1
|
Suominen A, Saldo Rubio G, Ruohonen S, Szabó Z, Pohjolainen L, Ghimire B, Ruohonen ST, Saukkonen K, Ijas J, Skarp S, Kaikkonen L, Cai M, Wardlaw SL, Ruskoaho H, Talman V, Savontaus E, Kerkelä R, Rinne P. α-Melanocyte-stimulating hormone alleviates pathological cardiac remodeling via melanocortin 5 receptor. EMBO Rep 2024; 25:1987-2014. [PMID: 38454158 PMCID: PMC11014855 DOI: 10.1038/s44319-024-00109-6] [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: 01/19/2024] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
α-Melanocyte-stimulating hormone (α-MSH) regulates diverse physiological functions by activating melanocortin receptors (MC-R). However, the role of α-MSH and its possible target receptors in the heart remain completely unknown. Here we investigate whether α-MSH could be involved in pathological cardiac remodeling. We found that α-MSH was highly expressed in the mouse heart with reduced ventricular levels after transverse aortic constriction (TAC). Administration of a stable α-MSH analog protected mice against TAC-induced cardiac hypertrophy and systolic dysfunction. In vitro experiments revealed that MC5-R in cardiomyocytes mediates the anti-hypertrophic signaling of α-MSH. Silencing of MC5-R in cardiomyocytes induced hypertrophy and fibrosis markers in vitro and aggravated TAC-induced cardiac hypertrophy and fibrosis in vivo. Conversely, pharmacological activation of MC5-R improved systolic function and reduced cardiac fibrosis in TAC-operated mice. In conclusion, α-MSH is expressed in the heart and protects against pathological cardiac remodeling by activating MC5-R in cardiomyocytes. These results suggest that analogs of naturally occurring α-MSH, that have been recently approved for clinical use and have agonistic activity at MC5-R, may be of benefit in treating heart failure.
Collapse
Affiliation(s)
- Anni Suominen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Drug Research Doctoral Programme (DRDP), University of Turku, Turku, Finland
| | - Guillem Saldo Rubio
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Saku Ruohonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Bishwa Ghimire
- Institute for Molecular Medicine Finland (FIMM), HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Faculty of Medicine, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Karla Saukkonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jani Ijas
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sini Skarp
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Minying Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Eriika Savontaus
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Petteri Rinne
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
2
|
Pohjolainen L, Ruskoaho H, Talman V. Transcriptomics reveal stretched human pluripotent stem cell-derived cardiomyocytes as an advantageous hypertrophy model. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 2:100020. [PMID: 39802492 PMCID: PMC11708431 DOI: 10.1016/j.jmccpl.2022.100020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 01/16/2025]
Abstract
Left ventricular hypertrophy, characterized by hypertrophy of individual cardiomyocytes, is an adaptive response to an increased cardiac workload that eventually leads to heart failure. Previous studies using neonatal rat ventricular myocytes (NRVMs) and animal models have revealed several genes and signaling pathways associated with hypertrophy and mechanical load. However, these models are not directly applicable to humans. Here, we studied the effect of cyclic mechanical stretch on gene expression of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using RNA sequencing. hiPSC-CMs showed distinct hypertrophic changes in gene expression at the level of individual genes and in biological processes. We also identified several differentially expressed genes that have not been previously associated with cardiomyocyte hypertrophy and thus serve as attractive targets for future studies. When compared to previously published data attained from stretched NRVMs and human embryonic stem cell-derived cardiomyocytes, hiPSC-CMs displayed a smaller number of changes in gene expression, but the differentially expressed genes revealed more pronounced enrichment of hypertrophy-related biological processes and pathways. Overall, these results establish hiPSC-CMs as a valuable in vitro model for studying human cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
3
|
Li J, Wiesinger A, Fokkert L, Boukens BJ, Verkerk AO, Christoffels VM, Boink GJ, Devalla HD. Molecular and electrophysiological evaluation of human cardiomyocyte subtypes to facilitate generation of composite cardiac models. J Tissue Eng 2022; 13:20417314221127908. [PMID: 36277058 PMCID: PMC9583221 DOI: 10.1177/20417314221127908] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/06/2022] [Indexed: 11/06/2022] Open
Abstract
Paucity of physiologically relevant cardiac models has limited the widespread application of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes in drug development. Here, we performed comprehensive characterization of hiPSC-derived cardiomyocyte subtypes from 2D and 3D cultures and established a novel 3D model to study impulse initiation and propagation. Directed differentiation approaches were used to generate sinoatrial nodal (SANCM), atrial (ACM) and ventricular cardiomyocytes (VCM). Single cell RNA sequencing established that the protocols yield distinct cell populations in line with expected identities, which was also confirmed by electrophysiological characterization. In 3D EHT cultures of all subtypes, we observed prominent expression of stretch-responsive genes such as NPPA. Response to rate modulating drugs noradrenaline, carbachol and ivabradine were comparable in single cells and EHTs. Differences in the speed of impulse propagation between the subtypes were more pronounced in EHTs compared with 2D monolayers owing to a progressive increase in conduction velocities in atrial and ventricular cardiomyocytes, in line with a more mature phenotype. In a novel binary EHT model of pacemaker-atrial interface, the SANCM end of the tissue consistently paced the EHTs under baseline conditions, which was inhibited by ivabradine. Taken together, our data provide comprehensive insights into molecular and electrophysiological properties of hiPSC-derived cardiomyocyte subtypes, facilitating the creation of next generation composite cardiac models for drug discovery, disease modeling and cell-based regenerative therapies.
Collapse
Affiliation(s)
- Jiuru Li
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Alexandra Wiesinger
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Lianne Fokkert
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Bastiaan J. Boukens
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Arie O. Verkerk
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands,Department of Experimental Cardiology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Vincent M. Christoffels
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Gerard J.J. Boink
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands,Department of Cardiology, Amsterdam
University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands
| | - Harsha D. Devalla
- Department of Medical Biology,
Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The
Netherlands,Harsha D Devalla, Department of Medical
Biology, Amsterdam University Medical Centers, University of Amsterdam,
Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands.
| |
Collapse
|
4
|
Gui Y, Zhang Y, Zhang Q, Chen X, Wang F, Wu F, Gui Y, Li Q. The functional verification and analysis of Fugu promoter of cardiac gene tnni1a in zebrafish. Cells Dev 2022; 171:203801. [PMID: 35787465 DOI: 10.1016/j.cdev.2022.203801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/09/2022] [Accepted: 06/28/2022] [Indexed: 01/25/2023]
Abstract
Troponin I type 1b (Tnni1b) is thought to be a novel isoform that is expressed only in the zebrafish heart. Knocking down of tnni1b can lead to cardiac defects in zebrafish. Although both the zebrafish tnni1b and human troponin I1 (TNNI1) genes are thought to be closely associated with fatal cardiac development, the regulatory molecular mechanisms of these genes are poorly understood. Analyzing the functionally conserved sequence, especially in the noncoding regulatory region involved in gene expression, clarified these mechanisms. In this study, we isolated a 3 kb fragment upstream of Fugu tnni1a that can regulate green fluorescence protein (GFP) expression in a heart-specific manner, similar to the pattern of zebrafish homologue expression. Three evolutionarily conserved regions (ECRs) in the 5'-flanking sequence of Fugu tnni1a were identified by sequence alignment. Deletion analysis led to the identification of ECR2 as a core sequence that affects the heart-specific expression function of the Fugu tnni1a promoter. Interestingly, both the Fugu tnni1a promoter and ECR2 sequence were functionally conserved in zebrafish, although they shared no sequence similarity. Together, the findings of our study provided further evidence for the important role of tnni1a homologous in cardiac development and demonstrated that two functionally conserved sequences in the zebrafish and Fugu genomes may be ECRs, despite their lack of similarity.
Collapse
Affiliation(s)
- Yiting Gui
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect Prevention and Control, NHC Key Laboratory of Neonatal Diseases, Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China; Cardiovascular Center, NHC Key Laboratory of Neonatal Diseases, Fudan University, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yawen Zhang
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect Prevention and Control, NHC Key Laboratory of Neonatal Diseases, Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China; Cardiovascular Center, NHC Key Laboratory of Neonatal Diseases, Fudan University, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Qi Zhang
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect Prevention and Control, NHC Key Laboratory of Neonatal Diseases, Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Xudong Chen
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect Prevention and Control, NHC Key Laboratory of Neonatal Diseases, Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Feng Wang
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect Prevention and Control, NHC Key Laboratory of Neonatal Diseases, Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China; Cardiovascular Center, NHC Key Laboratory of Neonatal Diseases, Fudan University, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Fang Wu
- Department of Neonatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Yonghao Gui
- Cardiovascular Center, NHC Key Laboratory of Neonatal Diseases, Fudan University, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China.
| | - Qiang Li
- Translational Medical Center for Development and Disease, Shanghai Key Laboratory of Birth Defect Prevention and Control, NHC Key Laboratory of Neonatal Diseases, Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China.
| |
Collapse
|
5
|
Kowalski WJ, Garcia-Pak IH, Li W, Uosaki H, Tampakakis E, Zou J, Lin Y, Patterson K, Kwon C, Mukouyama YS. Sympathetic Neurons Regulate Cardiomyocyte Maturation in Culture. Front Cell Dev Biol 2022; 10:850645. [PMID: 35359438 PMCID: PMC8961983 DOI: 10.3389/fcell.2022.850645] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/02/2022] [Indexed: 12/20/2022] Open
Abstract
Embryos devoid of autonomic innervation suffer sudden cardiac death. However, whether autonomic neurons have a role in heart development is poorly understood. To investigate if sympathetic neurons impact cardiomyocyte maturation, we co-cultured phenotypically immature cardiomyocytes derived from human induced pluripotent stem cells with mouse sympathetic ganglion neurons. We found that 1) multiple cardiac structure and ion channel genes related to cardiomyocyte maturation were up-regulated when co-cultured with sympathetic neurons; 2) sarcomere organization and connexin-43 gap junctions increased; 3) calcium imaging showed greater transient amplitudes. However, sarcomere spacing, relaxation time, and level of sarcoplasmic reticulum calcium did not show matured phenotypes. We further found that addition of endothelial and epicardial support cells did not enhance maturation to a greater extent beyond sympathetic neurons, while administration of isoproterenol alone was insufficient to induce changes in gene expression. These results demonstrate that sympathetic neurons have a significant and complex role in regulating cardiomyocyte development.
Collapse
Affiliation(s)
- William J. Kowalski
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Iris H. Garcia-Pak
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hideki Uosaki
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States,Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Emmanouil Tampakakis
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jizhong Zou
- IPSC Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yongshun Lin
- IPSC Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kira Patterson
- IPSC Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States,*Correspondence: Yoh-Suke Mukouyama,
| |
Collapse
|
6
|
Bourque K, Hawey C, Jones-Tabah J, Pétrin D, Martin RD, Ling Sun Y, Hébert TE. Measuring hypertrophy in neonatal rat primary cardiomyocytes and human iPSC-derived cardiomyocytes. Methods 2021; 203:447-464. [PMID: 34933120 DOI: 10.1016/j.ymeth.2021.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
In the heart, left ventricular hypertrophy is initially an adaptive mechanism that increases wall thickness to preserve normal cardiac output and function in the face of coronary artery disease or hypertension. Cardiac hypertrophy develops in response to pressure and volume overload but can also be seen in inherited cardiomyopathies. As the wall thickens, it becomes stiffer impairing the distribution of oxygenated blood to the rest of the body. With complex cellular signalling and transcriptional networks involved in the establishment of the hypertrophic state, several model systems have been developed to better understand the molecular drivers of disease. Immortalized cardiomyocyte cell lines, primary rodent and larger animal models have all helped understand the pathological mechanisms underlying cardiac hypertrophy. Induced pluripotent stem cell-derived cardiomyocytes are also used and have the additional benefit of providing access to human samples with direct disease relevance as when generated from patients suffering from hypertrophic cardiomyopathies. Here, we briefly review in vitro and in vivo model systems that have been used to model hypertrophy and provide detailed methods to isolate primary neonatal rat cardiomyocytes as well as to generate cardiomyocytes from human iPSCs. We also describe how to model hypertrophy in a "dish" using gene expression analysis and immunofluorescence combined with automated high-content imaging.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Yi Ling Sun
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
7
|
Environmental Alterations during Embryonic Development: Studying the Impact of Stressors on Pluripotent Stem Cell-Derived Cardiomyocytes. Genes (Basel) 2021; 12:genes12101564. [PMID: 34680959 PMCID: PMC8536136 DOI: 10.3390/genes12101564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022] Open
Abstract
Non-communicable diseases (NCDs) sauch as diabetes, obesity and cardiovascular diseases are rising rapidly in all countries world-wide. Environmental maternal factors (e.g., diet, oxidative stress, drugs and many others), maternal illnesses and other stressors can predispose the newborn to develop diseases during different stages of life. The connection between environmental factors and NCDs was formulated by David Barker and colleagues as the Developmental Origins of Health and Disease (DOHaD) hypothesis. In this review, we describe the DOHaD concept and the effects of several environmental stressors on the health of the progeny, providing both animal and human evidence. We focus on cardiovascular diseases which represent the leading cause of death worldwide. The purpose of this review is to discuss how in vitro studies with pluripotent stem cells (PSCs), such as embryonic and induced pluripotent stem cells (ESC, iPSC), can underpin the research on non-genetic heart conditions. The PSCs could provide a tool to recapitulate aspects of embryonic development “in a dish”, studying the effects of environmental exposure during cardiomyocyte (CM) differentiation and maturation, establishing a link to molecular mechanism and epigenetics.
Collapse
|
8
|
ERRγ enhances cardiac maturation with T-tubule formation in human iPSC-derived cardiomyocytes. Nat Commun 2021; 12:3596. [PMID: 34155205 PMCID: PMC8217550 DOI: 10.1038/s41467-021-23816-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/17/2021] [Indexed: 01/17/2023] Open
Abstract
One of the earliest maturation steps in cardiomyocytes (CMs) is the sarcomere protein isoform switch between TNNI1 and TNNI3 (fetal and neonatal/adult troponin I). Here, we generate human induced pluripotent stem cells (hiPSCs) carrying a TNNI1EmGFP and TNNI3mCherry double reporter to monitor and isolate mature sub-populations during cardiac differentiation. Extensive drug screening identifies two compounds, an estrogen-related receptor gamma (ERRγ) agonist and an S-phase kinase-associated protein 2 inhibitor, that enhances cardiac maturation and a significant change to TNNI3 expression. Expression, morphological, functional, and molecular analyses indicate that hiPSC-CMs treated with the ERRγ agonist show a larger cell size, longer sarcomere length, the presence of transverse tubules, and enhanced metabolic function and contractile and electrical properties. Here, we show that ERRγ-treated hiPSC-CMs have a mature cellular property consistent with neonatal CMs and are useful for disease modeling and regenerative medicine. Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSCs) suffer from limited maturation. Here the authors identify ERRγ agonist as a factor that enhances cardiac morphological, metabolic, contractile and electrical maturation of hiPSC-derived CMs with T-tubule formation.
Collapse
|
9
|
Orsolits B, Kovács Z, Kriston-Vizi J, Merkely B, Földes G. New Modalities of 3D Pluripotent Stem Cell-Based Assays in Cardiovascular Toxicity. Front Pharmacol 2021; 12:603016. [PMID: 33854431 PMCID: PMC8039822 DOI: 10.3389/fphar.2021.603016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/04/2021] [Indexed: 12/04/2022] Open
Abstract
The substantial progress of the human induced pluripotent stem cell (hiPSC) technologies over the last decade has provided us with new opportunities for cardiovascular drug discovery, regenerative medicine, and disease modeling. The combination of hiPSC with 3D culture techniques offers numerous advantages for generating and studying physiological and pathophysiological cardiac models. Cells grown in 3D can overcome many limitations of 2D cell cultures and animal models. Furthermore, it enables the investigation in an architecturally appropriate, complex cellular environment in vitro. Yet, generation and study of cardiac organoids-which may contain versatile cardiovascular cell types differentiated from hiPSC-remain a challenge. The large-scale and high-throughput applications require accurate and standardised models with highly automated processes in culturing, imaging and data collection. Besides the compound spatial structure of organoids, their biological processes also possess different temporal dynamics which require other methods and technologies to detect them. In this review, we summarise the possibilities and challenges of acquiring relevant information from 3D cardiovascular models. We focus on the opportunities during different time-scale processes in dynamic pharmacological experiments and discuss the putative steps toward one-size-fits-all assays.
Collapse
Affiliation(s)
- Barbara Orsolits
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - Zsófia Kovács
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - János Kriston-Vizi
- Bioinformatics Image Core (BIONIC), MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
| | - Gábor Földes
- Heart and Vascular Center, Semmelweis University Budapest, Budapest, Hungary
- National Heart and Lung Institute, Imperial Centre for Experimental and Translational Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
10
|
Gao Q, Wang P, Qiu H, Qiu B, Yi W, Tu W, Lin B, Sun D, Zeng R, Huang M, Chen J, Cen J, Zhuang J. Myogenin suppresses apoptosis induced by angiotensin II in human induced pluripotent stem cell-derived cardiomyocytes. Biochem Biophys Res Commun 2021; 552:84-90. [PMID: 33743352 DOI: 10.1016/j.bbrc.2021.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Angiotensin II (Ang II), an important component of the renin-angiotensin system (RAS), plays a critical role in the pathogenesis of cardiovascular disorders. In addition, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been considered as a promising platform for studying personalized medicine for heart diseases. However, whether Ang II can induce the apoptosis of hiPSC-CMs is not known. METHODS In this study, we treated hiPSC-CMs with different concentrations of Ang II [0 nM (vehicle as a control), 1 nM, 10 nM, 100 nM, 1 μM, 10 μM, 100 μM, and 1 mM] for various time periods (24 h, 48 h, 6 days, and 10 days) and analyzed the viability and apoptosis of hiPSC-CMs. RESULTS We found that treatment with 1 mM Ang II for 10 days reduced the viability of hiPSC-CMs by 41% (p = 2.073E-08) and increased apoptosis by 2.74-fold, compared to the control group (p = 6.248E-12). MYOG, which encodes the muscle-specific transcription factor myogenin, was also identified as an apoptosis-suppressor gene in Ang II-treated hiPSC-CMs. Ectopic MYOG expression decreased the apoptosis and increased the viability of Ang II-treated hiPSC-CMs. Further analysis of the RNA sequencing (RNA-seq) data illustrated that myogenin ameliorated Ang II-induced apoptosis of hiPSC-CMs by downregulating the expression of proinflammatory genes. CONCLUSION Our findings suggest that Ang II induces the apoptosis of hiPSC-CMs and that myogenin attenuates Ang II-induced apoptosis.
Collapse
Affiliation(s)
- Qiang Gao
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Ping Wang
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Hailong Qiu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Bin Qiu
- Department of Mechanical & Electrical Engineering, Xiamen University, Xiamen, Fujian, 361102, China
| | - Weijin Yi
- Department of Mechanical & Electrical Engineering, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenchang Tu
- Department of Mechanical & Electrical Engineering, Xiamen University, Xiamen, Fujian, 361102, China
| | - Bin Lin
- Guangdong Beating Origin Regenerative Medicine Co. Ltd., Foshan, Guangdong, 528231, China
| | - Daoheng Sun
- Department of Mechanical & Electrical Engineering, Xiamen University, Xiamen, Fujian, 361102, China
| | - Rong Zeng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Meiping Huang
- Department of Catheterization Lab, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences Guangzhou, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Jianzheng Cen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China.
| | - Jian Zhuang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China.
| |
Collapse
|
11
|
Prajapati C, Ojala M, Lappi H, Aalto-Setälä K, Pekkanen-Mattila M. Electrophysiological evaluation of human induced pluripotent stem cell-derived cardiomyocytes obtained by different methods. Stem Cell Res 2021; 51:102176. [PMID: 33485184 DOI: 10.1016/j.scr.2021.102176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/21/2020] [Accepted: 01/09/2021] [Indexed: 11/30/2022] Open
Abstract
The human induced pluripotent stem cells (hiPSCs) derived cardiomyocytes (CMs) (hiPSC-CMs) retain the same genetic information as the donor, and they have been shown to faithfully recapitulate the disease phenotypes of various genetic cardiac diseases. The hiPSC-CMs can be utilized in multiple types of studies and in most cases, the functionality of hiPSC-CMs is of interest. For the functional analyses, the hiPSC-CMs need to be manipulated after differentiation, e.g. enriched or dissociated into single-cell stage. For the functional assessments to be reliable and reproducible, the cell culture environment should support the cells in an optimal manner. The aim of the present study was to evaluate the effect of various differentiation methods, as well as coating materials used for the dissociated cells on the functionality of the differentiated hiPSC-CMs. The different protocols not only had different differentiation efficiencies, but they also yielded functionally different hiPSC-CMs. Additionally, the coating material had a major effect on the functionality of the hiPSC-CMs. The results of the present study emphasize that the cardiac differentiation method and the coating material have a major effect on hiPS-CMs' characteristics. Thus, when different hiPSC lines and results obtained in different labs are compared, extra care should be taken to check the conditions when results are compared.
Collapse
Affiliation(s)
- Chandra Prajapati
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, Tampere, Finland.
| | - Marisa Ojala
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, Tampere, Finland
| | - Henna Lappi
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, Tampere, Finland.
| | - Katriina Aalto-Setälä
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, Tampere, Finland; Heart Hospital, Tampere University Hospital, Tampere, Finland.
| | - Mari Pekkanen-Mattila
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, Tampere, Finland.
| |
Collapse
|
12
|
Johansson M, Ulfenborg B, Andersson CX, Heydarkhan-Hagvall S, Jeppsson A, Sartipy P, Synnergren J. Cardiac hypertrophy in a dish: a human stem cell based model. Biol Open 2020; 9:bio052381. [PMID: 32878883 PMCID: PMC7522030 DOI: 10.1242/bio.052381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/23/2020] [Indexed: 12/23/2022] Open
Abstract
Cardiac hypertrophy is an important and independent risk factor for the development of heart failure. To better understand the mechanisms and regulatory pathways involved in cardiac hypertrophy, there is a need for improved in vitro models. In this study, we investigated how hypertrophic stimulation affected human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs). The cells were stimulated with endothelin-1 (ET-1) for 8, 24, 48, 72, or 96 h. Parameters including cell size, ANP-, proBNP-, and lactate concentration were analyzed. Moreover, transcriptional profiling using RNA-sequencing was performed to identify differentially expressed genes following ET-1 stimulation. The results show that the CMs increase in size by approximately 13% when exposed to ET-1 in parallel to increases in ANP and proBNP protein and mRNA levels. Furthermore, the lactate concentration in the media was increased indicating that the CMs consume more glucose, a hallmark of cardiac hypertrophy. Using RNA-seq, a hypertrophic gene expression pattern was also observed in the stimulated CMs. Taken together, these results show that hiPSC-derived CMs stimulated with ET-1 display a hypertrophic response. The results from this study also provide new molecular insights about the underlying mechanisms of cardiac hypertrophy and may help accelerate the development of new drugs against this condition.
Collapse
Affiliation(s)
- Markus Johansson
- Systems Biology Research Center, School of Bioscience, Department for Biology and Bioinformatics, University of Skövde, SE-541 28 Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Benjamin Ulfenborg
- Systems Biology Research Center, School of Bioscience, Department for Biology and Bioinformatics, University of Skövde, SE-541 28 Skövde, Sweden
| | | | - Sepideh Heydarkhan-Hagvall
- Systems Biology Research Center, School of Bioscience, Department for Biology and Bioinformatics, University of Skövde, SE-541 28 Skövde, Sweden
- Bioscience, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals, R&D AstraZeneca, 431 50 Gothenburg, Sweden
| | - Anders Jeppsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, 405 30 Gothenburg, Sweden
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Peter Sartipy
- Systems Biology Research Center, School of Bioscience, Department for Biology and Bioinformatics, University of Skövde, SE-541 28 Skövde, Sweden
- Late-stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, Department for Biology and Bioinformatics, University of Skövde, SE-541 28 Skövde, Sweden
| |
Collapse
|
13
|
Martewicz S, Magnussen M, Elvassore N. Beyond Family: Modeling Non-hereditary Heart Diseases With Human Pluripotent Stem Cell-Derived Cardiomyocytes. Front Physiol 2020; 11:384. [PMID: 32390874 PMCID: PMC7188911 DOI: 10.3389/fphys.2020.00384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/30/2020] [Indexed: 12/23/2022] Open
Abstract
Non-genetic cardiac pathologies develop as an aftermath of extracellular stress-conditions. Nevertheless, the response to pathological stimuli depends deeply on intracellular factors such as physiological state and complex genetic backgrounds. Without a thorough characterization of their in vitro phenotype, modeling of maladaptive hypertrophy, ischemia and reperfusion injury or diabetes in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) has been more challenging than hereditary diseases with defined molecular causes. In past years, greater insights into hPSC-CM in vitro physiology and advancements in technological solutions and culture protocols have generated cell types displaying stress-responsive phenotypes reminiscent of in vivo pathological events, unlocking their application as a reductionist model of human cardiomyocytes, if not the adult human myocardium. Here, we provide an overview of the available literature of pathology models for cardiac non-genetic conditions employing healthy (or asymptomatic) hPSC-CMs. In terms of numbers of published articles, these models are significantly lagging behind monogenic diseases, which misrepresents the incidence of heart disease causes in the human population.
Collapse
Affiliation(s)
- Sebastian Martewicz
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China
| | - Michael Magnussen
- Stem Cells & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Nicola Elvassore
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China.,Stem Cells & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Industrial Engineering, University of Padova, Padua, Italy
| |
Collapse
|
14
|
Nemade H, Acharya A, Chaudhari U, Nembo E, Nguemo F, Riet N, Abken H, Hescheler J, Papadopoulos S, Sachinidis A. Cyclooxygenases Inhibitors Efficiently Induce Cardiomyogenesis in Human Pluripotent Stem Cells. Cells 2020; 9:cells9030554. [PMID: 32120775 PMCID: PMC7140528 DOI: 10.3390/cells9030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/28/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Application of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is limited by the challenges in their efficient differentiation. Recently, the Wingless (Wnt) signaling pathway has emerged as the key regulator of cardiomyogenesis. In this study, we evaluated the effects of cyclooxygenase inhibitors on cardiac differentiation of hPSCs. Cardiac differentiation was performed by adherent monolayer based method using 4 hPSC lines (HES3, H9, IMR90, and ES4SKIN). The efficiency of cardiac differentiation was evaluated by flow cytometry and RT-qPCR. Generated hPSC-CMs were characterised using immunocytochemistry, electrophysiology, electron microscopy, and calcium transient measurements. Our data show that the COX inhibitors Sulindac and Diclofenac in combination with CHIR99021 (GSK-3 inhibitor) efficiently induce cardiac differentiation of hPSCs. In addition, inhibition of COX using siRNAs targeted towards COX-1 and/or COX-2 showed that inhibition of COX-2 alone or COX-1 and COX-2 in combination induce cardiomyogenesis in hPSCs within 12 days. Using IMR90-Wnt reporter line, we showed that inhibition of COX-2 led to downregulation of Wnt signalling activity in hPSCs. In conclusion, this study demonstrates that COX inhibition efficiently induced cardiogenesis via modulation of COX and Wnt pathway and the generated cardiomyocytes express cardiac-specific structural markers as well as exhibit typical calcium transients and action potentials. These cardiomyocytes also responded to cardiotoxicants and can be relevant as an in vitro cardiotoxicity screening model.
Collapse
Affiliation(s)
- Harshal Nemade
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Aviseka Acharya
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Umesh Chaudhari
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Erastus Nembo
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Filomain Nguemo
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Nicole Riet
- Department I Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 21, 50931 Cologne, Germany;
| | - Hinrich Abken
- Regensburg Centre for Interventional Immunology (RCI), Deptartment Genetic Immunotherapy, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Jürgen Hescheler
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Symeon Papadopoulos
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
| | - Agapios Sachinidis
- Institute of Neurophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany; (H.N.); (A.A.); (U.C.); (E.N.); (F.N.); (J.H.); (S.P.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Correspondence: ; Tel.: +49-0221-4787373
| |
Collapse
|
15
|
High-Throughput Phenotyping Toolkit for Characterizing Cellular Models of Hypertrophic Cardiomyopathy In Vitro. Methods Protoc 2019; 2:mps2040083. [PMID: 31717790 PMCID: PMC6961126 DOI: 10.3390/mps2040083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/18/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a prevalent and complex cardiovascular disease characterised by multifarious hallmarks, a heterogeneous set of clinical manifestations, and several molecular mechanisms. Various disease models have been developed to study this condition, but they often show contradictory results, due to technical constraints and/or model limitations. Therefore, new tools are needed to better investigate pathological features in an unbiased and technically refined approach, towards improving understanding of disease progression. Herein, we describe three simple protocols to phenotype cellular models of HCM in vitro, in a high-throughput manner where technical artefacts are minimized. These are aimed at investigating: (1) Hypertrophy, by measuring cell volume by flow cytometry; (2) HCM molecular features, through the analysis of a hypertrophic marker, multinucleation, and sarcomeric disarray by high-content imaging; and (3) mitochondrial respiration and content via the Seahorse™ platform. Collectively, these protocols comprise straightforward tools to evaluate molecular and functional parameters of HCM phenotypes in cardiomyocytes in vitro. These facilitate greater understanding of HCM and high-throughput drug screening approaches and are accessible to all researchers of cardiac disease modelling. Whilst HCM is used as an exemplar, the approaches described are applicable to other cellular models where the investigation of identical biological changes is paramount.
Collapse
|
16
|
Gintant G, Burridge P, Gepstein L, Harding S, Herron T, Hong C, Jalife J, Wu JC. Use of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes in Preclinical Cancer Drug Cardiotoxicity Testing: A Scientific Statement From the American Heart Association. Circ Res 2019; 125:e75-e92. [PMID: 31533542 DOI: 10.1161/res.0000000000000291] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
It is now well recognized that many lifesaving oncology drugs may adversely affect the heart and cardiovascular system, including causing irreversible cardiac injury that can result in reduced quality of life. These effects, which may manifest in the short term or long term, are mechanistically not well understood. Research is hampered by the reliance on whole-animal models of cardiotoxicity that may fail to reflect the fundamental biology or cardiotoxic responses of the human myocardium. The emergence of human induced pluripotent stem cell-derived cardiomyocytes as an in vitro research tool holds great promise for understanding drug-induced cardiotoxicity of oncological drugs that may manifest as contractile and electrophysiological dysfunction, as well as structural abnormalities, making it possible to deliver novel drugs free from cardiac liabilities and guide personalized therapy. This article briefly reviews the challenges of cardio-oncology, the strengths and limitations of using human induced pluripotent stem cell-derived cardiomyocytes to represent clinical findings in the nonclinical research space, and future directions for their further use.
Collapse
|
17
|
Ribeiro AJS, Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Brock M, Chaudhary KW, Kanda Y, Berridge B. Considerations for an In Vitro, Cell-Based Testing Platform for Detection of Drug-Induced Inotropic Effects in Early Drug Development. Part 2: Designing and Fabricating Microsystems for Assaying Cardiac Contractility With Physiological Relevance Using Human iPSC-Cardiomyocytes. Front Pharmacol 2019; 10:934. [PMID: 31555128 PMCID: PMC6727630 DOI: 10.3389/fphar.2019.00934] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
Contractility of the myocardium engines the pumping function of the heart and is enabled by the collective contractile activity of its muscle cells: cardiomyocytes. The effects of drugs on the contractility of human cardiomyocytes in vitro can provide mechanistic insight that can support the prediction of clinical cardiac drug effects early in drug development. Cardiomyocytes differentiated from human-induced pluripotent stem cells have high potential for overcoming the current limitations of contractility assays because they attach easily to extracellular materials and last long in culture, while having human- and patient-specific properties. Under these conditions, contractility measurements can be non-destructive and minimally invasive, which allow assaying sub-chronic effects of drugs. For this purpose, the function of cardiomyocytes in vitro must reflect physiological settings, which is not observed in cultured cardiomyocytes derived from induced pluripotent stem cells because of the fetal-like properties of their contractile machinery. Primary cardiomyocytes or tissues of human origin fully represent physiological cellular properties, but are not easily available, do not last long in culture, and do not attach easily to force sensors or mechanical actuators. Microengineered cellular systems with a more mature contractile function have been developed in the last 5 years to overcome this limitation of stem cell-derived cardiomyocytes, while simultaneously measuring contractile endpoints with integrated force sensors/actuators and image-based techniques. Known effects of engineered microenvironments on the maturity of cardiomyocyte contractility have also been discovered in the development of these systems. Based on these discoveries, we review here design criteria of microengineered platforms of cardiomyocytes derived from pluripotent stem cells for measuring contractility with higher physiological relevance. These criteria involve the use of electromechanical, chemical and morphological cues, co-culture of different cell types, and three-dimensional cellular microenvironments. We further discuss the use and the current challenges for developing and improving these novel technologies for predicting clinical effects of drugs based on contractility measurements with cardiomyocytes differentiated from induced pluripotent stem cells. Future research should establish contexts of use in drug development for novel contractility assays with stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Brian D Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.,PreClinical Drug Development Platform (PCDDP), North-West University, Potchefstroom, South Africa
| | - Michael Engwall
- Safety Pharmacology and Animal Research Center, Amgen Research, Thousand Oaks, CA, United States
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - C Michael Foley
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - Liang Guo
- Laboratory of Investigative Toxicology, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - John Koerner
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Stanley T Parish
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, South San Francisco, CA, United States
| | - Khuram W Chaudhary
- Global Safety Pharmacology, GlaxoSmithKline plc, Collegeville, PA, United States
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Brian Berridge
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
18
|
Lyon A. Stress in a Dish: Exploring the Mechanisms of Takotsubo Syndrome. J Am Coll Cardiol 2019; 70:992-995. [PMID: 28818209 DOI: 10.1016/j.jacc.2017.07.716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 01/26/2023]
Affiliation(s)
- Alexander Lyon
- Cardiovascular Research Centre, Royal Brompton Hospital and Imperial College London, London, United Kingdom.
| |
Collapse
|
19
|
The Role of Reactive Oxygen Species in In Vitro Cardiac Maturation. Trends Mol Med 2019; 25:482-493. [PMID: 31080142 DOI: 10.1016/j.molmed.2019.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 12/27/2022]
Abstract
Recent advances in developmental biology and biomedical engineering have significantly improved the efficiency and purity of cardiomyocytes (CMs) generated from pluripotent stem cells (PSCs). Regardless of the protocol used to derive CMs, these cells exhibit hallmarks of functional immaturity. In this Opinion, we focus on reactive oxygen species (ROS), signaling molecules that can potentially modulate cardiac maturation. We outline how ROS impacts nearly every aspect associated with cardiac maturation, including contractility, calcium handling, metabolism, and hypertrophy. Though the precise role of ROS in cardiac maturation has yet to be elucidated, ROS may provide a valuable perspective for understanding the molecular mechanisms for cardiac maturation under various conditions.
Collapse
|
20
|
Prajapati C, Pölönen RP, Aalto-Setälä K. Simultaneous recordings of action potentials and calcium transients from human induced pluripotent stem cell derived cardiomyocytes. Biol Open 2018; 7:bio.035030. [PMID: 29970475 PMCID: PMC6078349 DOI: 10.1242/bio.035030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) offer a unique in vitro platform to study cardiac diseases, as they recapitulate many disease phenotypes. The membrane potential (Vm) and intracellular calcium (Ca2+) transient (CaT) are usually investigated separately, because incorporating different techniques to acquire both aspects concurrently is challenging. In this study, we recorded Vm and CaT simultaneously to understand the interrelation between these parameters in hiPSC-CMs. For this, we used a conventional patch clamp technique to record Vm, and synchronized this with a Ca2+ imaging system to acquire CaT from same hiPSC-CMs. Our results revealed that the CaT at 90% decay (CaT90) was longer than action potential (AP) duration at 90% repolarization (APD90). In addition, there was also a strong positive correlation between the different parameters of CaT and AP. The majority of delayed after depolarizations (DADs) observed in the Vm recording were also characterized by elevations in the intracellular Ca2+ level, but in some cases no abnormalities were observed in CaT. However, simultaneous fluctuations in CaT were always observed during early after depolarizations (EADs) in Vm In summary, simultaneous recording of Vm and CaT broadens the understanding of the interrelation between Vm and CaT and could be used to elucidate the mechanisms underlying arrhythmia in cardiac disease condition.
Collapse
Affiliation(s)
| | | | - Katriina Aalto-Setälä
- BioMediTech, University of Tampere, 33520 Tampere, Finland .,Faculty of Medicine and Life Science, University of Tampere, 33520 Tampere, Finland.,Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland
| |
Collapse
|
21
|
Stem cells are the most sensitive screening tool to identify toxicity of GATA4-targeted novel small-molecule compounds. Arch Toxicol 2018; 92:2897-2911. [PMID: 29987409 PMCID: PMC6132687 DOI: 10.1007/s00204-018-2257-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/04/2018] [Indexed: 01/10/2023]
Abstract
Safety assessment of drug candidates in numerous in vitro and experimental animal models is expensive, time consuming and animal intensive. More thorough toxicity profiling already in the early drug discovery projects using human cell models, which more closely resemble the physiological cell types, would help to decrease drug development costs. In this study we aimed to compare different cardiac and stem cell models for in vitro toxicity testing and to elucidate structure–toxicity relationships of novel compounds targeting the cardiac transcription factor GATA4. By screening the effects of eight compounds at concentrations ranging from 10 nM up to 30 µM on the viability of eight different cell types, we identified significant cell type- and structure-dependent toxicity profiles. We further characterized two compounds in more detail using high-content analysis. The results highlight the importance of cell type selection for toxicity screening and indicate that stem cells represent the most sensitive screening model, which can detect toxicity that may otherwise remain unnoticed. Furthermore, our structure–toxicity analysis reveals a characteristic dihedral angle in the GATA4-targeted compounds that causes stem cell toxicity and thus helps to direct further drug development efforts towards non-toxic derivatives.
Collapse
|
22
|
Rosales W, Lizcano F. The Histone Demethylase JMJD2A Modulates the Induction of Hypertrophy Markers in iPSC-Derived Cardiomyocytes. Front Genet 2018; 9:14. [PMID: 29479368 PMCID: PMC5811633 DOI: 10.3389/fgene.2018.00014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/10/2018] [Indexed: 01/04/2023] Open
Abstract
The development of cardiovascular pathologies is partly attributed to epigenetic causes, including histone methylation, which appears to be an important marker in hearts that develop cardiac hypertrophy. Previous studies showed that the histone demethylase JMJD2A can regulate the hypertrophic process in murine cardiomyocytes. However, the influence of JMJD2A on cardiac hypertrophy in a human cardiomyocyte model is still poorly understood. In the present study, cardiomyocytes derived from human induced pluripotent stem cells (iPSCs) were used. Hypertrophy was induced by angiotensin II and endothelin-1 (ET-1), and transfections were performed to overexpress JMJD2A and for small interfering RNA (siRNA)-induced silencing of JMJD2A. Gene expression analyses were determined using RT-PCR and Western blot. The expression levels of B-type natriuretic peptide (BNP), natriuretic peptide A (ANP), and beta myosin heavy chain (β-MHC) were increased by nearly 2–10-fold with ET-1 compared with the control. However, a higher level of JMJD2A and UTX was detected, whereas the level of JMJD2C was lower. When cardiomyocytes were transiently transfected with JMJD2A, an increase close to 150% in BNP was observed, and this increase was greater after treatment with ET-1. To verify the specificity of JMJD2A activity, a knockdown was performed by means of siRNA-JMJD2A, which led to a significant reduction in BNP. The involvement of JMJD2A suggests that histone-specific modifications are associated with genes encoding proteins that are actively transcribed during the hypertrophy process. Since BNP is closely related to JMJD2A expression, we suggest that there could be a direct influence of JMJD2A on the expression of BNP. These results may be studied further to reduce cardiac hypertrophy via the regulation of epigenetic modifiers.
Collapse
Affiliation(s)
- Wendy Rosales
- Center of Biomedical Research, Universidad de La Sabana, Chía, Colombia
| | - Fernando Lizcano
- Center of Biomedical Research, Universidad de La Sabana, Chía, Colombia
| |
Collapse
|
23
|
Magdy T, Schuldt AJT, Wu JC, Bernstein D, Burridge PW. Human Induced Pluripotent Stem Cell (hiPSC)-Derived Cells to Assess Drug Cardiotoxicity: Opportunities and Problems. Annu Rev Pharmacol Toxicol 2017; 58:83-103. [PMID: 28992430 DOI: 10.1146/annurev-pharmtox-010617-053110] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Billions of US dollars are invested every year by the pharmaceutical industry in drug development, with the aim of introducing new drugs that are effective and have minimal side effects. Thirty percent of in-pipeline drugs are excluded in an early phase of preclinical and clinical screening owing to cardiovascular safety concerns, and several lead molecules that pass the early safety screening make it to market but are later withdrawn owing to severe cardiac side effects. Although the current drug safety screening methodologies can identify some cardiotoxic drug candidates, they cannot accurately represent the human heart in many aspects, including genomics, transcriptomics, and patient- or population-specific cardiotoxicity. Despite some limitations, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are a powerful and evolving technology that has been shown to recapitulate many attributes of human cardiomyocytes and their drug responses. In this review, we discuss the potential impact of the inclusion of the hiPSC-CM platform in premarket candidate drug screening.
Collapse
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA; .,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Adam J T Schuldt
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA; .,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.,Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Daniel Bernstein
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA; .,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
24
|
Sayed N, Liu C, Wu JC. Translation of Human-Induced Pluripotent Stem Cells: From Clinical Trial in a Dish to Precision Medicine. J Am Coll Cardiol 2017; 67:2161-2176. [PMID: 27151349 DOI: 10.1016/j.jacc.2016.01.083] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 12/14/2022]
Abstract
The prospect of changing the plasticity of terminally differentiated cells toward pluripotency has completely altered the outlook for biomedical research. Human-induced pluripotent stem cells (iPSCs) provide a new source of therapeutic cells free from the ethical issues or immune barriers of human embryonic stem cells. iPSCs also confer considerable advantages over conventional methods of studying human diseases. Since its advent, iPSC technology has expanded with 3 major applications: disease modeling, regenerative therapy, and drug discovery. Here we discuss, in a comprehensive manner, the recent advances in iPSC technology in relation to basic, clinical, and population health.
Collapse
Affiliation(s)
- Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California.
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
25
|
Gara E, Molnár AÁ, Merkely B, Földes G. Assessing the therapeutic readiness of stem cells for cardiovascular repair. Expert Opin Biol Ther 2017; 17:911-914. [PMID: 28571479 DOI: 10.1080/14712598.2017.1338267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Edit Gara
- a Heart and Vascular Centre , Semmelweis University , Budapest , Hungary
| | | | - Béla Merkely
- a Heart and Vascular Centre , Semmelweis University , Budapest , Hungary
| | - Gábor Földes
- a Heart and Vascular Centre , Semmelweis University , Budapest , Hungary.,b Imperial Centre for Experimental and Translational Medicine , National Heart and Lung Institute, Imperial College , London , UK
| |
Collapse
|
26
|
Hypertrophy changes 3D shape of hiPSC-cardiomyocytes: Implications for cellular maturation in regenerative medicine. Cell Mol Bioeng 2016; 10:54-62. [PMID: 28163790 DOI: 10.1007/s12195-016-0462-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advances in the use of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes for heart regeneration and in vitro disease models demand a greater understanding of how these cells grow and mature in 3-dimensional space. In this study, we developed an analysis methodology of single cardiomyocytes plated on 2D surfaces to assess their 3D myofilament volume and its z-height distribution, or shape, upon hypertrophic stimulation via phenylephrine (PE) treatment or long-term culture ("aging"). Cardiomyocytes were fixed and labeled with α-actinin for confocal microscopy imaging to obtain z-stacks for 3D myofilament volume analysis. In primary neonatal rat ventricular myocytes (NRVMs), area increased 72% with PE, while volume increased 31%. In hiPSC-cardiomyocytes, area increased 70% with PE and 4-fold with aging; however, volume increased significantly only with aging by 2.3-fold. Analysis of z-height myofilament volume distribution in hiPSC-cardiomyocytes revealed a shift from a fairly uniform distribution in control cells to a basally located volume in a more flat and spread morphology with PE and even more so with aging, a shape that was akin to all NRVMs analyzed. These results suggest that 2D area is not a sufficient measure of hiPSC-cardiomyocyte growth and maturation, and that changes in 3D volume and its distribution are essential for understanding hiPSC-cardiomyocyte biology for disease modeling and regenerative medicine applications.
Collapse
|
27
|
Reid BG, Stratton MS, Bowers S, Cavasin MA, Demos-Davies KM, Susano I, McKinsey TA. Discovery of novel small molecule inhibitors of cardiac hypertrophy using high throughput, high content imaging. J Mol Cell Cardiol 2016; 97:106-13. [PMID: 27130278 PMCID: PMC5002372 DOI: 10.1016/j.yjmcc.2016.04.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/15/2016] [Accepted: 04/25/2016] [Indexed: 12/11/2022]
Abstract
Chronic cardiac hypertrophy is maladaptive and contributes to the pathogenesis of heart failure. The objective of this study was to identify small molecule inhibitors of pathological cardiomyocyte hypertrophy. High content screening was performed with primary neonatal rat ventricular myocytes (NRVMs) cultured on 96-well plates and treated with a library of 3241 distinct small molecules. Non-toxic hit compounds that blocked hypertrophy in response to phenylephrine (PE) and phorbol myristate acetate (PMA) were identified based on their ability to reduce cell size and inhibit expression of atrial natriuretic factor (ANF), which is a biomarker of pathological cardiac hypertrophy. Many of the hit compounds are existing drugs that have not previously been evaluated for benefit in the setting of cardiovascular disease. One such compound, the anti-malarial drug artesunate, blocked left ventricular hypertrophy (LVH) and improved cardiac function in adult mice subjected to transverse aortic constriction (TAC). These findings demonstrate that phenotypic screening with primary cardiomyocytes can be used to discover anti-hypertrophic lead compounds for heart failure drug discovery. Using annotated libraries of compounds with known selectivity profiles, this screening methodology also facilitates chemical biological dissection of signaling networks that control pathological growth of the heart.
Collapse
Affiliation(s)
- Brian G Reid
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, United States
| | - Matthew S Stratton
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States; Department of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Anschutz Medical Campus, United States
| | - Samantha Bowers
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, United States
| | - Maria A Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States; Department of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Anschutz Medical Campus, United States
| | - Kimberley M Demos-Davies
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States
| | - Isidro Susano
- Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO, United States
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado, Anschutz Medical Campus, United States; Department of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Anschutz Medical Campus, United States.
| |
Collapse
|
28
|
Kriston-Vizi J, Flotow H. Getting the whole picture: High content screening using three-dimensional cellular model systems and whole animal assays. Cytometry A 2016; 91:152-159. [PMID: 27403779 DOI: 10.1002/cyto.a.22907] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/09/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Phenotypic or High Content Screening (HCS) is becoming more widely used for primary screening campaigns in drug discovery. Currently the vast majority of HCS campaigns are using cell lines grown in well-established monolayer cultures (2D tissue culture). There is widespread recognition that the more biologically relevant 3D tissue culture technologies such as spheroids and organoids and even whole animal assays will eventually be run as primary HCS. Upgrading the IT infrastructure to cope with the increase in data volumes requires investments in hardware (and software) and this will be manageable. However, the main bottleneck for the effective adoption and use of 3D tissue culture and whole animal assays in HCS is anticipated to be the development of software for the analysis of 3D images. In this review we summarize the current state of the available software and how they may be applied to analyzing 3D images obtained from a HCS campaign. © 2016 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Janos Kriston-Vizi
- Bioinformatics Image Core, MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Horst Flotow
- HDC GmbH, Byk Gulden Strasse 2, Konstanz, Germany
| |
Collapse
|
29
|
Gouadon E, Moore-Morris T, Smit NW, Chatenoud L, Coronel R, Harding SE, Jourdon P, Lambert V, Rucker-Martin C, Pucéat M. Concise Review: Pluripotent Stem Cell-Derived Cardiac Cells, A Promising Cell Source for Therapy of Heart Failure: Where Do We Stand? Stem Cells 2016; 34:34-43. [PMID: 26352327 DOI: 10.1002/stem.2205] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 08/24/2015] [Indexed: 12/29/2022]
Abstract
Heart failure is still a major cause of hospitalization and mortality in developed countries. Many clinical trials have tested the use of multipotent stem cells as a cardiac regenerative medicine. The benefit for the patients of this therapeutic intervention has remained limited. Herein, we review the pluripotent stem cells as a cell source for cardiac regeneration. We more specifically address the various challenges of this cell therapy approach. We question the cell delivery systems, the immune tolerance of allogenic cells, the potential proarrhythmic effects, various drug mediated interventions to facilitate cell grafting and, finally, we describe the pathological conditions that may benefit from such an innovative approach. As members of a transatlantic consortium of excellence of basic science researchers and clinicians, we propose some guidelines to be applied to cell types and modes of delivery in order to translate pluripotent stem cell cardiac derivatives into safe and effective clinical trials.
Collapse
Affiliation(s)
- Elodie Gouadon
- INSERM UMR-S999, LabEx LERMIT, IPSIT Centre Chirurgical Marie Lanelongue, Le Plessis Robinson, Paris, France
| | | | - Nicoline W Smit
- Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, Amsterdam, The Netherlands
| | - Lucienne Chatenoud
- INSERM U1151, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ruben Coronel
- Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, Amsterdam, The Netherlands
| | | | - Philippe Jourdon
- INSERM UMR-S999, LabEx LERMIT, IPSIT Centre Chirurgical Marie Lanelongue, Le Plessis Robinson, Paris, France
| | - Virginie Lambert
- INSERM UMR-S999, LabEx LERMIT, IPSIT Centre Chirurgical Marie Lanelongue, Le Plessis Robinson, Paris, France
| | - Catherine Rucker-Martin
- INSERM UMR-S999, LabEx LERMIT, IPSIT Centre Chirurgical Marie Lanelongue, Le Plessis Robinson, Paris, France
| | - Michel Pucéat
- INSERM GMGF, a UMRS_910, Université Aix Marseille, Marseille, France
| |
Collapse
|
30
|
Dolatshad NF, Hellen N, Jabbour RJ, Harding SE, Földes G. G-protein Coupled Receptor Signaling in Pluripotent Stem Cell-derived Cardiovascular Cells: Implications for Disease Modeling. Front Cell Dev Biol 2015; 3:76. [PMID: 26697426 PMCID: PMC4673467 DOI: 10.3389/fcell.2015.00076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 11/09/2015] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell derivatives show promise as an in vitro platform to study a range of human cardiovascular diseases. A better understanding of the biology of stem cells and their cardiovascular derivatives will help to understand the strengths and limitations of this new model system. G-protein coupled receptors (GPCRs) are key regulators of stem cell maintenance and differentiation and have an important role in cardiovascular cell signaling. In this review, we will therefore describe the state of knowledge concerning the regulatory role of GPCRs in both the generation and function of pluripotent stem cell derived-cardiomyocytes, -endothelial, and -vascular smooth muscle cells. We will consider how far the in vitro disease models recapitulate authentic GPCR signaling and provide a useful basis for discovery of disease mechanisms or design of therapeutic strategies.
Collapse
Affiliation(s)
- Nazanin F Dolatshad
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Nicola Hellen
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Richard J Jabbour
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Sian E Harding
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK
| | - Gabor Földes
- Myocardial Function, National Heart and Lung Institute, Imperial College London London, UK ; The Heart and Vascular Center of Semmelweis University, Semmelweis University Budapest, Hungary
| |
Collapse
|
31
|
Cui H, Schlesinger J, Schoenhals S, Tönjes M, Dunkel I, Meierhofer D, Cano E, Schulz K, Berger MF, Haack T, Abdelilah-Seyfried S, Bulyk ML, Sauer S, Sperling SR. Phosphorylation of the chromatin remodeling factor DPF3a induces cardiac hypertrophy through releasing HEY repressors from DNA. Nucleic Acids Res 2015; 44:2538-53. [PMID: 26582913 PMCID: PMC4824069 DOI: 10.1093/nar/gkv1244] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/01/2015] [Indexed: 01/09/2023] Open
Abstract
DPF3 (BAF45c) is a member of the BAF chromatin remodeling complex. Two isoforms have been described, namely DPF3a and DPF3b. The latter binds to acetylated and methylated lysine residues of histones. Here, we elaborate on the role of DPF3a and describe a novel pathway of cardiac gene transcription leading to pathological cardiac hypertrophy. Upon hypertrophic stimuli, casein kinase 2 phosphorylates DPF3a at serine 348. This initiates the interaction of DPF3a with the transcriptional repressors HEY, followed by the release of HEY from the DNA. Moreover, BRG1 is bound by DPF3a, and is thus recruited to HEY genomic targets upon interaction of the two components. Consequently, the transcription of downstream targets such as NPPA and GATA4 is initiated and pathological cardiac hypertrophy is established. In human, DPF3a is significantly up-regulated in hypertrophic hearts of patients with hypertrophic cardiomyopathy or aortic stenosis. Taken together, we show that activation of DPF3a upon hypertrophic stimuli switches cardiac fetal gene expression from being silenced by HEY to being activated by BRG1. Thus, we present a novel pathway for pathological cardiac hypertrophy, whose inhibition is a long-term therapeutic goal for the treatment of the course of heart failure.
Collapse
Affiliation(s)
- Huanhuan Cui
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Jenny Schlesinger
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sophia Schoenhals
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Martje Tönjes
- Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Ilona Dunkel
- Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Elena Cano
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Kerstin Schulz
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Michael F Berger
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA Committee on Higher Degrees in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Timm Haack
- Hannover Medical School, Institute of Molecular Biology, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| | - Salim Abdelilah-Seyfried
- Hannover Medical School, Institute of Molecular Biology, Carl-Neuberg Str. 1, D-30625 Hannover, Germany Potsdam University, Institute of Biochemistry and Biology, Department of Animal Physiology, Karl-Liebknecht Str. 24-25, 14476 Potsdam-Golm, Germany
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA Committee on Higher Degrees in Biophysics, Harvard University, Cambridge, MA 02138, USA Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sascha Sauer
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany CU Systems Medicine, University of Würzburg, 97080 Würzburg, Germany
| | - Silke R Sperling
- Department of Cardiovascular Genetics, Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany Group of Cardiovascular Genetics, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
32
|
Bedada FB, Wheelwright M, Metzger JM. Maturation status of sarcomere structure and function in human iPSC-derived cardiac myocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1829-38. [PMID: 26578113 DOI: 10.1016/j.bbamcr.2015.11.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/05/2015] [Accepted: 11/09/2015] [Indexed: 12/15/2022]
Abstract
Human heart failure due to myocardial infarction is a major health concern. The paucity of organs for transplantation limits curative approaches for the diseased and failing adult heart. Human induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs) have the potential to provide a long-term, viable, regenerative-medicine alternative. Significant progress has been made with regard to efficient cardiac myocyte generation from hiPSCs. However, directing hiPSC-CMs to acquire the physiological structure, gene expression profile and function akin to mature cardiac tissue remains a major obstacle. Thus, hiPSC-CMs have several hurdles to overcome before they find their way into translational medicine. In this review, we address the progress that has been made, the void in knowledge and the challenges that remain. 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)
- Fikru B Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA
| | - Matthew Wheelwright
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA.
| |
Collapse
|
33
|
Denning C, Borgdorff V, Crutchley J, Firth KSA, George V, Kalra S, Kondrashov A, Hoang MD, Mosqueira D, Patel A, Prodanov L, Rajamohan D, Skarnes WC, Smith JGW, Young LE. Cardiomyocytes from human pluripotent stem cells: From laboratory curiosity to industrial biomedical platform. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1728-48. [PMID: 26524115 PMCID: PMC5221745 DOI: 10.1016/j.bbamcr.2015.10.014] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/12/2015] [Accepted: 10/20/2015] [Indexed: 12/14/2022]
Abstract
Cardiomyocytes from human pluripotent stem cells (hPSCs-CMs) could revolutionise biomedicine. Global burden of heart failure will soon reach USD $90bn, while unexpected cardiotoxicity underlies 28% of drug withdrawals. Advances in hPSC isolation, Cas9/CRISPR genome engineering and hPSC-CM differentiation have improved patient care, progressed drugs to clinic and opened a new era in safety pharmacology. Nevertheless, predictive cardiotoxicity using hPSC-CMs contrasts from failure to almost total success. Since this likely relates to cell immaturity, efforts are underway to use biochemical and biophysical cues to improve many of the ~30 structural and functional properties of hPSC-CMs towards those seen in adult CMs. Other developments needed for widespread hPSC-CM utility include subtype specification, cost reduction of large scale differentiation and elimination of the phenotyping bottleneck. This review will consider these factors in the evolution of hPSC-CM technologies, as well as their integration into high content industrial platforms that assess structure, mitochondrial function, electrophysiology, calcium transients and contractility. 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)
- Chris Denning
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom.
| | - Viola Borgdorff
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - James Crutchley
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Karl S A Firth
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Vinoj George
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Spandan Kalra
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Alexander Kondrashov
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Minh Duc Hoang
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Diogo Mosqueira
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Asha Patel
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Ljupcho Prodanov
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Divya Rajamohan
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - William C Skarnes
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - James G W Smith
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Lorraine E Young
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
34
|
Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR, van de Pol V, Dambrot C, Devalla HD, Davis RP, Mastroberardino PG, Atsma DE, Passier R, Mummery CL. Contractile Defect Caused by Mutation in MYBPC3 Revealed under Conditions Optimized for Human PSC-Cardiomyocyte Function. Cell Rep 2015; 13:733-745. [PMID: 26489474 PMCID: PMC4644234 DOI: 10.1016/j.celrep.2015.09.025] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/31/2015] [Accepted: 09/05/2015] [Indexed: 12/23/2022] Open
Abstract
Maximizing baseline function of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is essential for their effective application in models of cardiac toxicity and disease. Here, we aimed to identify factors that would promote an adequate level of function to permit robust single-cell contractility measurements in a human induced pluripotent stem cell (hiPSC) model of hypertrophic cardiomyopathy (HCM). A simple screen revealed the collaborative effects of thyroid hormone, IGF-1 and the glucocorticoid analog dexamethasone on the electrophysiology, bioenergetics, and contractile force generation of hPSC-CMs. In this optimized condition, hiPSC-CMs with mutations in MYBPC3, a gene encoding myosin-binding protein C, which, when mutated, causes HCM, showed significantly lower contractile force generation than controls. This was recapitulated by direct knockdown of MYBPC3 in control hPSC-CMs, supporting a mechanism of haploinsufficiency. Modeling this disease in vitro using human cells is an important step toward identifying therapeutic interventions for HCM.
Collapse
Affiliation(s)
- Matthew J Birket
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Marcelo C Ribeiro
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Georgios Kosmidis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Dorien Ward
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Ana Rita Leitoguinho
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Vera van de Pol
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Cheryl Dambrot
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Harsha D Devalla
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | | | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
35
|
Kimbrel EA, Lanza R. Current status of pluripotent stem cells: moving the first therapies to the clinic. Nat Rev Drug Discov 2015; 14:681-92. [PMID: 26391880 DOI: 10.1038/nrd4738] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pluripotent stem cells (PSCs) hold great promise for drug discovery and regenerative medicine owing to their ability to differentiate into any cell type in the body. After more than three decades of research, including delays due to the potential tumorigenicity of PSCs and inefficiencies in differentiation methods, the field is at a turning point, with a number of clinical trials across the globe now testing PSC-derived products in humans. Ocular diseases dominate these first-in-man trials, and Phase l/ll results are showing promising safety data as well as possible efficacy. In addition, the advent of induced PSC (iPSC) technology is enabling the development of a wide range of cell-based disease models from genetically predisposed patients, thereby facilitating drug discovery. In this Review, we discuss the recent progress and remaining challenges for the use of PSCs in regenerative medicine and drug development.
Collapse
Affiliation(s)
- Erin A Kimbrel
- Ocata Therapeutics, 33 Locke Drive, Marlborough, Massachusetts 01752, USA
| | - Robert Lanza
- Ocata Therapeutics, 33 Locke Drive, Marlborough, Massachusetts 01752, USA
| |
Collapse
|
36
|
Wang Y, Li ZC, Zhang P, Poon E, Kong CW, Boheler KR, Huang Y, Li RA, Yao X. Nitric Oxide-cGMP-PKG Pathway Acts on Orai1 to Inhibit the Hypertrophy of Human Embryonic Stem Cell-Derived Cardiomyocytes. Stem Cells 2015; 33:2973-84. [PMID: 26269433 DOI: 10.1002/stem.2118] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/15/2015] [Indexed: 11/08/2022]
Abstract
Cardiac hypertrophy is an abnormal enlargement of heart muscle. It frequently results in congestive heart failure, which is a leading cause of human death. Previous studies demonstrated that the nitric oxide (NO), cyclic GMP (cGMP), and protein kinase G (PKG) signaling pathway can inhibit cardiac hypertrophy and thus improve cardiac function. However, the underlying mechanisms are not fully understood. Here, based on the human embryonic stem cell-derived cardiomyocyte (hESC-CM) model system, we showed that Orai1, the pore-forming subunit of store-operated Ca(2+) entry (SOCE), is the downstream effector of PKG. Treatment of hESC-CMs with an α-adrenoceptor agonist phenylephrine (PE) caused a marked hypertrophy, which was accompanied by an upregulation of Orai1. Moreover, suppression of Orai1 expression/activity using Orai1-siRNAs or a dominant-negative construct Orai1(G98A) inhibited the hypertrophy, suggesting that Orai1-mediated SOCE is indispensable for the PE-induced hypertrophy of hESC-CMs. In addition, the hypertrophy was inhibited by NO and cGMP via activating PKG. Importantly, substitution of Ala for Ser(34) in Orai1 abolished the antihypertrophic effects of NO, cGMP, and PKG. Furthermore, PKG could directly phosphorylate Orai1 at Ser(34) and thus prevent Orai1-mediated SOCE. Together, we conclude that NO, cGMP, and PKG inhibit the hypertrophy of hESC-CMs via PKG-mediated phosphorylation on Orai1-Ser-34. These results provide novel mechanistic insights into the action of cGMP-PKG-related antihypertrophic agents, such as NO donors and sildenafil.
Collapse
Affiliation(s)
- Y Wang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Z C Li
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - P Zhang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - E Poon
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China.,The Department of Physiology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - C W Kong
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China
| | - K R Boheler
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China.,The Department of Physiology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Y Huang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - R A Li
- Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong, People's Republic of China
| | - X Yao
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| |
Collapse
|
37
|
Veerman CC, Kosmidis G, Mummery CL, Casini S, Verkerk AO, Bellin M. Immaturity of Human Stem-Cell-Derived Cardiomyocytes in Culture: Fatal Flaw or Soluble Problem? Stem Cells Dev 2015; 24:1035-52. [DOI: 10.1089/scd.2014.0533] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Christiaan C. Veerman
- Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Georgios Kosmidis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Simona Casini
- Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arie O. Verkerk
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
38
|
Simonson OE, Domogatskaya A, Volchkov P, Rodin S. The safety of human pluripotent stem cells in clinical treatment. Ann Med 2015; 47:370-80. [PMID: 26140342 DOI: 10.3109/07853890.2015.1051579] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) have practically unlimited proliferation potential and a capability to differentiate into any cell type in the human body. Since the first derivation in 1998, they have been an attractive source of cells for regenerative medicine. Numerous ethical, technological, and regulatory complications have been hampering hPSC use in clinical applications. Human embryonic stem cells (ESCs), parthenogenetic human ESCs, human nuclear transfer ESCs, and induced pluripotent stem cells are four types of hPSCs that are different in many clinically relevant features such as propensity to epigenetic abnormalities, generation methods, and ability for development of autologous cell lines. Propensity to genetic mutations and tumorigenicity are common features of all pluripotent cells that complicate hPSC-based therapies. Several recent advances in methods of derivation, culturing, and monitoring of hPSCs have addressed many ethical concerns and technological challenges in development of clinical-grade hPSC lines. Generation of banks of such lines may be useful to minimize immune rejection of hPSC-derived allografts. In this review, we discuss different sources of hPSCs available at the moment, various safety risks associated with them, and possible solutions for successful use of hPSCs in the clinic. We also discuss ongoing clinical trials of hPSC-based treatments.
Collapse
Affiliation(s)
- Oscar E Simonson
- a Division of Cardiothoracic Surgery and Anesthesiology, Department of Molecular Medicine and Surgery , Karolinska Institutet, Karolinska University Hospital , 171 77 Stockholm , Sweden
| | | | | | | |
Collapse
|