellular phenotypes. Mol Psychiatry 2016; 21:1167-79. [PMID: 27240529 PMCID: PMC4995546 DOI: 10.1038/mp.2016.89] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/15/2022]
Abstract
Major programs in psychiatric genetics have identified >150 risk loci for psychiatric disorders. These loci converge on a small number of functional pathways, which span conventional diagnostic criteria, suggesting a partly common biology underlying schizophrenia, autism and other psychiatric disorders. Nevertheless, the cellular phenotypes that capture the fundamental features of psychiatric disorders have not yet been determined. Recent advances in genetics and stem cell biology offer new prospects for cell-based modeling of psychiatric disorders. The advent of cell reprogramming and induced pluripotent stem cells (iPSC) provides an opportunity to translate genetic findings into patient-specific in vitro models. iPSC technology is less than a decade old but holds great promise for bridging the gaps between patients, genetics and biology. Despite many obvious advantages, iPSC studies still present multiple challenges. In this expert review, we critically review the challenges for modeling of psychiatric disorders, potential solutions and how iPSC technology can be used to develop an analytical framework for the evaluation and therapeutic manipulation of fundamental disease processes.
Collapse
|
34
|
Jin S, Collin J, Zhu L, Montaner D, Armstrong L, Neganova I, Lako M. A Novel Role for miR-1305 in Regulation of Pluripotency-Differentiation Balance, Cell Cycle, and Apoptosis in Human Pluripotent Stem Cells. Stem Cells 2016; 34:2306-17. [PMID: 27339422 PMCID: PMC5031214 DOI: 10.1002/stem.2444] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/24/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022]
Abstract
Human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) are defined as pluripotent in view of their self‐renewal ability and potential to differentiate to cells of all three germ layers. Recent studies have indicated that microRNAs (miRNAs) play an important role in the maintenance of pluripotency and cell cycle regulation. We used a microarray based approach to identify miRNAs that were enriched in hESCs when compared to differentiated cells and at the same time showed significant expression changes between different phases of cell cycle. We identified 34 candidate miRNAs and performed functional studies on one of these, miR‐1305, which showed the highest expression change during cell cycle transition. Overexpression of miR‐1305 induced differentiation of pluripotent stem cells, increased cell apoptosis and sped up G1/S transition, while its downregulation facilitated the maintenance of pluripotency and increased cell survival. Using target prediction software and luciferase based reporter assays we identified POLR3G as a downstream target by which miR‐1305 regulates the fine balance between maintenance of pluripotency and onset of differentiation. Overexpression of POLR3G rescued pluripotent stem cell differentiation induced by miR‐1305 overexpression. In contrast, knock‐down of POLR3G expression abolished the miR‐1305‐knockdown mediated enhancement of pluripotency, thus validating its role as miR‐1305 target in human pluripotent stem cells. Together our data point to an important role for miR‐1305 as a novel regulator of pluripotency, cell survival and cell cycle and uncovers new mechanisms and networks by which these processes are intertwined in human pluripotent stem cells. Stem Cells2016;34:2306–2317
Collapse
Affiliation(s)
- Shibo Jin
- Institute of Genetic Medicine, Newcastle University, UK
| | - Joseph Collin
- Institute of Genetic Medicine, Newcastle University, UK
| | - Lili Zhu
- Institute of Genetic Medicine, Newcastle University, UK
| | - David Montaner
- Centro De Investigacion Principe Felipe, Valencia, Spain
| | | | - Irina Neganova
- Institute of Genetic Medicine, Newcastle University, UK.
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, UK.
| |
Collapse
|
35
|
Laurila E, Ahola A, Hyttinen J, Aalto-Setälä K. Methods for in vitro functional analysis of iPSC derived cardiomyocytes - Special focus on analyzing the mechanical beating behavior. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1864-72. [PMID: 26707468 DOI: 10.1016/j.bbamcr.2015.12.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 12/09/2015] [Accepted: 12/16/2015] [Indexed: 02/06/2023]
Abstract
A rapidly increasing number of papers describing novel iPSC models for cardiac diseases are being published. To be able to understand the disease mechanisms in more detail, we should also take the full advantage of the various methods for analyzing these cell models. The traditionally and commonly used electrophysiological analysis methods have been recently accompanied by novel approaches for analyzing the mechanical beatingbehavior of the cardiomyocytes. In this review, we provide first a concise overview on the methodology for cardiomyocyte functional analysis and then concentrate on the video microscopy, which provides a promise for a new faster yet reliable method for cardiomyocyte functional analysis. We also show how analysis conditions may affect the results. Development of the methodology not only serves the basic research on the disease models, but could also provide the much needed efficient early phase screening method for cardiac safety toxicology. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Eeva Laurila
- University of Tampere, BioMediTech and School of Medicine, Tampere, Finland.
| | - Antti Ahola
- Tampere University of Technology, Department of Electronics and Communications Engineering, BioMediTech, Tampere, Finland
| | - Jari Hyttinen
- Tampere University of Technology, Department of Electronics and Communications Engineering, BioMediTech, Tampere, Finland
| | - Katriina Aalto-Setälä
- University of Tampere, BioMediTech and School of Medicine, Tampere, Finland; Heart Hospital, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
36
|
Yeger H. Come together, right now…. J Cell Commun Signal 2015. [PMID: 26208947 DOI: 10.1007/s12079-015-0301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Editing the genome using approaches like TALEN and siRNA are already well tested. The new kid on the block is CRISPR-Cas9. CRISPR-Cas9 is rapidly evolving with impressive refinements for specificity, eliminating off-target effects, and versatility. One can adjust constructs and conditions to produce opposite effects on the genome and for a specific purpose. The nuances of the system, the means to significantly reduce off-targeting, and numerous applications are now emerging rapidly. This B&B commentary looks forward into how the CRISPR-Cas9 tool might serve the CCN field.
Collapse
Affiliation(s)
- Herman Yeger
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|