51
|
Crasto S, Di Pasquale E. Induced Pluripotent Stem Cells to Study Mechanisms of Laminopathies: Focus on Epigenetics. Front Cell Dev Biol 2018; 6:172. [PMID: 30619852 PMCID: PMC6306496 DOI: 10.3389/fcell.2018.00172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022] Open
Abstract
Laminopathies are a group of rare degenerative disorders that manifest with a wide spectrum of clinical phenotypes, including both systemic multi-organ disorders, such as the Hutchinson-Gilford Progeria Syndrome (HGPS), and tissue-restricted diseases, such as Emery-Dreifuss muscular dystrophy, dilated cardiomyopathy and lipodystrophies, often overlapping. Despite their clinical heterogeneity, which remains an open question, laminopathies are commonly caused by mutations in the LMNA gene, encoding the nuclear proteins Lamin A and C. These two proteins are main components of the nuclear lamina and are involved in several biological processes. Besides the well-known structural function in the nucleus, their role in regulating chromatin organization and transcription has emerged in the last decade, supporting the hypothesis that the disruption of this layer of regulation may be mechanism underlying the disease. Indeed, recent studies that show various epigenetic defects in cells carrying LMNA mutations, such as loss of heterochromatin, changes in gene expression and chromatin remodeling, strongly support this view. However, those findings are restricted to few cell types in humans, mainly because of the limited accessibility of primary cells and the difficulties to culture them ex-vivo. On the other hand, animal models might fail to recapitulate phenotypic hallmarks of the disease as of humans. To fill this gap, models based on induced pluripotent stem cell (iPSCs) technology have been recently generated that allowed investigations on diverse cells types, such as mesenchymal stem cells (MSCs), vascular and smooth muscle cells and cardiomyocytes, and provided a platform for investigating mechanisms underlying the pathogenesis of laminopathies in a cell-type specific human context. Nevertheless, studies on iPSC-based models of laminopathy have expanded only in the last few years and, with the advancement of reprogramming and differentiation protocols, their number is expecting to further increase over time. This review will give an overview of models developed thus far, with a focus on the novel insights on epigenetic mechanisms underlying the disease in different human cellular contexts. Perspectives and future directions of the field will be also given, highlighting the potential of those models for preclinical studies for identifying molecular targets and their translational impact on patients' cure.
Collapse
Affiliation(s)
- Silvia Crasto
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Milan, Milan, Italy.,Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elisa Di Pasquale
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Milan, Milan, Italy.,Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
52
|
van Mil A, Balk GM, Neef K, Buikema JW, Asselbergs FW, Wu SM, Doevendans PA, Sluijter JPG. Modelling inherited cardiac disease using human induced pluripotent stem cell-derived cardiomyocytes: progress, pitfalls, and potential. Cardiovasc Res 2018; 114:1828-1842. [PMID: 30169602 PMCID: PMC6887927 DOI: 10.1093/cvr/cvy208] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/06/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
In the past few years, the use of specific cell types derived from induced pluripotent stem cells (iPSCs) has developed into a powerful approach to investigate the cellular pathophysiology of numerous diseases. Despite advances in therapy, heart disease continues to be one of the leading causes of death in the developed world. A major difficulty in unravelling the underlying cellular processes of heart disease is the extremely limited availability of viable human cardiac cells reflecting the pathological phenotype of the disease at various stages. Thus, the development of methods for directed differentiation of iPSCs to cardiomyocytes (iPSC-CMs) has provided an intriguing option for the generation of patient-specific cardiac cells. In this review, a comprehensive overview of the currently published iPSC-CM models for hereditary heart disease is compiled and analysed. Besides the major findings of individual studies, detailed methodological information on iPSC generation, iPSC-CM differentiation, characterization, and maturation is included. Both, current advances in the field and challenges yet to overcome emphasize the potential of using patient-derived cell models to mimic genetic cardiac diseases.
Collapse
Affiliation(s)
- Alain van Mil
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Geerthe Margriet Balk
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Klaus Neef
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jan Willem Buikema
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Folkert W Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science, University College London, London, UK
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, Utrecht, the Netherlands
- Farr Institute of Health Informatics Research and Institute of Health Informatics, University College London, London, UK
| | - Sean M Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pieter A Doevendans
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
53
|
Steele-Stallard HB, Pinton L, Sarcar S, Ozdemir T, Maffioletti SM, Zammit PS, Tedesco FS. Modeling Skeletal Muscle Laminopathies Using Human Induced Pluripotent Stem Cells Carrying Pathogenic LMNA Mutations. Front Physiol 2018; 9:1332. [PMID: 30405424 PMCID: PMC6201196 DOI: 10.3389/fphys.2018.01332] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/04/2018] [Indexed: 01/03/2023] Open
Abstract
Laminopathies are a clinically heterogeneous group of disorders caused by mutations in LMNA. The main proteins encoded by LMNA are Lamin A and C, which together with Lamin B1 and B2, form the nuclear lamina: a mesh-like structure located underneath the inner nuclear membrane. Laminopathies show striking tissue specificity, with subtypes affecting striated muscle, peripheral nerve, and adipose tissue, while others cause multisystem disease with accelerated aging. Although several pathogenic mechanisms have been proposed, the exact pathophysiology of laminopathies remains unclear, compounded by the rarity of these disorders and lack of easily accessible cell types to study. To overcome this limitation, we used induced pluripotent stem cells (iPSCs) from patients with skeletal muscle laminopathies such as LMNA-related congenital muscular dystrophy and limb-girdle muscular dystrophy 1B, to model disease phenotypes in vitro. iPSCs can be derived from readily accessible cell types, have unlimited proliferation potential and can be differentiated into cell types that would otherwise be difficult and invasive to obtain. iPSC lines from three skeletal muscle laminopathy patients were differentiated into inducible myogenic cells and myotubes. Disease-associated phenotypes were observed in these cells, including abnormal nuclear shape and mislocalization of nuclear lamina proteins. Nuclear abnormalities were less pronounced in monolayer cultures of terminally differentiated skeletal myotubes than in proliferating myogenic cells. Notably, skeletal myogenic differentiation of LMNA-mutant iPSCs in artificial muscle constructs improved detection of myonuclear abnormalities compared to conventional monolayer cultures across multiple pathogenic genotypes, providing a high-fidelity modeling platform for skeletal muscle laminopathies. Our results lay the foundation for future iPSC-based therapy development and screening platforms for skeletal muscle laminopathies.
Collapse
Affiliation(s)
- Heather B Steele-Stallard
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.,Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Luca Pinton
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.,Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Shilpita Sarcar
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Tanel Ozdemir
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.,Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Sara M Maffioletti
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.,The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
54
|
Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 2018; 471:755-768. [PMID: 30324321 PMCID: PMC6475632 DOI: 10.1007/s00424-018-2214-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) can be differentiated to cardiomyocytes at high efficiency and are increasingly used to study cardiac disease in a human context. This review evaluated 38 studies on hypertrophic (HCM) and dilated cardiomyopathy (DCM) of different genetic causes asking to which extent published data allow the definition of an in vitro HCM/DCM hiPSC-CM phenotype. The data are put in context with the prevailing hypotheses on HCM/DCM dysfunction and pathophysiology. Relatively consistent findings in HCM not reported in DCM were larger cell size (156 ± 85%, n = 15), more nuclear localization of nuclear factor of activated T cells (NFAT; 175 ± 65%, n = 3), and higher β-myosin heavy chain gene expression levels (500 ± 547%, n = 8) than respective controls. Conversely, DCM lines showed consistently less force development than controls (47 ± 23%, n = 9), while HCM forces scattered without clear trend. Both HCM and DCM lines often showed sarcomere disorganization, higher NPPA/NPPB expression levels, and arrhythmic beating behaviour. The data have to be taken with the caveat that reporting frequencies of the various parameters (e.g. cell size, NFAT expression) differ widely between HCM and DCM lines, in which data scatter is large and that only 9/38 studies used isogenic controls. Taken together, the current data provide interesting suggestions for disease-specific phenotypes in HCM/DCM hiPSC-CM but indicate that the field is still in its early days. Systematic, quantitative comparisons and robust, high content assays are warranted to advance the field.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| |
Collapse
|
55
|
Ng KM, Lau YM, Dhandhania V, Cai ZJ, Lee YK, Lai WH, Tse HF, Siu CW. Empagliflozin Ammeliorates High Glucose Induced-Cardiac Dysfuntion in Human iPSC-Derived Cardiomyocytes. Sci Rep 2018; 8:14872. [PMID: 30291295 PMCID: PMC6173708 DOI: 10.1038/s41598-018-33293-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
Empagliflozin, a sodium-glucose co-transporter (SGLT) inhibitor, reduces heart failure and sudden cardiac death but the underlying mechanisms remain elusive. In cardiomyocytes, SGLT1 and SGLT2 expression is upregulated in diabetes mellitus, heart failure, and myocardial infarction. We hypothesise that empagliflozin exerts direct effects on cardiomyocytes that attenuate diabetic cardiomyopathy. To test this hypothesis, cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) were used to test the potential effects of empagliflozin on neutralization of cardiac dysfunction induced by diabetic-like cultures. Our results indicated that insulin-free high glucose culture significantly increased the size of and NPPB, SGLT1 and SGLT2 expression of hiPSC-derived cardiomyocytes. In addition, high glucose-treated hiPSC-derived cardiomyocytes exhibited reduced contractility regardless of the increased calcium transient capacity. Interestingly, application of empagliflozin before or after high glucose treatment effectively reduced the high glucose-induced cardiac abnormalities. Since application of empagliflozin did not significantly alter viability or glycolytic capacity of the hiPSC-derived cardiomyocytes, it is plausible that empagliflozin exerts its effects via the down-regulation of SGLT1, SGLT2 and GLUT1 expression. These observations provide supportive evidence that may help explain its unexpected benefit observed in the EMPA-REG trial.
Collapse
Affiliation(s)
- Kwong-Man Ng
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Vidhu Dhandhania
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Zhu-Jun Cai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Yee-Ki Lee
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China.
| |
Collapse
|
56
|
Durbin MD, Cadar AG, Chun YW, Hong CC. Investigating pediatric disorders with induced pluripotent stem cells. Pediatr Res 2018; 84:499-508. [PMID: 30065271 PMCID: PMC6265074 DOI: 10.1038/s41390-018-0064-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
The study of disease pathophysiology has long relied on model systems, including animal models and cultured cells. In 2006, Shinya Yamanaka achieved a breakthrough by reprogramming somatic cells into induced pluripotent stem cells (iPSCs). This revolutionary discovery provided new opportunities for disease modeling and therapeutic intervention. With established protocols, investigators can generate iPSC lines from patient blood, urine, and tissue samples. These iPSCs retain ability to differentiate into every human cell type. Advances in differentiation and organogenesis move cellular in vitro modeling to a multicellular model capable of recapitulating physiology and disease. Here, we discuss limitations of traditional animal and tissue culture models, as well as the application of iPSC models. We highlight various techniques, including reprogramming strategies, directed differentiation, tissue engineering, organoid developments, and genome editing. We extensively summarize current established iPSC disease models that utilize these techniques. Confluence of these technologies will advance our understanding of pediatric diseases and help usher in new personalized therapies for patients.
Collapse
Affiliation(s)
- Matthew D. Durbin
- Department of Pediatrics – Division of Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Adrian G. Cadar
- Departments of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Young W. Chun
- Department of Medicine - Cardiovascular Medicine Division University of Maryland School of Medicine, Baltimore, MD 21201
| | - Charles C. Hong
- Department of Medicine - Cardiovascular Medicine Division University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
57
|
Affiliation(s)
- Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
58
|
Linker of nucleoskeleton and cytoskeleton complex proteins in cardiomyopathy. Biophys Rev 2018; 10:1033-1051. [PMID: 29869195 PMCID: PMC6082319 DOI: 10.1007/s12551-018-0431-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/24/2018] [Indexed: 12/21/2022] Open
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex couples the nuclear lamina to the cytoskeleton. The LINC complex and its associated proteins play diverse roles in cells, ranging from genome organization, nuclear morphology, gene expression, to mechanical stability. The importance of a functional LINC complex is highlighted by the large number of mutations in genes encoding LINC complex proteins that lead to skeletal and cardiac myopathies. In this review, the structure, function, and interactions between components of the LINC complex will be described. Mutations that are known to cause cardiomyopathy in patients will be discussed alongside their respective mouse models. Furthermore, future challenges for the field and emerging technologies to investigate LINC complex function will be discussed.
Collapse
|
59
|
Ye L, Ni X, Zhao ZA, Lei W, Hu S. The Application of Induced Pluripotent Stem Cells in Cardiac Disease Modeling and Drug Testing. J Cardiovasc Transl Res 2018; 11:366-374. [PMID: 29845439 DOI: 10.1007/s12265-018-9811-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/06/2018] [Indexed: 12/18/2022]
Abstract
In recent decades, cardiovascular diseases have become the greatest health threat to human beings, and thus it is particularly important to explore the subtle underlying pathogenesis of cardiovascular diseases. Although many molecular pathways have been explored to be essential in the development of cardiovascular diseases, their clinical significances are still uncertain. With the emergence of induced pluripotent stem cells (iPSCs), a unique platform for cardiovascular diseases has been established to model cardiovascular diseases on specific genetic background in vitro. This review summarizes current progresses of iPSCs in cardiovascular disease modeling and drug testing. This review highlighted iPSC-based cardiovascular disease modeling and drug testing. The technical advances in iPSC-based researches and various clinically relevant applications are discussed. With further intensive research, iPSC technology will shape the future of clinical translational research in cardiovascular diseases.
Collapse
Affiliation(s)
- Lingqun Ye
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China
| | - Xuan Ni
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China
| | - Zhen-Ao Zhao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, 215000, Suzhou, China
| | - Wei Lei
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China.,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, 215000, Suzhou, China
| | - Shijun Hu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China. .,Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, 215000, Suzhou, China.
| |
Collapse
|
60
|
Brandão KO, Tabel VA, Atsma DE, Mummery CL, Davis RP. Human pluripotent stem cell models of cardiac disease: from mechanisms to therapies. Dis Model Mech 2018; 10:1039-1059. [PMID: 28883014 PMCID: PMC5611968 DOI: 10.1242/dmm.030320] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is now a decade since human induced pluripotent stem cells (hiPSCs) were first described. The reprogramming of adult somatic cells to a pluripotent state has become a robust technology that has revolutionised our ability to study human diseases. Crucially, these cells capture all the genetic aspects of the patient from which they were derived. Combined with advances in generating the different cell types present in the human heart, this has opened up new avenues to study cardiac disease in humans and investigate novel therapeutic approaches to treat these pathologies. Here, we provide an overview of the current state of the field regarding the generation of cardiomyocytes from human pluripotent stem cells and methods to assess them functionally, an essential requirement when investigating disease and therapeutic outcomes. We critically evaluate whether treatments suggested by these in vitro models could be translated to clinical practice. Finally, we consider current shortcomings of these models and propose methods by which they could be further improved.
Collapse
Affiliation(s)
- Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Viola A Tabel
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
61
|
Kalra S, Montanaro F, Denning C. Can Human Pluripotent Stem Cell-Derived Cardiomyocytes Advance Understanding of Muscular Dystrophies? J Neuromuscul Dis 2018; 3:309-332. [PMID: 27854224 PMCID: PMC5123622 DOI: 10.3233/jnd-150133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Muscular dystrophies (MDs) are clinically and molecularly a highly heterogeneous group of single-gene disorders that primarily affect striated muscles. Cardiac disease is present in several MDs where it is an important contributor to morbidity and mortality. Careful monitoring of cardiac issues is necessary but current management of cardiac involvement does not effectively protect from disease progression and cardiac failure. There is a critical need to gain new knowledge on the diverse molecular underpinnings of cardiac disease in MDs in order to guide cardiac treatment development and assist in reaching a clearer consensus on cardiac disease management in the clinic. Animal models are available for the majority of MDs and have been invaluable tools in probing disease mechanisms and in pre-clinical screens. However, there are recognized genetic, physiological, and structural differences between human and animal hearts that impact disease progression, manifestation, and response to pharmacological interventions. Therefore, there is a need to develop parallel human systems to model cardiac disease in MDs. This review discusses the current status of cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSC) to model cardiac disease, with a focus on Duchenne muscular dystrophy (DMD) and myotonic dystrophy (DM1). We seek to provide a balanced view of opportunities and limitations offered by this system in elucidating disease mechanisms pertinent to human cardiac physiology and as a platform for treatment development or refinement.
Collapse
Affiliation(s)
- Spandan Kalra
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, UK
| | - Federica Montanaro
- Dubowitz Neuromuscular Centre, Department of Molecular Neurosciences, University College London - Institute of Child Health, London, UK
| | - Chris Denning
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, UK
| |
Collapse
|
62
|
Musunuru K, Sheikh F, Gupta RM, Houser SR, Maher KO, Milan DJ, Terzic A, Wu JC. Induced Pluripotent Stem Cells for Cardiovascular Disease Modeling and Precision Medicine: A Scientific Statement From the American Heart Association. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e000043. [PMID: 29874173 PMCID: PMC6708586 DOI: 10.1161/hcg.0000000000000043] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Induced pluripotent stem cells (iPSCs) offer an unprece-dented opportunity to study human physiology and disease at the cellular level. They also have the potential to be leveraged in the practice of precision medicine, for example, personalized drug testing. This statement comprehensively describes the provenance of iPSC lines, their use for cardiovascular disease modeling, their use for precision medicine, and strategies through which to promote their wider use for biomedical applications. Human iPSCs exhibit properties that render them uniquely qualified as model systems for studying human diseases: they are of human origin, which means they carry human genomes; they are pluripotent, which means that in principle, they can be differentiated into any of the human body's somatic cell types; and they are stem cells, which means they can be expanded from a single cell into millions or even billions of cell progeny. iPSCs offer the opportunity to study cells that are genetically matched to individual patients, and genome-editing tools allow introduction or correction of genetic variants. Initial progress has been made in using iPSCs to better understand cardiomyopathies, rhythm disorders, valvular and vascular disorders, and metabolic risk factors for ischemic heart disease. This promising work is still in its infancy. Similarly, iPSCs are only just starting to be used to identify the optimal medications to be used in patients from whom the cells were derived. This statement is intended to (1) summarize the state of the science with respect to the use of iPSCs for modeling of cardiovascular traits and disorders and for therapeutic screening; (2) identify opportunities and challenges in the use of iPSCs for disease modeling and precision medicine; and (3) outline strategies that will facilitate the use of iPSCs for biomedical applications. This statement is not intended to address the use of stem cells as regenerative therapy, such as transplantation into the body to treat ischemic heart disease or heart failure.
Collapse
|
63
|
Affiliation(s)
- Gianfranco Sinagra
- From the Cardiovascular Department, Cardiomyopathy Centre, University of Trieste, Italy
| | - Matteo Dal Ferro
- From the Cardiovascular Department, Cardiomyopathy Centre, University of Trieste, Italy
| | - Marco Merlo
- From the Cardiovascular Department, Cardiomyopathy Centre, University of Trieste, Italy
| |
Collapse
|
64
|
Giacomelli E, Mummery CL, Bellin M. Human heart disease: lessons from human pluripotent stem cell-derived cardiomyocytes. Cell Mol Life Sci 2017; 74:3711-3739. [PMID: 28573431 PMCID: PMC5597692 DOI: 10.1007/s00018-017-2546-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/09/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023]
Abstract
Technical advances in generating and phenotyping cardiomyocytes from human pluripotent stem cells (hPSC-CMs) are now driving their wider acceptance as in vitro models to understand human heart disease and discover therapeutic targets that may lead to new compounds for clinical use. Current literature clearly shows that hPSC-CMs recapitulate many molecular, cellular, and functional aspects of human heart pathophysiology and their responses to cardioactive drugs. Here, we provide a comprehensive overview of hPSC-CMs models that have been described to date and highlight their most recent and remarkable contributions to research on cardiovascular diseases and disorders with cardiac traits. We conclude discussing immediate challenges, limitations, and emerging solutions.
Collapse
Affiliation(s)
- E Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - C L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Building Zuidhorst, 7500 AE, Enschede, The Netherlands
| | - M Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands.
| |
Collapse
|
65
|
Liu N, Zheng M, Li S, Bai H, Liu Z, Hou CH, Zhang S, Pu J. Genetic Mechanisms Contribute to the Development of Heart Failure in Patients with Atrioventricular Block and Right Ventricular Apical Pacing. Sci Rep 2017; 7:10676. [PMID: 28878402 PMCID: PMC5587648 DOI: 10.1038/s41598-017-11211-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Right ventricular apical (RVA) pacing can lead to progressive left ventricular dysfunction and heart failure (HF), even in patients with normal cardiac structure and function. Our study conducted candidate gene screening and lentivirus transfected neonatal rat cardiomyocytes (NRCMs) to explore the genetic and pathogenic mechanisms of RVA pacing induced cardiomyopathy in third degree atrioventricular block (III AVB) patients. We followed 887 III AVB patients with baseline normal cardiac function and RVA pacing. After a median follow-up of 2.5 years, 10 patients (four males, mean age 47.6 ± 10.0 years) were diagnosed with RVA pacing induced HF with left ventricular ejection fraction (LVEF) reducing dramatically to 37.8 ± 7.1% (P < 0.05). Candidate genes sequencing found cardiomyopathy associated genetic variations in all ten HF patients and six SCN5A variations in 6 of 20 control patients. Transfected NRCMs of Lamin A/C mutations (R216C and L379F) disrupted Lamin A/C location on nucleus membrane and finally resulted in increased apoptotic rate after serum starvation. In conclusion, cardiomyopathy associated genetic variations play an essential role in occurrence of newly onset HF in the III AVB patients with RVA pacing. RVA pacing, serving as extra stimulator, might accelerate the deterioration of cardiac structure and function.
Collapse
Affiliation(s)
- Nana Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Min Zheng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
- Department of cardiovascular diseases, Dongfang Hospital Affiliated to Tongji University, Shanghai, 200120, People's Republic of China
| | - Shijie Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Hui Bai
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Zhouying Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Cui Hong Hou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Shu Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Jielin Pu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China.
- Department of cardiovascular diseases, Dongfang Hospital Affiliated to Tongji University, Shanghai, 200120, People's Republic of China.
| |
Collapse
|
66
|
Çağlayan AO, Sezer RG, Kaymakçalan H, Ulgen E, Yavuz T, Baranoski JF, Bozaykut A, Harmanci AS, Yalcin Y, Youngblood MW, Yasuno K, Bilgüvar K, Gunel M. ALPK3 gene mutation in a patient with congenital cardiomyopathy and dysmorphic features. Cold Spring Harb Mol Case Stud 2017. [PMID: 28630369 PMCID: PMC5593152 DOI: 10.1101/mcs.a001859] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Primary cardiomyopathy is one of the most common inherited cardiac diseases and harbors significant phenotypic and genetic heterogeneity. Because of this, genetic testing has become standard in treatment of this disease group. Indeed, in recent years, next-generation DNA sequencing has found broad applications in medicine, both as a routine diagnostic tool for genetic disorders and as a high-throughput discovery tool for identifying novel disease-causing genes. We describe a male infant with primary dilated cardiomyopathy who was diagnosed using intrauterine echocardiography and found to progress to hypertrophic cardiomyopathy after birth. This proband was born to a nonconsanguineous family with a past history of a male fetus that died because of cardiac abnormalities at 30 wk of gestation. Using whole-exome sequencing, a novel homozygous frameshift mutation (c.2018delC; p.Gln675SerfsX30) in ALPK3 was identified and confirmed with Sanger sequencing. Heterozygous family members were normal with echocardiographic examination. To date, only two studies have reported homozygous pathogenic variants of ALPK3, with a total of seven affected individuals with cardiomyopathy from four unrelated consanguineous families. We include a discussion of the patient's phenotypic features and a review of relevant literature findings.
Collapse
Affiliation(s)
- Ahmet Okay Çağlayan
- Department of Medical Genetics, School of Medicine, Istanbul Bilim University, Istanbul 34394, Turkey.,Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Rabia Gonul Sezer
- Department of Pediatrics, University of Health Sciences, Zeynep Kamil Maternity and Childrens' Diseases Training and Research Hospital, Istanbul 34668, Turkey
| | - Hande Kaymakçalan
- Department of Pediatrics, School of Medicine, Istanbul Bilim University, Istanbul 34394, Turkey
| | - Ege Ulgen
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Taner Yavuz
- Division of Pediatric Cardiology, Department of Pediatrics, Zeynep Kamil Maternity and Childrens' Diseases Training and Research Hospital, Istanbul 34668, Turkey
| | - Jacob F Baranoski
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Abdulkadir Bozaykut
- Department of Pediatrics, University of Health Sciences, Zeynep Kamil Maternity and Childrens' Diseases Training and Research Hospital, Istanbul 34668, Turkey
| | - Akdes Serin Harmanci
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Yalim Yalcin
- Division of Pediatric Cardiology, Department of Pediatrics, School of Medicine, Istanbul Bilim University, Istanbul 34394, Turkey
| | - Mark W Youngblood
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Katsuhito Yasuno
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Kaya Bilgüvar
- Department of Genetics, Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Murat Gunel
- Departments of Neurosurgery, Neurobiology and Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
67
|
Arbustini E, Favalli V, Narula N. LMNA
Mutations Associated With Mild and Late-Onset Phenotype. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.117.001816. [DOI: 10.1161/circgenetics.117.001816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Eloisa Arbustini
- From the Centre for Inherited Cardiovascular Diseases, IRCCS Foundation University Hospital Policlinico San Matteo, Pavia, Italy (E.A., V.F., N.N.); and Icahn School of Medicine at Mount Sinai, New York, NY (N.N.)
| | - Valentina Favalli
- From the Centre for Inherited Cardiovascular Diseases, IRCCS Foundation University Hospital Policlinico San Matteo, Pavia, Italy (E.A., V.F., N.N.); and Icahn School of Medicine at Mount Sinai, New York, NY (N.N.)
| | - Nupoor Narula
- From the Centre for Inherited Cardiovascular Diseases, IRCCS Foundation University Hospital Policlinico San Matteo, Pavia, Italy (E.A., V.F., N.N.); and Icahn School of Medicine at Mount Sinai, New York, NY (N.N.)
| |
Collapse
|
68
|
Lee YK, Lau YM, Cai ZJ, Lai WH, Wong LY, Tse HF, Ng KM, Siu CW. Modeling Treatment Response for Lamin A/C Related Dilated Cardiomyopathy in Human Induced Pluripotent Stem Cells. J Am Heart Assoc 2017; 6:e005677. [PMID: 28754655 PMCID: PMC5586427 DOI: 10.1161/jaha.117.005677] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/02/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Precision medicine is an emerging approach to disease treatment and prevention that takes into account individual variability in the environment, lifestyle, and genetic makeup of patients. Patient-specific human induced pluripotent stem cells hold promise to transform precision medicine into real-life clinical practice. Lamin A/C (LMNA)-related cardiomyopathy is the most common inherited cardiomyopathy in which a substantial proportion of mutations in the LMNA gene are of nonsense mutation. PTC124 induces translational read-through over the premature stop codon and restores production of the full-length proteins from the affected genes. In this study we generated human induced pluripotent stem cells-derived cardiomyocytes from patients who harbored different LMNA mutations (nonsense and frameshift) to evaluate the potential therapeutic effects of PTC124 in LMNA-related cardiomyopathy. METHODS AND RESULTS We generated human induced pluripotent stem cells lines from 3 patients who carried distinctive mutations (R225X, Q354X, and T518fs) in the LMNA gene. The cardiomyocytes derived from these human induced pluripotent stem cells lines reproduced the pathophysiological hallmarks of LMNA-related cardiomyopathy. Interestingly, PTC124 treatment increased the production of full-length LMNA proteins in only the R225X mutant, not in other mutations. Functional evaluation experiments on the R225X mutant further demonstrated that PTC124 treatment not only reduced nuclear blebbing and electrical stress-induced apoptosis but also improved the excitation-contraction coupling of the affected cardiomyocytes. CONCLUSIONS Using cardiomyocytes derived from human induced pluripotent stem cells carrying different LMNA mutations, we demonstrated that the effect of PTC124 is codon selective. A premature stop codon UGA appeared to be most responsive to PTC124 treatment.
Collapse
Affiliation(s)
- Yee-Ki Lee
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Zhu-Jun Cai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Lai-Yung Wong
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
69
|
Affiliation(s)
- Yoshinori Yoshida
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| | - Shinya Yamanaka
- From the Center for iPS Cell Research and Application, Kyoto University, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA (S.Y.)
| |
Collapse
|
70
|
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
|
71
|
Differential Sarcomere and Electrophysiological Maturation of Human iPSC-Derived Cardiac Myocytes in Monolayer vs. Aggregation-Based Differentiation Protocols. Int J Mol Sci 2017; 18:ijms18061173. [PMID: 28587156 PMCID: PMC5485997 DOI: 10.3390/ijms18061173] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) represent a powerful human model to study cardiac disease in vitro, notably channelopathies and sarcomeric cardiomyopathies. Different protocols for cardiac differentiation of iPSCs have been proposed either based on embroid body formation (3D) or, more recently, on monolayer culture (2D). We performed a direct comparison of the characteristics of the derived cardiomyocytes (iPSC-CMs) on day 27 ± 2 of differentiation between 3D and 2D differentiation protocols with two different Wnt-inhibitors were compared: IWR1 (inhibitor of Wnt response) or IWP2 (inhibitor of Wnt production). We firstly found that the level of Troponin T (TNNT2) expression measured by FACS was significantly higher for both 2D protocols as compared to the 3D protocol. In the three methods, iPSC-CM show sarcomeric structures. However, iPSC-CM generated in 2D protocols constantly displayed larger sarcomere lengths as compared to the 3D protocol. In addition, mRNA and protein analyses reveal higher cTNi to ssTNi ratios in the 2D protocol using IWP2 as compared to both other protocols, indicating a higher sarcomeric maturation. Differentiation of cardiac myocytes with 2D monolayer-based protocols and the use of IWP2 allows the production of higher yield of cardiac myocytes that have more suitable characteristics to study sarcomeric cardiomyopathies.
Collapse
|
72
|
Abstract
The nuclear lamina is a critical structural domain for the maintenance of genomic stability and whole-cell mechanics. Mutations in the LMNA gene, which encodes nuclear A-type lamins lead to the disruption of these key cellular functions, resulting in a number of devastating diseases known as laminopathies. Cardiomyopathy is a common laminopathy and is highly penetrant with poor prognosis. To date, cell mechanical instability and dysregulation of gene expression have been proposed as the main mechanisms driving cardiac dysfunction, and indeed discoveries in these areas have provided some promising leads in terms of therapeutics. However, important questions remain unanswered regarding the role of lamin A dysfunction in the heart, including a potential role for the toxicity of lamin A precursors in LMNA cardiomyopathy, which has yet to be rigorously investigated.
Collapse
Affiliation(s)
- Daniel Brayson
- a King's College London, The James Black Centre , London , United Kingdom
| | | |
Collapse
|
73
|
Fine B, Vunjak-Novakovic G. Shortcomings of Animal Models and the Rise of Engineered Human Cardiac Tissue. ACS Biomater Sci Eng 2017; 3:1884-1897. [PMID: 33440547 DOI: 10.1021/acsbiomaterials.6b00662] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We provide here an historical context of how studies utilizing engineered human cardiac muscle can complement and in some cases substitute animal and cell models for studies of disease and drug testing. We give an overview of the development of animal models and discuss the ability of novel human tissue models to overcome limited predictive power of cell culture and animal models in studies of drug efficacy and safety. The in vitro generation of cardiac tissue is discussed in the context of state of the art in the field. Finally we describe the assembly of multitissue platforms for more accurate representation of integrated human cardiac physiology and consider the advantages of in silico drug trials to augment our ability to predict drug-drug and organ-organ interactions in humans.
Collapse
Affiliation(s)
- Barry Fine
- Department of Biomedical Engineering and ‡Department of Medicine, Columbia University, New York, New York 10027, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering and Department of Medicine, Columbia University, New York, New York 10027, United States
| |
Collapse
|
74
|
Moreau A, Mercier A, Thériault O, Boutjdir M, Burger B, Keller DI, Chahine M. Biophysical, Molecular, and Pharmacological Characterization of Voltage-Dependent Sodium Channels From Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Can J Cardiol 2017; 33:269-278. [DOI: 10.1016/j.cjca.2016.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 10/03/2016] [Accepted: 10/05/2016] [Indexed: 12/16/2022] Open
|
75
|
Smith AST, Macadangdang J, Leung W, Laflamme MA, Kim DH. Human iPSC-derived cardiomyocytes and tissue engineering strategies for disease modeling and drug screening. Biotechnol Adv 2017; 35:77-94. [PMID: 28007615 PMCID: PMC5237393 DOI: 10.1016/j.biotechadv.2016.12.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 12/16/2016] [Accepted: 12/17/2016] [Indexed: 01/13/2023]
Abstract
Improved methodologies for modeling cardiac disease phenotypes and accurately screening the efficacy and toxicity of potential therapeutic compounds are actively being sought to advance drug development and improve disease modeling capabilities. To that end, much recent effort has been devoted to the development of novel engineered biomimetic cardiac tissue platforms that accurately recapitulate the structure and function of the human myocardium. Within the field of cardiac engineering, induced pluripotent stem cells (iPSCs) are an exciting tool that offer the potential to advance the current state of the art, as they are derived from somatic cells, enabling the development of personalized medical strategies and patient specific disease models. Here we review different aspects of iPSC-based cardiac engineering technologies. We highlight methods for producing iPSC-derived cardiomyocytes (iPSC-CMs) and discuss their application to compound efficacy/toxicity screening and in vitro modeling of prevalent cardiac diseases. Special attention is paid to the application of micro- and nano-engineering techniques for the development of novel iPSC-CM based platforms and their potential to advance current preclinical screening modalities.
Collapse
Affiliation(s)
- Alec S T Smith
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jesse Macadangdang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Winnie Leung
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Michael A Laflamme
- Toronto General Research Institute, McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
76
|
Zaunbrecher R, Regnier M. Connecting Sarcomere Protein Mutations to Pathogenesis in Cardiomyopathies: The Development of "Disease in a Dish" Models. Front Physiol 2016; 7:566. [PMID: 27920728 PMCID: PMC5118458 DOI: 10.3389/fphys.2016.00566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/07/2016] [Indexed: 12/22/2022] Open
Abstract
Recent technological and protocol developments have greatly increased the ability to utilize stem cells transformed into cardiomyocytes as models to study human heart muscle development and how this is affected by disease associated mutations in a variety of sarcomere proteins. In this perspective we provide an overview of these emerging technologies and how they are being used to create better models of “disease in a dish” for both research and screening assays. We also consider the value of these assays as models to explore the seminal processes in initiation of the disease development and the possibility of early interventions.
Collapse
Affiliation(s)
| | - Michael Regnier
- Department of Bioengineering, University of WashingtonSeattle, WA, USA; Center for Cardiovascular BiologySeattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of WashingtonSeattle, WA, USA
| |
Collapse
|
77
|
Lee YK, Jiang Y, Ran XR, Lau YM, Ng KM, Lai WHK, Siu CW, Tse HF. Recent advances in animal and human pluripotent stem cell modeling of cardiac laminopathy. Stem Cell Res Ther 2016; 7:139. [PMID: 27649756 PMCID: PMC5029055 DOI: 10.1186/s13287-016-0401-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Laminopathy is a disease closely related to deficiency of the nuclear matrix protein lamin A/C or failure in prelamin A processing, and leads to accumulation of the misfold protein causing progeria. The resultant disrupted lamin function is highly associated with abnormal nuclear architecture, cell senescence, apoptosis, and unstable genome integrity. To date, the effects of loss in nuclear integrity on the susceptible organ, striated muscle, have been commonly associated with muscular dystrophy, dilated cardiac myopathy (DCM), and conduction defeats, but have not been studied intensively. In this review, we aim to summarize recent breakthroughs in an in vivo laminopathy model and in vitro study using patient-specific human induced pluripotent stem cells (iPSCs) that reproduce the pathophysiological phenotype for further drug screening. We describe several in-vivo transgenic mouse models to elucidate the effects of Lmna H222P, N195K mutations, and LMNA knockout on cardiac function, in terms of hemodynamic and electrical signal propagation; certain strategies targeted on stress-related MAPK are mentioned. We will also discuss human iPSC cardiomyocytes serving as a platform to reveal the underlying mechanisms, such as the altered mechanical sensation in electrical coupling of the heart conduction system and ion channel alternation in relation to altered nuclear architecture, and furthermore to enable screening of drugs that can attenuate this cardiac premature aging phenotype by inhibition of prelamin misfolding and oxidative stress, and also enhancement of autophagy protein clearance and cardiac-protective microRNA.
Collapse
Affiliation(s)
- Yee-Ki Lee
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Yu Jiang
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Xin-Ru Ran
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Wing-Hon Kevin Lai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, People's Republic of China. .,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China. .,Shenzhen Institutes of Research and Innovation, University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
78
|
Chamberlain SJ. Disease modelling using human iPSCs. Hum Mol Genet 2016; 25:R173-R181. [PMID: 27493026 DOI: 10.1093/hmg/ddw209] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 12/17/2022] Open
Affiliation(s)
- Stormy J Chamberlain
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
79
|
Li C, Chen Z, Yang H, Luo F, Chen L, Cai H, Li Y, You G, Long D, Li S, Zhang Q, Rao L. Selumetinib, an Oral Anti-Neoplastic Drug, May Attenuate Cardiac Hypertrophy via Targeting the ERK Pathway. PLoS One 2016; 11:e0159079. [PMID: 27438013 PMCID: PMC4954659 DOI: 10.1371/journal.pone.0159079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/27/2016] [Indexed: 02/05/2023] Open
Abstract
Aims Although extracellular-regulated kinases (ERK) are a well-known central mediator in cardiac hypertrophy, no clinically available ERK antagonist has been tested for preventing cardiac hypertrophy. Selumetinib is a novel oral MEK inhibitor that is currently under Phase II and Phase III clinical investigation for advanced solid tumors. In this study, we investigated whether Selumetinib could inhibit the aberrant ERK activation of the heart in response to stress as well as prevent cardiac hypertrophy. Methods and Results In an in vitro model of PE-induced cardiac hypertrophy, Selumetinib significantly inhibited the ERK activation and prevented enlargement of cardiomyocytes or reactivation of certain fetal genes. In the pathologic cardiac hypertrophy model of ascending aortic constriction, Selumetinib provided significant ERK inhibition in the stressed heart but not in the other organs. This selective ERK inhibition prevented left ventricular (LV) wall thickening, LV mass increase, fetal gene reactivation and cardiac fibrosis. In another distinct physiologic cardiac hypertrophy model of a swimming rat, Selumetinib provided a similar anti-hypertrophy effect, except that no significant fetal gene reactivation or cardiac fibrosis was observed. Conclusions Selumetinib, a novel oral anti-cancer drug with good safety records in a number of Phase II clinical trials, can inhibit ERK activity in the heart and prevent cardiac hypertrophy. These promising results indicate that Selumetinib could potentially be used to treat cardiac hypertrophy. However, this hypothesis needs to be validated in human clinical trials.
Collapse
Affiliation(s)
- Chen Li
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhongxiu Chen
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Hao Yang
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Fangbo Luo
- Department of Rehabilitation, Community Health Center of Shuangnan Wuhou District, Chengdu, Sichuan, China
| | - Lihong Chen
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yajiao Li
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Guiying You
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital of Sichuan University, High-tech Zone, Chengdu, Sichuan, China
| | - Shengfu Li
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital of Sichuan University, High-tech Zone, Chengdu, Sichuan, China
| | - Qiuping Zhang
- King’s College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London, United Kingdom
| | - Li Rao
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
80
|
Matsa E, Ahrens JH, Wu JC. Human Induced Pluripotent Stem Cells as a Platform for Personalized and Precision Cardiovascular Medicine. Physiol Rev 2016; 96:1093-126. [PMID: 27335446 PMCID: PMC6345246 DOI: 10.1152/physrev.00036.2015] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have revolutionized the field of human disease modeling, with an enormous potential to serve as paradigm shifting platforms for preclinical trials, personalized clinical diagnosis, and drug treatment. In this review, we describe how hiPSCs could transition cardiac healthcare away from simple disease diagnosis to prediction and prevention, bridging the gap between basic and clinical research to bring the best science to every patient.
Collapse
Affiliation(s)
- Elena Matsa
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - John H Ahrens
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
81
|
Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as a Model for Heart Development and Congenital Heart Disease. Stem Cell Rev Rep 2016; 11:710-27. [PMID: 26085192 DOI: 10.1007/s12015-015-9596-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Congenital heart disease (CHD) remains a significant health problem, with a growing population of survivors with chronic disease. Despite intense efforts to understand the genetic basis of CHD in humans, the etiology of most CHD is unknown. Furthermore, new models of CHD are required to better understand the development of CHD and to explore novel therapies for this patient population. In this review, we highlight the role that human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes can serve to enhance our understanding of the development, pathophysiology and potential therapeutic targets for CHD. We highlight the use of hiPSC-derived cardiomyocytes to model gene regulatory interactions, cell-cell interactions and tissue interactions contributing to CHD. We further emphasize the importance of using hiPSC-derived cardiomyocytes as personalized research models. The use of hiPSCs presents an unprecedented opportunity to generate disease-specific cellular models, investigate the underlying molecular mechanisms of disease and uncover new therapeutic targets for CHD.
Collapse
|
82
|
West G, Gullmets J, Virtanen L, Li SP, Keinänen A, Shimi T, Mauermann M, Heliö T, Kaartinen M, Ollila L, Kuusisto J, Eriksson JE, Goldman RD, Herrmann H, Taimen P. Deleterious assembly of the lamin A/C mutant p.S143P causes ER stress in familial dilated cardiomyopathy. J Cell Sci 2016; 129:2732-43. [PMID: 27235420 DOI: 10.1242/jcs.184150] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/20/2016] [Indexed: 01/12/2023] Open
Abstract
Mutation of the LMNA gene, encoding nuclear lamin A and lamin C (hereafter lamin A/C), is a common cause of familial dilated cardiomyopathy (DCM). Among Finnish DCM patients, the founder mutation c.427T>C (p.S143P) is the most frequently reported genetic variant. Here, we show that p.S143P lamin A/C is more nucleoplasmic and soluble than wild-type lamin A/C and accumulates into large intranuclear aggregates in a fraction of cultured patient fibroblasts as well as in cells ectopically expressing either FLAG- or GFP-tagged p.S143P lamin A. In fluorescence loss in photobleaching (FLIP) experiments, non-aggregated EGFP-tagged p.S143P lamin A was significantly more dynamic. In in vitro association studies, p.S143P lamin A failed to form appropriate filament structures but instead assembled into disorganized aggregates similar to those observed in patient cell nuclei. A whole-genome expression analysis revealed an elevated unfolded protein response (UPR) in cells expressing p.S143P lamin A/C. Additional endoplasmic reticulum (ER) stress induced by tunicamycin reduced the viability of cells expressing mutant lamin further. In summary, p.S143P lamin A/C affects normal lamina structure and influences the cellular stress response, homeostasis and viability.
Collapse
Affiliation(s)
- Gun West
- Department of Pathology, University of Turku and Turku University Hospital, 20520 Turku, Finland MediCity Research Laboratory, 20520 Turku, Finland
| | - Josef Gullmets
- Department of Pathology, University of Turku and Turku University Hospital, 20520 Turku, Finland MediCity Research Laboratory, 20520 Turku, Finland Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Laura Virtanen
- Department of Pathology, University of Turku and Turku University Hospital, 20520 Turku, Finland MediCity Research Laboratory, 20520 Turku, Finland
| | - Song-Ping Li
- Department of Pathology, University of Turku and Turku University Hospital, 20520 Turku, Finland MediCity Research Laboratory, 20520 Turku, Finland
| | - Anni Keinänen
- Department of Pathology, University of Turku and Turku University Hospital, 20520 Turku, Finland MediCity Research Laboratory, 20520 Turku, Finland
| | - Takeshi Shimi
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Monika Mauermann
- Division of Molecular Genetics, German Cancer Research Center, 69120 Heidelberg, Germany Institute of Neuropathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Tiina Heliö
- Heart and Lung Center Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Maija Kaartinen
- Heart and Lung Center Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Laura Ollila
- Heart and Lung Center Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - John E Eriksson
- Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Robert D Goldman
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Harald Herrmann
- Division of Molecular Genetics, German Cancer Research Center, 69120 Heidelberg, Germany Institute of Neuropathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Pekka Taimen
- Department of Pathology, University of Turku and Turku University Hospital, 20520 Turku, Finland MediCity Research Laboratory, 20520 Turku, Finland
| |
Collapse
|
83
|
Chen IY, Matsa E, Wu JC. Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat Rev Cardiol 2016; 13:333-49. [PMID: 27009425 DOI: 10.1038/nrcardio.2016.36] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The advent of human induced pluripotent stem cell (hiPSC) technology has revitalized the efforts in the past decade to realize more fully the potential of human embryonic stem cells for scientific research. Adding to the possibility of generating an unlimited amount of any cell type of interest, hiPSC technology now enables the derivation of cells with patient-specific phenotypes. Given the introduction and implementation of the large-scale Precision Medicine Initiative, hiPSC technology will undoubtedly have a vital role in the advancement of cardiovascular research and medicine. In this Review, we summarize the progress that has been made in the field of hiPSC technology, with particular emphasis on cardiovascular disease modelling and drug development. The growing roles of hiPSC technology in the practice of precision medicine will also be discussed.
Collapse
Affiliation(s)
- Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
84
|
An Overview of Direct Somatic Reprogramming: The Ins and Outs of iPSCs. Int J Mol Sci 2016; 17:ijms17010141. [PMID: 26805822 PMCID: PMC4730380 DOI: 10.3390/ijms17010141] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
Stem cells are classified into embryonic stem cells and adult stem cells. An evolving alternative to conventional stem cell therapies is induced pluripotent stem cells (iPSCs), which have a multi-lineage potential comparable to conventionally acquired embryonic stem cells with the additional benefits of being less immunoreactive and avoiding many of the ethical concerns raised with the use of embryonic material. The ability to generate iPSCs from somatic cells provides tremendous promise for regenerative medicine. The breakthrough of iPSCs has raised the possibility that patient-specific iPSCs can provide autologous cells for cell therapy without the concern for immune rejection. iPSCs are also relevant tools for modeling human diseases and drugs screening. However, there are still several hurdles to overcome before iPSCs can be used for translational purposes. Here, we review the recent advances in somatic reprogramming and the challenges that must be overcome to move this strategy closer to clinical application.
Collapse
|
85
|
Lee YK, Lau YM, Ng KM, Lai WH, Ho SL, Tse HF, Siu CW, Ho PWL. Efficient attenuation of Friedreich's ataxia (FRDA) cardiomyopathy by modulation of iron homeostasis-human induced pluripotent stem cell (hiPSC) as a drug screening platform for FRDA. Int J Cardiol 2016; 203:964-71. [PMID: 26625322 DOI: 10.1016/j.ijcard.2015.11.101] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/16/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Friedreich's ataxia (FRDA), a recessive neurodegenerative disorder commonly associated with hypertrophic cardiomyopathy, is caused by silencing of the frataxin (FXN) gene encoding the mitochondrial protein involved in iron-sulfur cluster biosynthesis. METHODS Application of our previously established FRDA human induced pluripotent stem cell (hiPSC) derived cardiomyocytes model as a platform to assess the efficacy of treatment with either the antioxidant coenzyme Q10 analog, idebenone (IDE) or the iron chelator, deferiprone (DFP), which are both under clinical trial. RESULTS DFP was able to more significantly suppress synthesis of reactive oxygen species (ROS) than IDE at the dosages of 25 μM and 10nM respectively which agreed with the reduced rate of intracellular accumulation of iron by DFP treatment from 25 to 50 μM. With regard to cardiac electrical-contraction (EC) coupling function, decay velocity of calcium handling kinetics in FRDA-hiPSC-cardiomyocytes was significantly improved by DFP treatment but not by IDE. Further mechanistic studies revealed that DFP also modulated iron induced mitochondrial stress as reflected by mitochondria network disorganization and decline level of respiratory chain protein, succinate dehydrogenase (CxII) and cytochrome c oxidase (COXIV). In addition, iron-response protein (IRP-1) regulatory loop was overridden by DFP as reflected by resumed level of ferritin (FTH) back to basal level and the attenuated transferrin receptor (TSFR) mRNA level suppression thereby reducing further iron uptake. CONCLUSIONS DFP modulated iron homeostasis in FRDA-hiPSC-cardiomyocytes and effectively relieved stress-stimulation related to cardiomyopathy. The resuming of redox condition led to the significantly improved cardiac prime events, cardiac electrical-coupling during contraction.
Collapse
Affiliation(s)
- Yee-Ki Lee
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shu-Leong Ho
- Neurology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong, China; Hong Kong - Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Philip Wing-Lok Ho
- Neurology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
86
|
Jones AR, Edwards DH, Cummins MJ, Williams AJ, George CH. A Systemized Approach to Investigate Ca(2+) Synchronization in Clusters of Human Induced Pluripotent Stem-Cell Derived Cardiomyocytes. Front Cell Dev Biol 2016; 3:89. [PMID: 26793710 DOI: 10.3389/fcell.2015.00089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 12/20/2015] [Indexed: 12/13/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (IPS-CM) are considered by many to be the cornerstone of future approaches to repair the diseased heart. However, current methods for producing IPS-CM typically yield highly variable populations with low batch-to-batch reproducibility. The underlying reasons for this are not fully understood. Here we report on a systematized approach to investigate the effect of maturation in embryoid bodies (EB) vs. "on plate" culture on spontaneous activity and regional Ca(2+) synchronization in IPS-CM clusters. A detailed analysis of the temporal and spatial organization of Ca(2+) spikes in IPS-CM clusters revealed that the disaggregation of EBs between 0.5 and 2 weeks produced IPS-CM characterized by spontaneous beating and high levels of regional Ca(2+) synchronization. These phenomena were typically absent in IPS-CM obtained from older EBs (>2 weeks). The maintenance of all spontaneously active IPS-CM clusters under "on plate" culture conditions promoted the progressive reduction in regional Ca(2+) synchronization and the loss of spontaneous Ca(2+) spiking. Raising the extracellular [Ca(2+)] surrounding these quiescent IPS-CM clusters from ~0.4 to 1.8 mM unmasked discrete behaviors typified by either (a) long-lasting Ca(2+) elevation that returned to baseline or (b) persistent, large-amplitude Ca(2+) oscillations around an increased cytoplasmic [Ca(2+)]. The different responses of IPS-CM to elevated extracellular [Ca(2+)] could be traced back to their routes of derivation. The data point to the possibility of predictably influencing IPS-CM phenotype and response to external activation via defined interventions at early stages in their maturation.
Collapse
Affiliation(s)
- Aled R Jones
- Ionic Cell Signalling, School of Medicine, Wales Heart Research Institute, Cardiff University Wales, UK
| | - David H Edwards
- Ionic Cell Signalling, School of Medicine, Wales Heart Research Institute, Cardiff University Wales, UK
| | - Michael J Cummins
- Ionic Cell Signalling, School of Medicine, Wales Heart Research Institute, Cardiff University Wales, UK
| | - Alan J Williams
- Ionic Cell Signalling, School of Medicine, Wales Heart Research Institute, Cardiff University Wales, UK
| | - Christopher H George
- Ionic Cell Signalling, School of Medicine, Wales Heart Research Institute, Cardiff University Wales, UK
| |
Collapse
|
87
|
Campbell KA, Terzic A, Nelson TJ. Induced pluripotent stem cells for cardiovascular disease: from product-focused disease modeling to process-focused disease discovery. Regen Med 2015; 10:773-83. [PMID: 26439809 DOI: 10.2217/rme.15.41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Induced pluripotent stem (iPS) cell technology offers an unprecedented opportunity to study patient-specific disease. This biotechnology platform enables recapitulation of individualized disease signatures in a dish through differentiation of patient-derived iPS cells. Beyond disease modeling, the in vitro process of differentiation toward genuine patient tissue offers a blueprint to inform disease etiology and molecular pathogenesis. Here, we highlight recent advances in patient-specific cardiac disease modeling and outline the future promise of iPS cell-based disease discovery applications.
Collapse
Affiliation(s)
- Katherine A Campbell
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andre Terzic
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.,Department of Medical Genetics, Mayo Clinic, Rochester, MN, USA
| | - Timothy J Nelson
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.,Center for Transplantation & Clinical Regeneration, Mayo Clinic, Rochester, MN, USA.,Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
88
|
Cardiovascular Disease Modeling Using Patient-Specific Induced Pluripotent Stem Cells. Int J Mol Sci 2015; 16:18894-922. [PMID: 26274955 PMCID: PMC4581278 DOI: 10.3390/ijms160818894] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022] Open
Abstract
The generation of induced pluripotent stem cells (iPSCs) has opened up a new scientific frontier in medicine. This technology has made it possible to obtain pluripotent stem cells from individuals with genetic disorders. Because iPSCs carry the identical genetic anomalies related to those disorders, iPSCs are an ideal platform for medical research. The pathophysiological cellular phenotypes of genetically heritable heart diseases such as arrhythmias and cardiomyopathies, have been modeled on cell culture dishes using disease-specific iPSC-derived cardiomyocytes. These model systems can potentially provide new insights into disease mechanisms and drug discoveries. This review focuses on recent progress in cardiovascular disease modeling using iPSCs, and discusses problems and future perspectives concerning their use.
Collapse
|
89
|
Kamdar F, Klaassen Kamdar A, Koyano-Nakagawa N, Garry MG, Garry DJ. Cardiomyopathy in a dish: using human inducible pluripotent stem cells to model inherited cardiomyopathies. J Card Fail 2015; 21:761-70. [PMID: 25934595 DOI: 10.1016/j.cardfail.2015.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 04/11/2015] [Accepted: 04/17/2015] [Indexed: 12/15/2022]
Abstract
Inherited cardiomyopathies, including hypertrophic cardiomyopathy, dilated cardiomyopathies, arrythmogenic right ventricular cardiomyopathy, and other inherited forms of heart failure, represent a unique set of genetically defined cardiovascular disease processes. Unraveling the molecular mechanisms of these deadly forms of human heart disease has been challenging, but recent groundbreaking scientific advances in stem cell technology have allowed for the generation of patient-specific human inducible stem cell (hiPSC)-derived cardiomyocytes (CMs). hiPSC-derived CMs retain the genetic blueprint of the patient, they can be maintained in culture, and they recapitulate the phenotypic characteristics of the disease in vitro, thus serving as a disease in a dish. This review provides an overview of in vitro modeling of inherited cardiomyopathies with the use of patient-specific hiPSC-derived CMs.
Collapse
Affiliation(s)
- Forum Kamdar
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Andre Klaassen Kamdar
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Naoko Koyano-Nakagawa
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Mary G Garry
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Daniel J Garry
- Cardiovascular Division and Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
90
|
Matsa E, Burridge PW, Wu JC. Human stem cells for modeling heart disease and for drug discovery. Sci Transl Med 2015; 6:239ps6. [PMID: 24898747 DOI: 10.1126/scitranslmed.3008921] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A major research focus in the field of cardiovascular medicine is the prospect of using stem cells and progenitor cells for cardiac regeneration. With the advent of induced pluripotent stem cell (iPSC) technology, major efforts are also underway to use iPSCs to model heart disease, to screen for new drugs, and to test candidate drugs for cardiotoxicity. Here, we discuss recent advances in the exciting fields of stem cells and cardiovascular disease.
Collapse
Affiliation(s)
- Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA. Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul W Burridge
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA. Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA. Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
91
|
Eschenhagen T, Mummery C, Knollmann BC. Modelling sarcomeric cardiomyopathies in the dish: from human heart samples to iPSC cardiomyocytes. Cardiovasc Res 2015; 105:424-38. [PMID: 25618410 PMCID: PMC4349163 DOI: 10.1093/cvr/cvv017] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
One of the obstacles to a better understanding of the pathogenesis of human cardiomyopathies has been poor availability of heart-tissue samples at early stages of disease development. This has possibly changed by the advent of patient-derived induced pluripotent stem cell (hiPSC) from which cardiomyocytes can be derived in vitro. The main promise of hiPSC technology is that by capturing the effects of thousands of individual gene variants, the phenotype of differentiated derivatives of these cells will provide more information on a particular disease than simple genotyping. This article summarizes what is known about the ‘human cardiomyopathy or heart failure phenotype in vitro’, which constitutes the reference for modelling sarcomeric cardiomyopathies in hiPSC-derived cardiomyocytes. The current techniques for hiPSC generation and cardiac myocyte differentiation are briefly reviewed and the few published reports of hiPSC models of sarcomeric cardiomyopathies described. A discussion of promises and challenges of hiPSC-modelling of sarcomeric cardiomyopathies and individualized approaches is followed by a number of questions that, in the view of the authors, need to be answered before the true potential of this technology can be evaluated.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Martinistr. 52, 20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333ZC Leiden, The Netherlands
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, 2215 Garland Ave, Nashville, TN 37232, USA
| |
Collapse
|
92
|
Abstract
PURPOSE OF REVIEW This article provides an overview of the latest advances in in-vitro modeling of inherited cardiomyopathies using human-induced pluripotent stem cells (iPSCs). RECENT FINDINGS Inherited cardiomyopathies have been recently modeled by generating iPSCs from patients harboring mutations in genes associated with the pathogenesis of hypertrophic cardiomyopathy, dilated cardiomyopathy, and arrhythmogenic right ventricular cardiomyopathy/dysplasia. SUMMARY Patient-specific iPSCs and their differentiated cardiomyocytes (induced pluripotent stem cell-derived cardiomyocytes) now provide a novel model to study the underlying molecular mechanism of the pathogenesis of familial cardiomyopathies as well as for in-vitro drug screening and drug discovery.
Collapse
|
93
|
Savla JJ, Nelson BC, Perry CN, Adler ED. Induced Pluripotent Stem Cells for the Study of Cardiovascular Disease. J Am Coll Cardiol 2014; 64:512-9. [DOI: 10.1016/j.jacc.2014.05.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 12/16/2022]
|
94
|
Fedorchak GR, Kaminski A, Lammerding J. Cellular mechanosensing: getting to the nucleus of it all. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:76-92. [PMID: 25008017 PMCID: PMC4252489 DOI: 10.1016/j.pbiomolbio.2014.06.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 12/12/2022]
Abstract
Cells respond to mechanical forces by activating specific genes and signaling pathways that allow the cells to adapt to their physical environment. Examples include muscle growth in response to exercise, bone remodeling based on their mechanical load, or endothelial cells aligning under fluid shear stress. While the involved downstream signaling pathways and mechanoresponsive genes are generally well characterized, many of the molecular mechanisms of the initiating 'mechanosensing' remain still elusive. In this review, we discuss recent findings and accumulating evidence suggesting that the cell nucleus plays a crucial role in cellular mechanotransduction, including processing incoming mechanoresponsive signals and even directly responding to mechanical forces. Consequently, mutations in the involved proteins or changes in nuclear envelope composition can directly impact mechanotransduction signaling and contribute to the development and progression of a variety of human diseases, including muscular dystrophy, cancer, and the focus of this review, dilated cardiomyopathy. Improved insights into the molecular mechanisms underlying nuclear mechanotransduction, brought in part by the emergence of new technologies to study intracellular mechanics at high spatial and temporal resolution, will not only result in a better understanding of cellular mechanosensing in normal cells but may also lead to the development of novel therapies in the many diseases linked to defects in nuclear envelope proteins.
Collapse
Affiliation(s)
- Gregory R Fedorchak
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Ashley Kaminski
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jan Lammerding
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
95
|
Abstract
OPINION STATEMENT Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) represent a powerful new model system to study the basic mechanisms of inherited cardiomyopathies. hiPSC-CMs have been utilized to model several cardiovascular diseases, achieving the most success in the inherited arrhythmias, including long QT and Timothy syndromes (Moretti et al. N Engl J Med. 363:1397-409, 2010; Yazawa et al. Nature. 471:230-4, 2011) and arrhythmogenic right ventricular dysplasia (ARVD) (Ma et al. Eur Heart J. 34:1122-33, 2013). Recently, studies have applied hiPSC-CMs to the study of both dilated (DCM) (Sun et al. Sci Transl Med. 4:130ra47, 2012) and hypertrophic (HCM) cardiomyopathies (Lan et al. Cell Stem Cell. 12:101-13, 2013; Carvajal-Vergara et al. Nature. 465:808-12, 2010), providing new insights into basic mechanisms of disease. However, hiPSC-CMs do not recapitulate many of the structural and functional aspects of mature human cardiomyocytes, instead mirroring an immature - embryonic or fetal - phenotype. Much work remains in order to better understand these differences, as well as to develop methods to induce hiPSC-CMs into a fully mature phenotype. Despite these limitations, hiPSC-CMs represent the best current in vitro correlate of the human heart and an invaluable tool in the search for mechanisms underlying cardiomyopathy and for screening new pharmacologic therapies.
Collapse
|
96
|
Cau P, Navarro C, Harhouri K, Roll P, Sigaudy S, Kaspi E, Perrin S, De Sandre-Giovannoli A, Lévy N. WITHDRAWN: Nuclear matrix, nuclear envelope and premature aging syndromes in a translational research perspective. Semin Cell Dev Biol 2014:S1084-9521(14)00058-5. [PMID: 24685615 DOI: 10.1016/j.semcdb.2014.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/03/2014] [Accepted: 03/09/2014] [Indexed: 10/25/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, http://dx.doi.org/10.1016/j.semcdb.2014.03.022. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Pierre Cau
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); AP-HM, Service de Biologie Cellulaire, Hôpital La Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France(2).
| | - Claire Navarro
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1)
| | - Karim Harhouri
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1)
| | - Patrice Roll
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); AP-HM, Service de Biologie Cellulaire, Hôpital La Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France(2)
| | - Sabine Sigaudy
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); AP-HM, Département de Génétique Médicale, Hôpital d'enfants Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France(3)
| | - Elise Kaspi
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); AP-HM, Service de Biologie Cellulaire, Hôpital La Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France(2)
| | - Sophie Perrin
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1)
| | - Annachiara De Sandre-Giovannoli
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); AP-HM, Département de Génétique Médicale, Hôpital d'enfants Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France(3)
| | - Nicolas Lévy
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France(1); AP-HM, Département de Génétique Médicale, Hôpital d'enfants Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France(3).
| |
Collapse
|
97
|
Cau P, Navarro C, Harhouri K, Roll P, Sigaudy S, Kaspi E, Perrin S, De Sandre-Giovannoli A, Lévy N. Nuclear matrix, nuclear envelope and premature aging syndromes in a translational research perspective. Semin Cell Dev Biol 2014; 29:125-47. [PMID: 24662892 DOI: 10.1016/j.semcdb.2014.03.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lamin A-related progeroid syndromes are genetically determined, extremely rare and severe. In the past ten years, our knowledge and perspectives for these diseases has widely progressed, through the progressive dissection of their pathophysiological mechanisms leading to precocious and accelerated aging, from the genes mutations discovery until therapeutic trials in affected children. A-type lamins are major actors in several structural and functional activities at the nuclear periphery, as they are major components of the nuclear lamina. However, while this is usually poorly considered, they also play a key role within the rest of the nucleoplasm, whose defects are related to cell senescence. Although nuclear shape and nuclear envelope deformities are obvious and visible events, nuclear matrix disorganization and abnormal composition certainly represent the most important causes of cell defects with dramatic pathological consequences. Therefore, lamin-associated diseases should be better referred as laminopathies instead of envelopathies, this later being too restrictive, considering neither the key structural and functional roles of soluble lamins in the entire nucleoplasm, nor the nuclear matrix contribution to the pathophysiology of lamin-associated disorders and in particular in defective lamin A processing-associated aging diseases. Based on both our understanding of pathophysiological mechanisms and the biological and clinical consequences of progeria and related diseases, therapeutic trials have been conducted in patients and were terminated less than 10 years after the gene discovery, a quite fast issue for a genetic disease. Pharmacological drugs have been repurposed and used to decrease the toxicity of the accumulated, unprocessed and truncated prelaminA in progeria. To date, none of them may be considered as a cure for progeria and these clinical strategies were essentially designed toward reducing a subset of the most dramatic and morbid features associated to progeria. New therapeutic strategies under study, in particular targeting the protein expression pathway at the mRNA level, have shown a remarkable efficacy both in vitro in cells and in vivo in mice models. Strategies intending to clear the toxic accumulated proteins from the nucleus are also under evaluation. However, although exceedingly rare, improving our knowledge of genetic progeroid syndromes and searching for innovative and efficient therapies in these syndromes is of paramount importance as, even before they can be used to save lives, they may significantly (i) expand the affected childrens' lifespan and preserve their quality of life; (ii) improve our understanding of aging-related disorders and other more common diseases; and (iii) expand our fundamental knowledge of physiological aging and its links with major physiological processes such as those involved in oncogenesis.
Collapse
Affiliation(s)
- Pierre Cau
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; AP-HM, Service de Biologie Cellulaire, Hôpital La Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France.
| | - Claire Navarro
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France
| | - Karim Harhouri
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France
| | - Patrice Roll
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; AP-HM, Service de Biologie Cellulaire, Hôpital La Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France
| | - Sabine Sigaudy
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; AP-HM, Département de Génétique Médicale, Hôpital d'enfants Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France
| | - Elise Kaspi
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; AP-HM, Service de Biologie Cellulaire, Hôpital La Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France
| | - Sophie Perrin
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France
| | - Annachiara De Sandre-Giovannoli
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; AP-HM, Département de Génétique Médicale, Hôpital d'enfants Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France
| | - Nicolas Lévy
- Aix-Marseille Université, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; INSERM, UMR_S 910, Génétique Médicale et Génomique Fonctionnelle, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 5, France; AP-HM, Département de Génétique Médicale, Hôpital d'enfants Timone, 264 Rue Saint Pierre, 13385 Marseille Cedex 5, France.
| |
Collapse
|
98
|
Abstract
Advances in the understanding and treatment of cardiac disorders have been thwarted by the inability to study beating human cardiac cells in vitro. Induced pluripotent stem cells (iPSCs) bypass this hurdle by enabling the creation of patient-specific iPSC-derived cardiomyocytes (iPSC-CMs). These cells provide a unique platform to study cardiac diseases in vitro, especially hereditary cardiac conditions. To date, iPSC-CMs have been used to successfully model arrhythmic disorders, showing excellent recapitulation of cardiac channel function and electrophysiologic features of long QT syndrome types 1, 2, 3, and 8, and catecholaminergic polymorphic ventricular tachycardia (CPVT). Similarly, iPSC-CM models of dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) have shown robust correlation of predicted morphologic, contractile, and electrical phenotypes. In addition, iPSC-CMs have shown some features of the respective phenotypes for arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C), LEOPARD syndrome, Pompe's disease, and Friedriech's ataxia. In this review, we examine the progress of utilizing iPSC-CMs as a model for cardiac conditions and analyze the potential for the platform in furthering the biology and treatment of cardiac disorders.
Collapse
|
99
|
Abstract
Lamins A and C, encoded by LMNA, are constituent of the nuclear lamina, a meshwork of proteins underneath the nuclear envelope first described as scaffolding proteins of the nucleus. Since the discovery of LMNA mutations in highly heterogeneous human disorders (including cardiac and muscular dystrophies, lipodystrophies and progeria), the number of functions described for lamin A/C has expanded. Lamin A/C is notably involved in the regulation of chromatin structure and gene transcription, and in the resistance of cells to mechanical stress. This review focuses on studies performed on knock-out and knock-in Lmna mouse models, which have led to decipher some of the lamin A/C functions in striated muscles and to the first preclinical trials of pharmaceutical therapies.
Collapse
|
100
|
Starke S, Meinke P, Camozzi D, Mattioli E, Pfaeffle R, Siekmeyer M, Hirsch W, Horn LC, Paasch U, Mitter D, Lattanzi G, Wehnert M, Kiess W. Progeroid laminopathy with restrictive dermopathy-like features caused by an isodisomic LMNA mutation p.R435C. Aging (Albany NY) 2013; 5:445-59. [PMID: 23804595 PMCID: PMC3824411 DOI: 10.18632/aging.100566] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The clinical course of a female patient affected by a progeroid syndrome with Restrictive Dermopathy (RD)-like features was followed up. Besides missing hairiness, stagnating weight and growth, RD-like features including progressive skin swelling and solidification, acrocontractures, osteolysis and muscular hypotension were observed until the patient died at the age of 11 months. A homozygous LMNA mutation c.1303C>T (p.R435C) was found by Sanger sequencing. Haplotyping revealed a partial uniparental disomy of chromosome 1 (1q21.3 to 1q23.1) including the LMNA gene. In contrast to reported RD patients with LMNA mutations, LMNA p.R435C is not located at the cleavage site necessary for processing of prelamin A by ZMPSTE24 and leads to a distinct phenotype combining clinical features of Restrictive Dermopathy, Mandibuloacral Dysplasia and Hutchinson-Gilford Progeria. Functionally, LMNA p.R435C is associated with increasing DNA double strand breaks and decreased recruitment of P53 binding protein 1 (53BP1) to DNA-damage sites indicating delayed DNA repair. The follow-up of the complete clinical course in the patient combined with functional studies showed for the first time that a progressive loss of lamin A rather than abnormal accumulation of prelamin A species could be a pathophysiological mechanism in progeroid laminopathies, which leads to DNA repair deficiency accompanied by advancing tissue degeneration.
Collapse
Affiliation(s)
- Sven Starke
- Department of Women and Child Health, Hospital for Children and Adolescents, Centre of Pediatric Research, University Hospital, University of Leipzig, Leipzig, Germany. ‐leipzig.de
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|