1
|
Gregorich ZR, Larson EJ, Zhang Y, Braz CU, Liu C, Ge Y, Guo W. Integrated proteomics and transcriptomics analysis reveals insights into differences in premature mortality associated with disparate pathogenic RBM20 variants. J Mol Cell Cardiol 2024; 197:78-89. [PMID: 39490642 PMCID: PMC11588510 DOI: 10.1016/j.yjmcc.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Variants in RNA binding motif protein 20 (RBM20) are causative in a severe form of dilated cardiomyopathy referred to as RBM20 cardiomyopathy, yet the mechanisms are unclear. Moreover, the reason(s) for phenotypic heterogeneity in carriers with different pathogenic variants are similarly opaque. To gain insight, we carried out multi-omics analysis, including the first analysis of gene expression changes at the protein level, of mice carrying two different pathogenic variants in the RBM20 nuclear localization signal (NLS). Direct comparison of the phenotypes confirmed greater premature morality in S639G variant carrying mice compared to mice with the S637A variant despite similar cardiac remodeling and dysfunction. Analysis of differentially spliced genes uncovered alterations in the splicing of both RBM20 target genes and non-target genes, including several genes previously implicated in arrhythmia. Global proteomics analysis found that a greater number of proteins were differentially expressed in the hearts of Rbm20S639G mice relative to WT than in Rbm20S637A versus WT. Gene ontology analysis suggested greater mitochondrial dysfunction in Rbm20S639G mice, although direct comparison of protein expression in the hearts of Rbm20S639G versus Rbm20S637A mice failed to identify any significant differences. Similarly, few differences were found by direct comparison of gene expression at the transcript level in Rbm20S639G and Rbm20S637A despite greater coverage. Our data provide a comprehensive overview of gene splicing and expression differences associated with pathogenic variants in RBM20, as well as insights into the molecular underpinnings of phenotypic heterogeneity associated with different dilated cardiomyopathy-associated variants.
Collapse
Affiliation(s)
- Zachery R Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eli J Larson
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yanghai Zhang
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Camila U Braz
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Chunling Liu
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Human Proteomics Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
2
|
Du X, Jia H, Chang Y, Zhao Y, Song J. Progress of organoid platform in cardiovascular research. Bioact Mater 2024; 40:88-103. [PMID: 38962658 PMCID: PMC11220467 DOI: 10.1016/j.bioactmat.2024.05.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiovascular disease is a significant cause of death in humans. Various models are necessary for the study of cardiovascular diseases, but once cellular and animal models have some defects, such as insufficient fidelity. As a new technology, organoid has certain advantages and has been used in many applications in the study of cardiovascular diseases. This article aims to summarize the application of organoid platforms in cardiovascular diseases, including organoid construction schemes, modeling, and application of cardiovascular organoids. Advances in cardiovascular organoid research have provided many models for different cardiovascular diseases in a variety of areas, including myocardium, blood vessels, and valves. Physiological and pathological models of different diseases, drug research models, and methods for evaluating and promoting the maturation of different kinds of organ tissues are provided for various cardiovascular diseases, including cardiomyopathy, myocardial infarction, and atherosclerosis. This article provides a comprehensive overview of the latest research progress in cardiovascular organ tissues, including construction protocols for cardiovascular organoid tissues and their evaluation system, different types of disease models, and applications of cardiovascular organoid models in various studies. The problems and possible solutions in organoid development are summarized.
Collapse
Affiliation(s)
- Xingchao Du
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| |
Collapse
|
3
|
Akagi K, Baba S, Fujita H, Fuseya Y, Yoshinaga D, Kubota H, Kume E, Fukumura F, Matsuda K, Tanaka T, Hirata T, Saito MK, Iwai K, Takita J. HOIL-1L deficiency induces cell cycle alteration which causes immaturity of skeletal muscle and cardiomyocytes. Sci Rep 2024; 14:8871. [PMID: 38632277 PMCID: PMC11024103 DOI: 10.1038/s41598-024-57504-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
HOIL-1L deficiency was recently reported to be one of the causes of myopathy and dilated cardiomyopathy (DCM). However, the mechanisms by which myopathy and DCM develop have not been clearly elucidated. Here, we sought to elucidate these mechanisms using the murine myoblast cell line C2C12 and disease-specific human induced pluripotent stem cells (hiPSCs). Myotubes differentiated from HOIL-1L-KO C2C12 cells exhibited deteriorated differentiation and mitotic cell accumulation. CMs differentiated from patient-derived hiPSCs had an abnormal morphology with a larger size and were excessively multinucleated compared with CMs differentiated from control hiPSCs. Further analysis of hiPSC-derived CMs showed that HOIL-1L deficiency caused cell cycle alteration and mitotic cell accumulation. These results demonstrate that abnormal cell maturation possibly contribute to the development of myopathy and DCM. In conclusion, HOIL-1L is an important intrinsic regulator of cell cycle-related myotube and CM maturation and cell proliferation.
Collapse
Affiliation(s)
- Kentaro Akagi
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Shiro Baba
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan.
| | - Hiroaki Fujita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto City, Kyoto, 606-8501, Japan
| | - Yasuhiro Fuseya
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto City, Kyoto, 606-8501, Japan
| | - Daisuke Yoshinaga
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Hirohito Kubota
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto City, Kyoto, 606-8501, Japan
| | - Eitaro Kume
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Fumiaki Fukumura
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Koichi Matsuda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Takayuki Tanaka
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Takuya Hirata
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto City, Kyoto, 606-8501, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| |
Collapse
|
4
|
Butler D, Reyes DR. Heart-on-a-chip systems: disease modeling and drug screening applications. LAB ON A CHIP 2024; 24:1494-1528. [PMID: 38318723 DOI: 10.1039/d3lc00829k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, casting a substantial economic footprint and burdening the global healthcare system. Historically, pre-clinical CVD modeling and therapeutic screening have been performed using animal models. Unfortunately, animal models oftentimes fail to adequately mimic human physiology, leading to a poor translation of therapeutics from pre-clinical trials to consumers. Even those that make it to market can be removed due to unforeseen side effects. As such, there exists a clinical, technological, and economical need for systems that faithfully capture human (patho)physiology for modeling CVD, assessing cardiotoxicity, and evaluating drug efficacy. Heart-on-a-chip (HoC) systems are a part of the broader organ-on-a-chip paradigm that leverages microfluidics, tissue engineering, microfabrication, electronics, and gene editing to create human-relevant models for studying disease, drug-induced side effects, and therapeutic efficacy. These compact systems can be capable of real-time measurements and on-demand characterization of tissue behavior and could revolutionize the drug development process. In this review, we highlight the key components that comprise a HoC system followed by a review of contemporary reports of their use in disease modeling, drug toxicity and efficacy assessment, and as part of multi-organ-on-a-chip platforms. We also discuss future perspectives and challenges facing the field, including a discussion on the role that standardization is expected to play in accelerating the widespread adoption of these platforms.
Collapse
Affiliation(s)
- Derrick Butler
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| | - Darwin R Reyes
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| |
Collapse
|
5
|
Gregorich ZR, Yanghai Z, Kamp TJ, Granzier H, Guo W. Mechanisms of RBM20 Cardiomyopathy: Insights From Model Systems. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004355. [PMID: 38288598 PMCID: PMC10923161 DOI: 10.1161/circgen.123.004355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2024]
Abstract
RBM20 (RNA-binding motif protein 20) is a vertebrate- and muscle-specific RNA-binding protein that belongs to the serine-arginine-rich family of splicing factors. The RBM20 gene was first identified as a dilated cardiomyopathy-linked gene over a decade ago. Early studies in Rbm20 knockout rodents implicated disrupted splicing of RBM20 target genes as a causative mechanism. Clinical studies show that pathogenic variants in RBM20 are linked to aggressive dilated cardiomyopathy with early onset heart failure and high mortality. Subsequent studies employing pathogenic variant knock-in animal models revealed that variants in a specific portion of the arginine-serine-rich domain in RBM20 not only disrupt splicing but also hinder nucleocytoplasmic transport and lead to the formation of RBM20 biomolecular condensates in the sarcoplasm. Conversely, mice harboring a disease-associated variant in the RRM (RNA recognition motif) do not show evidence of adverse remodeling or exhibit sudden death despite disrupted splicing of RBM20 target genes. Thus, whether disrupted splicing, biomolecular condensates, or both contribute to dilated cardiomyopathy is under debate. Beyond this, additional questions remain, such as whether there is sexual dimorphism in the presentation of RBM20 cardiomyopathy. What are the clinical features of RBM20 cardiomyopathy and why do some individuals develop more severe disease than others? In this review, we summarize the reported observations and discuss potential mechanisms of RBM20 cardiomyopathy derived from studies employing in vivo animal models and in vitro human-induced pluripotent stem cell-derived cardiomyocytes. Potential therapeutic strategies to treat RBM20 cardiomyopathy are also discussed.
Collapse
Affiliation(s)
- Zachery R. Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI
| | - Zhang Yanghai
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI
| | - Timothy J. Kamp
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
6
|
Eberl H, Rebs S, Hoppe S, Sedaghat-Hamedani F, Kayvanpour E, Meder B, Streckfuss-Bömeke K. Generation of an RBM20-mutation-associated left-ventricular non-compaction cardiomyopathy iPSC line (UMGi255-A) into a DCM genetic background to investigate monogenetic cardiomyopathies. Stem Cell Res 2024; 74:103290. [PMID: 38141360 DOI: 10.1016/j.scr.2023.103290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/23/2023] [Accepted: 12/15/2023] [Indexed: 12/25/2023] Open
Abstract
RBM20 mutations account for 3 % of genetic cardiomypathies and manifest with high penetrance and arrhythmogenic effects. Numerous mutations in the conserved RS domain have been described as causing dilated cardiomyopathy (DCM), whereas a particular mutation (p.R634L) drives development of a different cardiac phenotype: left-ventricular non-compaction cardiomyopathy. We generated a mutation-induced pluripotent stem cell (iPSC) line in which the RBM20-LVNC mutation p.R634L was introduced into a DCM patient line with rescued RBM20-p.R634W mutation. These DCM-634L-iPSC can be differentiated into functional cardiomyocytes to test whether this RBM20 mutation induces development of the LVNC phenotype within the genetic context of a DCM patient.
Collapse
Affiliation(s)
- Hanna Eberl
- Institute of Pharmacology and Toxicology, University of Würzburg, Germany
| | - Sabine Rebs
- Institute of Pharmacology and Toxicology, University of Würzburg, Germany; Clinic for Cardiology and Pneumology, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Stefanie Hoppe
- Institute of Pharmacology and Toxicology, University of Würzburg, Germany
| | - Farbod Sedaghat-Hamedani
- Department of Internal Medicine III, University of Heidelberg, and DZHK, Partner Site Heidelberg, Germany
| | - Elham Kayvanpour
- Department of Internal Medicine III, University of Heidelberg, and DZHK, Partner Site Heidelberg, Germany
| | - Benjamin Meder
- Department of Internal Medicine III, University of Heidelberg, and DZHK, Partner Site Heidelberg, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Germany; Clinic for Cardiology and Pneumology, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Germany; Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany.
| |
Collapse
|
7
|
Hawey C, Bourque K, Alim K, Derish I, Rody E, Khan K, Gendron N, Cecere R, Giannetti N, Hébert TE. Measuring Single-Cell Calcium Dynamics Using a Myofilament-Localized Optical Biosensor in hiPSC-CMs Derived from DCM Patients. Cells 2023; 12:2526. [PMID: 37947605 PMCID: PMC10647603 DOI: 10.3390/cells12212526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Synchronized contractions of cardiomyocytes within the heart are tightly coupled to electrical stimulation known as excitation-contraction coupling. Calcium plays a key role in this process and dysregulated calcium handling can significantly impair cardiac function and lead to the development of cardiomyopathies and heart failure. Here, we describe a method and analytical technique to study myofilament-localized calcium signaling using the intensity-based fluorescent biosensor, RGECO-TnT. Dilated cardiomyopathy is a heart muscle disease that negatively impacts the heart's contractile function following dilatation of the left ventricle. We demonstrate how this biosensor can be used to characterize 2D hiPSC-CMs monolayers generated from a healthy control subject compared to two patients diagnosed with dilated cardiomyopathy. Lastly, we provide a step-by-step guide for single-cell data analysis and describe a custom Transient Analysis application, specifically designed to quantify features of calcium transients. All in all, we explain how this analytical approach can be applied to phenotype hiPSC-CM behaviours and stratify patient responses to identify perturbations in calcium signaling.
Collapse
Affiliation(s)
- Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| | - Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| | - Karima Alim
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| | - Ida Derish
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Elise Rody
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Kashif Khan
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Natalie Gendron
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Renzo Cecere
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Nadia Giannetti
- Research Institute, McGull University Hospital Centre, 1001 Decarie Blvd, Montréal, QC H4A 3J1, Canada; (I.D.); (E.R.); (K.K.); (N.G.); (R.C.); (N.G.)
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada; (C.H.); (K.B.); (K.A.)
| |
Collapse
|
8
|
Rebs S, Streckfuss-Bömeke K. How can we use stem cell-derived cardiomyocytes to understand the involvement of energetic metabolism in alterations of cardiac function? FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1222986. [PMID: 39086669 PMCID: PMC11285589 DOI: 10.3389/fmmed.2023.1222986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/15/2023] [Indexed: 08/02/2024]
Abstract
Mutations in the mitochondrial-DNA or mitochondria related nuclear-encoded-DNA lead to various multisystemic disorders collectively termed mitochondrial diseases. One in three cases of mitochondrial disease affects the heart muscle, which is called mitochondrial cardiomyopathy (MCM) and is associated with hypertrophic, dilated, and noncompact cardiomyopathy. The heart is an organ with high energy demand, and mitochondria occupy 30%-40% of its cardiomyocyte-cell volume. Mitochondrial dysfunction leads to energy depletion and has detrimental effects on cardiac performance. However, disease development and progression in the context of mitochondrial and nuclear DNA mutations, remains incompletely understood. The system of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) is an excellent platform to study MCM since the unique genetic identity to their donors enables a robust recapitulation of the predicted phenotypes in a dish on a patient-specific level. Here, we focus on recent insights into MCM studied by patient-specific iPSC-CM and further discuss research gaps and advances in metabolic maturation of iPSC-CM, which is crucial for the study of mitochondrial dysfunction and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Sabine Rebs
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen and DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen and DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
Korover N, Etzion S, Cherniak A, Rabinski T, Levitas A, Etzion Y, Ofir R, Parvari R, Cohen S. Functional defects in hiPSCs-derived cardiomyocytes from patients with a PLEKHM2-mutation associated with dilated cardiomyopathy and left ventricular non-compaction. Biol Res 2023; 56:34. [PMID: 37349842 PMCID: PMC10288792 DOI: 10.1186/s40659-023-00442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/17/2023] [Indexed: 06/24/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a primary myocardial disease, leading to heart failure and excessive risk of sudden cardiac death with rather poorly understood pathophysiology. In 2015, Parvari's group identified a recessive mutation in the autophagy regulator, PLEKHM2 gene, in a family with severe recessive DCM and left ventricular non-compaction (LVNC). Fibroblasts isolated from these patients exhibited abnormal subcellular distribution of endosomes, Golgi apparatus, lysosomes and had impaired autophagy flux. To better understand the effect of mutated PLEKHM2 on cardiac tissue, we generated and characterized induced pluripotent stem cells-derived cardiomyocytes (iPSC-CMs) from two patients and a healthy control from the same family. The patient iPSC-CMs showed low expression levels of genes encoding for contractile functional proteins (α and β-myosin heavy chains and 2v and 2a-myosin light chains), structural proteins integral to heart contraction (Troponin C, T and I) and proteins participating in Ca2+ pumping action (SERCA2 and Calsequestrin 2) compared to their levels in control iPSC-derived CMs. Furthermore, the sarcomeres of the patient iPSC-CMs were less oriented and aligned compared to control cells and generated slowly beating foci with lower intracellular calcium amplitude and abnormal calcium transient kinetics, measured by IonOptix system and MuscleMotion software. Autophagy in patient's iPSC-CMs was impaired as determined from a decrease in the accumulation of autophagosomes in response to chloroquine and rapamycin treatment, compared to control iPSC-CMs. Impairment in autophagy together with the deficiency in the expression of NKX2.5, MHC, MLC, Troponins and CASQ2 genes, which are related to contraction-relaxation coupling and intracellular Ca2+ signaling, may contribute to the defective function of the patient CMs and possibly affect cell maturation and cardiac failure with time.
Collapse
Affiliation(s)
- Nataly Korover
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel.
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Alexander Cherniak
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Tatiana Rabinski
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Aviva Levitas
- Department of Pediatric Cardiology, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Yoram Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Rivka Ofir
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Dead Sea & Arava Science Center, 8691000, Masada, Israel
| | - Ruti Parvari
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Smadar Cohen
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| |
Collapse
|
10
|
Körtl T, Stehle T, Riedl D, Trausel J, Rebs S, Pabel S, Paulus M, Holzamer A, Marrouche N, Maier LS, Sohns C, Streckfuss-Bömeke K, Sossalla S. Atrial Fibrillation Burden Specifically Determines Human Ventricular Cellular Remodeling. JACC Clin Electrophysiol 2022; 8:1357-1366. [PMID: 36424002 DOI: 10.1016/j.jacep.2022.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/27/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) can either be a consequence or an underlying mechanism of left ventricular systolic dysfunction. Patients included in the CASTLE-AF (Catheter Ablation vs. Standard Conventional Treatment in Patients With LV Dysfunction and AF) trial who suffered from AF and left ventricular systolic dysfunction benefited from an AF burden <50% after catheter ablation compared with those patients with an AF burden >50%. OBJECTIVES This analysis tried to explain the clinical findings of the CASTLE-AF trial regarding AF burden in a "back-to-bench" approach. METHODS To study the ventricular effects of different AF burdens, experiments were performed using human ventricular induced pluripotent stem cell-derived cardiomyocytes undergoing in vitro AF simulation. Epifluorescence microscopy, action potential measurements, and measurements of sarcomere regularity were conducted. RESULTS Induced pluripotent stem cell-derived cardiomyocytes stimulated with AF burden of 60% or higher displayed typical hallmarks of heart failure. Ca2+ transient amplitude was significantly reduced indicating negative inotropic effects. Action potential duration was significantly prolonged, which represents a potential trigger for arrhythmias. A significant decrease of sarcomere regularity could explain impaired cardiac contractility in patients with high AF burden. These effects were more pronounced after 7 days of AF simulation compared with 48 hours. CONCLUSIONS Significant functional and structural alterations occurred at the cellular level at a threshold of ∼50% AF burden as it was observed to be harmful in the CASTLE-AF trial. Therefore, these translational results may help to understand the findings of the CASTLE-AF trial.
Collapse
Affiliation(s)
- Thomas Körtl
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Thea Stehle
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Dominic Riedl
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Johanna Trausel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Sabine Rebs
- Clinic for Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany; German Center for Cardiovascular Research, partner site Göttingen, Göttingen, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Würzberg, Germany
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Michael Paulus
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Andreas Holzamer
- Department of Cardiothoracic Surgery, University of Regensburg Medical Center, Regensburg, Germany
| | - Nassir Marrouche
- Tulane Research and Innovation for Arrhythmia Discoveries Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Lars S Maier
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Christian Sohns
- Clinic for Electrophysiology, Herz-und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Zenhaeusern, Germany
| | - Katrin Streckfuss-Bömeke
- Clinic for Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany; German Center for Cardiovascular Research, partner site Göttingen, Göttingen, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Würzberg, Germany
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany; Clinic for Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany; German Center for Cardiovascular Research, partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
11
|
Criscione J, Rezaei Z, Hernandez Cantu CM, Murphy S, Shin SR, Kim DH. Heart-on-a-chip platforms and biosensor integration for disease modeling and phenotypic drug screening. Biosens Bioelectron 2022; 220:114840. [DOI: 10.1016/j.bios.2022.114840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/09/2022] [Accepted: 10/18/2022] [Indexed: 11/02/2022]
|
12
|
Li J, Feng X, Wei X. Modeling hypertrophic cardiomyopathy with human cardiomyocytes derived from induced pluripotent stem cells. Stem Cell Res Ther 2022; 13:232. [PMID: 35659761 PMCID: PMC9166443 DOI: 10.1186/s13287-022-02905-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/18/2022] [Indexed: 12/16/2022] Open
Abstract
One of the obstacles in studying the pathogenesis of hypertrophic cardiomyopathy (HCM) is the poor availability of myocardial tissue samples at the early stages of disease development. This has been addressed by the advent of induced pluripotent stem cells (iPSCs), which allow us to differentiate patient-derived iPSCs into cardiomyocytes (iPSC-CMs) in vitro. In this review, we summarize different approaches to establishing iPSC models and the application of genome editing techniques in iPSC. Because iPSC-CMs cultured at the present stage are immature in structure and function, researchers have attempted several methods to mature iPSC-CMs, such as prolonged culture duration, and mechanical and electrical stimulation. Currently, many researchers have established iPSC-CM models of HCM and employed diverse methods for performing measurements of cellular morphology, contractility, electrophysiological property, calcium handling, mitochondrial function, and metabolism. Here, we review published results in humans to date within the growing field of iPSC-CM models of HCM. Although there is no unified consensus, preliminary results suggest that this approach to modeling disease would provide important insights into our understanding of HCM pathogenesis and facilitate drug development and safety testing.
Collapse
Affiliation(s)
- Jiangtao Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Xin Feng
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
13
|
Malakootian M, Bagheri Moghaddam M, Kalayinia S, Farrashi M, Maleki M, Sadeghipour P, Amin A. Dilated cardiomyopathy caused by a pathogenic nucleotide variant in RBM20 in an Iranian family. BMC Med Genomics 2022; 15:106. [PMID: 35527250 PMCID: PMC9079971 DOI: 10.1186/s12920-022-01262-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/05/2022] [Indexed: 12/22/2022] Open
Abstract
Abstract
Introduction
Dilated cardiomyopathy (DCM) is characterized by the dilation and impaired contraction of 1 or both ventricles and can be caused by a variety of disorders. Up to 50% of idiopathic DCM cases have heritable familial diseases, and the clinical screening of family members is recommended. Identifying a genetic cause that can explain the DCM risk in the family can help with better screening planning and clinical decision-making. Whole-exome sequencing (WES) has aided significantly in the detection of causative genes in many genetically heterogeneous diseases. In the present study, we applied WES to identify the causative genetic variant in a family with heritable DCM.
Methods
WES was applied to identify genetic variants on a 26-year-old man as the proband of a family with DCM. Subsequently, Sanger sequencing was performed to confirm the variant in the patient and all the available affected and unaffected family members. The pathogenicity of the variant was evaluated through co-segregation analysis in the family and employment of in silico predictive software.
Results
WES demonstrated the missense pathogenic heterozygous nucleotide variant, c.1907G > A, (p.Arg636His, rs267607004, NM_0011343), in exon 9 of the RBM20 gene in the proband. The variant was co-segregated in all the affected family members in a heterozygous form and the unaffected family members. The in silico analysis confirmed the variant as pathogenic.
Conclusion
Pathogenic RBM20 nucleotide variants are associated with arrhythmogenic DCM. We believe that our report is the first to show an RBM20 variant in Iranian descent associated with DCM.
Collapse
|
14
|
Tani H, Tohyama S. Human Engineered Heart Tissue Models for Disease Modeling and Drug Discovery. Front Cell Dev Biol 2022; 10:855763. [PMID: 35433691 PMCID: PMC9008275 DOI: 10.3389/fcell.2022.855763] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/08/2022] [Indexed: 12/29/2022] Open
Abstract
The emergence of human induced pluripotent stem cells (hiPSCs) and efficient differentiation of hiPSC-derived cardiomyocytes (hiPSC-CMs) induced from diseased donors have the potential to recapitulate the molecular and functional features of the human heart. Although the immaturity of hiPSC-CMs, including the structure, gene expression, conduct, ion channel density, and Ca2+ kinetics, is a major challenge, various attempts to promote maturation have been effective. Three-dimensional cardiac models using hiPSC-CMs have achieved these functional and morphological maturations, and disease models using patient-specific hiPSC-CMs have furthered our understanding of the underlying mechanisms and effective therapies for diseases. Aside from the mechanisms of diseases and drug responses, hiPSC-CMs also have the potential to evaluate the safety and efficacy of drugs in a human context before a candidate drug enters the market and many phases of clinical trials. In fact, novel drug testing paradigms have suggested that these cells can be used to better predict the proarrhythmic risk of candidate drugs. In this review, we overview the current strategies of human engineered heart tissue models with a focus on major cardiac diseases and discuss perspectives and future directions for the real application of hiPSC-CMs and human engineered heart tissue for disease modeling, drug development, clinical trials, and cardiotoxicity tests.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- *Correspondence: Shugo Tohyama,
| |
Collapse
|
15
|
Haupt LP, Rebs S, Maurer W, Hübscher D, Tiburcy M, Pabel S, Maus A, Köhne S, Tappu R, Haas J, Li Y, Sasse A, Santos CCX, Dressel R, Wojnowski L, Bunt G, Möbius W, Shah AM, Meder B, Wollnik B, Sossalla S, Hasenfuss G, Streckfuss-Bömeke K. Doxorubicin induces cardiotoxicity in a pluripotent stem cell model of aggressive B cell lymphoma cancer patients. Basic Res Cardiol 2022; 117:13. [PMID: 35260914 PMCID: PMC8904375 DOI: 10.1007/s00395-022-00918-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/22/2022] [Accepted: 02/07/2022] [Indexed: 01/31/2023]
Abstract
Cancer therapies with anthracyclines have been shown to induce cardiovascular complications. The aims of this study were to establish an in vitro induced pluripotent stem cell model (iPSC) of anthracycline-induced cardiotoxicity (ACT) from patients with an aggressive form of B-cell lymphoma and to examine whether doxorubicin (DOX)-treated ACT-iPSC cardiomyocytes (CM) can recapitulate the clinical features exhibited by patients, and thus help uncover a DOX-dependent pathomechanism. ACT-iPSC CM generated from individuals with CD20+ B-cell lymphoma who had received high doses of DOX and suffered cardiac dysfunction were studied and compared to control-iPSC CM from cancer survivors without cardiac symptoms. In cellular studies, ACT-iPSC CM were persistently more susceptible to DOX toxicity including augmented disorganized myofilament structure, changed mitochondrial shape, and increased apoptotic events. Consistently, ACT-iPSC CM and cardiac fibroblasts isolated from fibrotic human ACT myocardium exhibited higher DOX-dependent reactive oxygen species. In functional studies, Ca2+ transient amplitude of ACT-iPSC CM was reduced compared to control cells, and diastolic sarcoplasmic reticulum Ca2+ leak was DOX-dependently increased. This could be explained by overactive CaMKIIδ in ACT CM. Together with DOX-dependent augmented proarrhythmic cellular triggers and prolonged action potentials in ACT CM, this suggests a cellular link to arrhythmogenic events and contractile dysfunction especially found in ACT engineered human myocardium. CamKIIδ inhibition prevented proarrhythmic triggers in ACT. In contrast, control CM upregulated SERCA2a expression in a DOX-dependent manner, possibly to avoid heart failure conditions. In conclusion, we developed the first human patient-specific stem cell model of DOX-induced cardiac dysfunction from patients with B-cell lymphoma. Our results suggest that DOX-induced stress resulted in arrhythmogenic events associated with contractile dysfunction and finally in heart failure after persistent stress activation in ACT patients.
Collapse
Affiliation(s)
- Luis Peter Haupt
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Sabine Rebs
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, Würzburg University, Würzburg, Germany
| | - Wiebke Maurer
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Daniela Hübscher
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Malte Tiburcy
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Centre Göttingen, Göttingen, Germany
| | - Steffen Pabel
- Department of Internal Medicine 2, Cardiology, University Medical Centre Regensburg, Regensburg, Germany
| | - Andreas Maus
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,King's College London, British Heart Foundation Centre of Excellence, London, UK
| | - Steffen Köhne
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Rewati Tappu
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centrefor Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany
| | - Jan Haas
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centrefor Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany
| | - Yun Li
- Institute of Human Genetics, University Hospital Centre Göttingen, Göttingen, Germany
| | - Andre Sasse
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| | - Celio C X Santos
- King's College London, British Heart Foundation Centre of Excellence, London, UK
| | - Ralf Dressel
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| | - Leszek Wojnowski
- Department of Pharmacology, University Medical Centre Mainz, Mainz, Germany
| | - Gertrude Bunt
- Clinical Optical Microscopy, University Medical Centre Göttingen, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Electron Microscopy Core Unit, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Ajay M Shah
- King's College London, British Heart Foundation Centre of Excellence, London, UK
| | - Benjamin Meder
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centrefor Cardiovascular Research), partner site Heidelberg, Heidelberg, Germany
| | - Bernd Wollnik
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Human Genetics, University Hospital Centre Göttingen, Göttingen, Germany
| | - Samuel Sossalla
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Internal Medicine 2, Cardiology, University Medical Centre Regensburg, Regensburg, Germany
| | - Gerd Hasenfuss
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- Clinic for Cardiology and Pneumology, University Medical Centre Göttingen, Göttingen, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany. .,Institute of Pharmacology and Toxicology, Würzburg University, Würzburg, Germany.
| |
Collapse
|
16
|
Rebs S, Buchwald TA, Streckfuss-Bömeke K. A quantitative RT-PCR protocol to adapt and quantify RBM20-dependent exon splicing of targets at the human locus. STAR Protoc 2022; 3:101117. [PMID: 35106501 PMCID: PMC8784397 DOI: 10.1016/j.xpro.2021.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gene splicing is a fine-tuned process orchestrated by splice factors including RNA-binding motif 20 (RBM20), and their mutations are linked to the development of cardiac diseases. Here, we provide a step-by-step protocol to transfer RBM20-dependent splicing from rat to human. This protocol describes a PCR-based approach to adapt and quantify RBM20-dependent exon-expression of human target genes. We detail the primer design, the use of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) for RNA isolation, followed by quantification of splicing products. For complete details on the use and execution of this profile, please refer to Streckfuss-Bömeke et al. (2017). Humanized primer usage to detect multiple splicing products by RT-PCR Use of Sanger sequencing to annotate the exons included within a splicing product Detailed description for primer design to quantify specific exon expression by qRT-PCR Use of patient-specific iPSC-CM recapitulating RBM20-based dilated cardiomyopathy
Collapse
|
17
|
Camman M, Joanne P, Agbulut O, Hélary C. 3D models of dilated cardiomyopathy: Shaping the chemical, physical and topographical properties of biomaterials to mimic the cardiac extracellular matrix. Bioact Mater 2022; 7:275-291. [PMID: 34466733 PMCID: PMC8379361 DOI: 10.1016/j.bioactmat.2021.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of dilated cardiomyopathy (DCM), one major cause of heart failure, is characterized by the dilation of the heart but remains poorly understood because of the lack of adequate in vitro models. Current 2D models do not allow for the 3D organotypic organization of cardiomyocytes and do not reproduce the ECM perturbations. In this review, the different strategies to mimic the chemical, physical and topographical properties of the cardiac tissue affected by DCM are presented. The advantages and drawbacks of techniques generating anisotropy required for the cardiomyocytes alignment are discussed. In addition, the different methods creating macroporosity and favoring organotypic organization are compared. Besides, the advances in the induced pluripotent stem cells technology to generate cardiac cells from healthy or DCM patients will be described. Thanks to the biomaterial design, some features of the DCM extracellular matrix such as stiffness, porosity, topography or chemical changes can impact the cardiomyocytes function in vitro and increase their maturation. By mimicking the affected heart, both at the cellular and at the tissue level, 3D models will enable a better understanding of the pathology and favor the discovery of novel therapies.
Collapse
Affiliation(s)
- Marie Camman
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
| |
Collapse
|
18
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
19
|
Radke MH, Badillo-Lisakowski V, Britto-Borges T, Kubli DA, Jüttner R, Parakkat P, Carballo JL, Hüttemeister J, Liss M, Hansen A, Dieterich C, Mullick AE, Gotthardt M. Therapeutic inhibition of RBM20 improves diastolic function in a murine heart failure model and human engineered heart tissue. Sci Transl Med 2021; 13:eabe8952. [PMID: 34851694 DOI: 10.1126/scitranslmed.abe8952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael H Radke
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany
| | - Victor Badillo-Lisakowski
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany.,Humboldt-Universität zu Berlin, 10099 Berlin, Germany
| | - Thiago Britto-Borges
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | | | - René Jüttner
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Pragati Parakkat
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Jacobo Lopez Carballo
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Judith Hüttemeister
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany
| | - Martin Liss
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University Hospital Heidelberg, 69120 Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | | | - Michael Gotthardt
- Department of Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, 10785 Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| |
Collapse
|
20
|
Fenix AM, Miyaoka Y, Bertero A, Blue SM, Spindler MJ, Tan KKB, Perez-Bermejo JA, Chan AH, Mayerl SJ, Nguyen TD, Russell CR, Lizarraga PP, Truong A, So PL, Kulkarni A, Chetal K, Sathe S, Sniadecki NJ, Yeo GW, Murry CE, Conklin BR, Salomonis N. Gain-of-function cardiomyopathic mutations in RBM20 rewire splicing regulation and re-distribute ribonucleoprotein granules within processing bodies. Nat Commun 2021; 12:6324. [PMID: 34732726 PMCID: PMC8566601 DOI: 10.1038/s41467-021-26623-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/22/2021] [Indexed: 12/16/2022] Open
Abstract
Mutations in the cardiac splicing factor RBM20 lead to malignant dilated cardiomyopathy (DCM). To understand the mechanism of RBM20-associated DCM, we engineered isogenic iPSCs with DCM-associated missense mutations in RBM20 as well as RBM20 knockout (KO) iPSCs. iPSC-derived engineered heart tissues made from these cell lines recapitulate contractile dysfunction of RBM20-associated DCM and reveal greater dysfunction with missense mutations than KO. Analysis of RBM20 RNA binding by eCLIP reveals a gain-of-function preference of mutant RBM20 for 3' UTR sequences that are shared with amyotrophic lateral sclerosis (ALS) and processing-body associated RNA binding proteins (FUS, DDX6). Deep RNA sequencing reveals that the RBM20 R636S mutant has unique gene, splicing, polyadenylation and circular RNA defects that differ from RBM20 KO. Super-resolution microscopy verifies that mutant RBM20 maintains very limited nuclear localization potential; rather, the mutant protein associates with cytoplasmic processing bodies (DDX6) under basal conditions, and with stress granules (G3BP1) following acute stress. Taken together, our results highlight a pathogenic mechanism in cardiac disease through splicing-dependent and -independent pathways.
Collapse
Affiliation(s)
- Aidan M Fenix
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, 850 Republican Street, Brotman Building, Seattle, WA, 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Yuichiro Miyaoka
- Regenerative Medicine Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Alessandro Bertero
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, 850 Republican Street, Brotman Building, Seattle, WA, 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
| | - Steven M Blue
- Department of Cellular and Molecular Medicine, Stem Cell Program, and Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Kenneth K B Tan
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | | | - Amanda H Chan
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Steven J Mayerl
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Trieu D Nguyen
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | | | | | - Annie Truong
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Po-Lin So
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Aishwarya Kulkarni
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45221, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Shashank Sathe
- Department of Cellular and Molecular Medicine, Stem Cell Program, and Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nathan J Sniadecki
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, 850 Republican Street, Brotman Building, Seattle, WA, 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, 3720 15th Avenue NE, Seattle, WA, 98105, USA
- Department of Bioengineering, University of Washington, 3720 15th Avenue NE, Seattle, WA, 98105, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Stem Cell Program, and Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Charles E Murry
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
- Center for Cardiovascular Biology, University of Washington, 850 Republican Street, Brotman Building, Seattle, WA, 98109, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA.
- Department of Bioengineering, University of Washington, 3720 15th Avenue NE, Seattle, WA, 98105, USA.
- Department of Medicine/Cardiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
- Sana Biotechnology, 188 E Blaine Street, Seattle, WA, 98102, USA.
| | - Bruce R Conklin
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA.
- Department of Medicine, Cellular and Molecular Pharmacology, and Ophthalmology, University of California San Francisco, San Francisco, CA, 94158, USA.
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, 45229, USA.
| |
Collapse
|
21
|
Loescher CM, Hobbach AJ, Linke WA. Titin (TTN): from molecule to modifications, mechanics and medical significance. Cardiovasc Res 2021; 118:2903-2918. [PMID: 34662387 PMCID: PMC9648829 DOI: 10.1093/cvr/cvab328] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022] Open
Abstract
The giant sarcomere protein titin is a major determinant of cardiomyocyte stiffness and contributor to cardiac strain sensing. Titin-based forces are highly regulated in health and disease, which aids in the regulation of myocardial function, including cardiac filling and output. Due to the enormous size, complexity, and malleability of the titin molecule, titin properties are also vulnerable to dysregulation, as observed in various cardiac disorders. This review provides an overview of how cardiac titin properties can be changed at a molecular level, including the role isoform diversity and post-translational modifications (acetylation, oxidation, and phosphorylation) play in regulating myocardial stiffness and contractility. We then consider how this regulation becomes unbalanced in heart disease, with an emphasis on changes in titin stiffness and protein quality control. In this context, new insights into the key pathomechanisms of human cardiomyopathy due to a truncation in the titin gene (TTN) are discussed. Along the way, we touch on the potential for titin to be therapeutically targeted to treat acquired or inherited cardiac conditions, such as HFpEF or TTN-truncation cardiomyopathy.
Collapse
Affiliation(s)
- Christine M Loescher
- Institute of Physiology II, University Hospital Münster, Robert-Koch-Str. 27B, Münster, 48149 Germany
| | - Anastasia J Hobbach
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, University Hospital Münster, Münster, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, Robert-Koch-Str. 27B, Münster, 48149 Germany
| |
Collapse
|
22
|
Alternative Splicing in Cardiovascular Disease-A Survey of Recent Findings. Genes (Basel) 2021; 12:genes12091457. [PMID: 34573439 PMCID: PMC8469243 DOI: 10.3390/genes12091457] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
Alternative splicing, a driver of posttranscriptional variance, differs from canonical splicing by arranging the introns and exons of an immature pre-mRNA transcript in a multitude of different ways. Although alternative splicing was discovered almost half a century ago, estimates of the proportion of genes that undergo alternative splicing have risen drastically over the last two decades. Deep sequencing methods and novel bioinformatic algorithms have led to new insights into the prevalence of spliced variants, tissue-specific splicing patterns and the significance of alternative splicing in development and disease. Thus far, the role of alternative splicing has been uncovered in areas ranging from heart development, the response to myocardial infarction to cardiac structural disease. Circular RNAs, a product of alternative back-splicing, were initially discovered in 1976, but landmark publications have only recently identified their regulatory role, tissue-specific expression, and transcriptomic abundance, spurring a renewed interest in the topic. The aim of this review is to provide a brief insight into some of the available findings on the role of alternative splicing in cardiovascular disease, with a focus on atherosclerosis, myocardial infarction, heart failure, dilated cardiomyopathy and circular RNAs in myocardial infarction.
Collapse
|
23
|
RBM20-Related Cardiomyopathy: Current Understanding and Future Options. J Clin Med 2021; 10:jcm10184101. [PMID: 34575212 PMCID: PMC8468976 DOI: 10.3390/jcm10184101] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Splice regulators play an essential role in the transcriptomic diversity of all eukaryotic cell types and organ systems. Recent evidence suggests a contribution of splice-regulatory networks in many diseases, such as cardiomyopathies. Adaptive splice regulators, such as RNA-binding motif protein 20 (RBM20) determine the physiological mRNA landscape formation, and rare variants in the RBM20 gene explain up to 6% of genetic dilated cardiomyopathy (DCM) cases. With ample knowledge from RBM20-deficient mice, rats, swine and induced pluripotent stem cells (iPSCs), the downstream targets and quantitative effects on splicing are now well-defined and the prerequisites for corrective therapeutic approaches are set. This review article highlights some of the recent advances in the field, ranging from aspects of granule formation to 3D genome architectures underlying RBM20-related cardiomyopathy. Promising therapeutic strategies are presented and put into context with the pathophysiological characteristics of RBM20-related diseases.
Collapse
|
24
|
Sewanan LR, Park J, Rynkiewicz MJ, Racca AW, Papoutsidakis N, Schwan J, Jacoby DL, Moore JR, Lehman W, Qyang Y, Campbell SG. Loss of crossbridge inhibition drives pathological cardiac hypertrophy in patients harboring the TPM1 E192K mutation. J Gen Physiol 2021; 153:212516. [PMID: 34319370 PMCID: PMC8321830 DOI: 10.1085/jgp.202012640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/14/2021] [Accepted: 07/09/2021] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disorder caused primarily by mutations to thick and thinfilament proteins. Although thin filament mutations are less prevalent than their oft-studied thick filament counterparts, they are frequently associated with severe patient phenotypes and can offer important insight into fundamental disease mechanisms. We have performed a detailed study of tropomyosin (TPM1) E192K, a variant of uncertain significance associated with HCM. Molecular dynamics revealed that E192K results in a more flexible TPM1 molecule, which could affect its ability to regulate crossbridges. In vitro motility assays of regulated actin filaments containing TPM1 E192K showed an overall loss of Ca2+ sensitivity. To understand these effects, we used multiscale computational models that suggested a subtle phenotype in which E192K leads to an inability to completely inhibit actin-myosin crossbridge activity at low Ca2+. To assess the physiological impact of the mutation, we generated patient-derived engineered heart tissues expressing E192K. These tissues showed disease features similar to those of the patients, including cellular hypertrophy, hypercontractility, and diastolic dysfunction. We hypothesized that excess residual crossbridge activity could be triggering cellular hypertrophy, even if the overall Ca2+ sensitivity was reduced by E192K. To test this hypothesis, the cardiac myosin-specific inhibitor mavacamten was applied to patient-derived engineered heart tissues for 4 d followed by 24 h of washout. Chronic mavacamten treatment abolished contractile differences between control and TPM1 E192K engineered heart tissues and reversed hypertrophy in cardiomyocytes. These results suggest that the TPM1 E192K mutation triggers cardiomyocyte hypertrophy by permitting excess residual crossbridge activity. These studies also provide direct evidence that myosin inhibition by mavacamten can counteract the hypertrophic effects of mutant tropomyosin.
Collapse
Affiliation(s)
- Lorenzo R Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Jinkyu Park
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT.,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT
| | - Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA
| | - Alice W Racca
- Department of Biological Sciences, University of Massachusetts, Lowell, MA
| | - Nikolaos Papoutsidakis
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Jonas Schwan
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Daniel L Jacoby
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts, Lowell, MA
| | - William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA
| | - Yibing Qyang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT.,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT.,Department of Pathology, Yale University, New Haven, CT
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
25
|
Giri P, Mukhopadhyay A, Gupta M, Mohapatra B. Dilated cardiomyopathy: a new insight into the rare but common cause of heart failure. Heart Fail Rev 2021; 27:431-454. [PMID: 34245424 DOI: 10.1007/s10741-021-10125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
Heart failure is a global health burden responsible for high morbidity and mortality with a prevalence of greater than 60 million individuals worldwide. One of the major causes of heart failure is dilated cardiomyopathy (DCM), characterized by associated systolic dysfunction. During the last few decades, there have been remarkable advances in our understanding about the genetics of dilated cardiomyopathy. The genetic causes were initially thought to be associated with mutations in genes encoding proteins that are localized to cytoskeleton and sarcomere only; however, with the advancement in mechanistic understanding, the roles of ion channels, Z-disc, mitochondria, nuclear proteins, cardiac transcription factors (e.g., NKX-2.5, TBX20, GATA4), and the factors involved in calcium homeostasis have also been identified and found to be implicated in both familial and sporadic DCM cases. During past few years, next-generation sequencing (NGS) has been established as a diagnostic tool for genetic analysis and it has added significantly to the existing candidate gene list for DCM. The animal models have also provided novel insights to develop a better treatment strategy based on phenotype-genotype correlation, epigenetic and phenomic profiling. Most of the DCM biomarkers that are used in routine genetic and clinical testing are structural proteins, but during the last few years, the role of mi-RNA has also emerged as a biomarker due to their accessibility through noninvasive methods. Our increasing genetic knowledge can improve the clinical management of DCM by bringing clinicians and geneticists on one platform, thereby influencing the individualized clinical decision making and leading to precision medicine.
Collapse
Affiliation(s)
- Prerna Giri
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Amrita Mukhopadhyay
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Mohini Gupta
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India.
| |
Collapse
|
26
|
Gähwiler EKN, Motta SE, Martin M, Nugraha B, Hoerstrup SP, Emmert MY. Human iPSCs and Genome Editing Technologies for Precision Cardiovascular Tissue Engineering. Front Cell Dev Biol 2021; 9:639699. [PMID: 34262897 PMCID: PMC8273765 DOI: 10.3389/fcell.2021.639699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) originate from the reprogramming of adult somatic cells using four Yamanaka transcription factors. Since their discovery, the stem cell (SC) field achieved significant milestones and opened several gateways in the area of disease modeling, drug discovery, and regenerative medicine. In parallel, the emergence of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) revolutionized the field of genome engineering, allowing the generation of genetically modified cell lines and achieving a precise genome recombination or random insertions/deletions, usefully translated for wider applications. Cardiovascular diseases represent a constantly increasing societal concern, with limited understanding of the underlying cellular and molecular mechanisms. The ability of iPSCs to differentiate into multiple cell types combined with CRISPR-Cas9 technology could enable the systematic investigation of pathophysiological mechanisms or drug screening for potential therapeutics. Furthermore, these technologies can provide a cellular platform for cardiovascular tissue engineering (TE) approaches by modulating the expression or inhibition of targeted proteins, thereby creating the possibility to engineer new cell lines and/or fine-tune biomimetic scaffolds. This review will focus on the application of iPSCs, CRISPR-Cas9, and a combination thereof to the field of cardiovascular TE. In particular, the clinical translatability of such technologies will be discussed ranging from disease modeling to drug screening and TE applications.
Collapse
Affiliation(s)
- Eric K. N. Gähwiler
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Marcy Martin
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Bramasta Nugraha
- Molecular Parasitology Lab, Institute of Parasitology, University of Zurich, Zurich, Switzerland
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Wyss Zurich, University and ETH Zurich, Zurich, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| |
Collapse
|
27
|
Briganti F, Sun H, Wei W, Wu J, Zhu C, Liss M, Karakikes I, Rego S, Cipriano A, Snyder M, Meder B, Xu Z, Millat G, Gotthardt M, Mercola M, Steinmetz LM. iPSC Modeling of RBM20-Deficient DCM Identifies Upregulation of RBM20 as a Therapeutic Strategy. Cell Rep 2021; 32:108117. [PMID: 32905764 PMCID: PMC8168789 DOI: 10.1016/j.celrep.2020.108117] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/11/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
Recent advances in induced pluripotent stem cell (iPSC) technology and directed differentiation of iPSCs into cardiomyocytes (iPSC-CMs) make it possible to model genetic heart disease in vitro. We apply CRISPR/Cas9 genome editing technology to introduce three RBM20 mutations in iPSCs and differentiate them into iPSC-CMs to establish an in vitro model of RBM20 mutant dilated cardiomyopathy (DCM). In iPSC-CMs harboring a known causal RBM20 variant, the splicing of RBM20 target genes, calcium handling, and contractility are impaired consistent with the disease manifestation in patients. A variant (Pro633Leu) identified by exome sequencing of patient genomes displays the same disease phenotypes, thus establishing this variant as disease causing. We find that all-trans retinoic acid upregulates RBM20 expression and reverts the splicing, calcium handling, and contractility defects in iPSC-CMs with different causal RBM20 mutations. These results suggest that pharmacological upregulation of RBM20 expression is a promising therapeutic strategy for DCM patients with a heterozygous mutation in RBM20. Briganti et al. use iPSC and CRISPR/Cas9 to create a model of RBM20-deficient dilated cardiomyopathy (DCM) that recapitulates mRNA splicing and contractile defects of the disease. They evaluate pharmacological upregulation of RBM20 as a therapeutic strategy. All-trans retinoic acid upregulates RBM20 expression and ameliorates the in vitro hallmarks of disease.
Collapse
Affiliation(s)
- Francesca Briganti
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany; Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA, USA
| | - Han Sun
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Wu Wei
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA
| | - Jingyan Wu
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Chenchen Zhu
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Martin Liss
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ioannis Karakikes
- Cardiovascular Institute and Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Shannon Rego
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Andrea Cipriano
- Department of Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Michael Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Benjamin Meder
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA; Institute for Cardiomyopathies Heidelberg and Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Zhenyu Xu
- SOPHiA Genetics, St. Sulpice, Switzerland
| | - Gilles Millat
- Laboratoire de Cardiogénétique Moléculaire, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Lyon, France
| | - Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany; Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK: German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA, USA.
| | - Lars M Steinmetz
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA, USA; Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA; DZHK: German Center for Cardiovascular Research, Partner Site EMBL Heidelberg, Heidelberg, Germany.
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW This review aims to give an update on recent findings related to the cardiac splicing factor RNA-binding motif protein 20 (RBM20) and RBM20 cardiomyopathy, a form of dilated cardiomyopathy caused by mutations in RBM20. RECENT FINDINGS While most research on RBM20 splicing targets has focused on titin (TTN), multiple studies over the last years have shown that other splicing targets of RBM20 including Ca2+/calmodulin-dependent kinase IIδ (CAMK2D) might be critically involved in the development of RBM20 cardiomyopathy. In this regard, loss of RBM20 causes an abnormal intracellular calcium handling, which may relate to the arrhythmogenic presentation of RBM20 cardiomyopathy. In addition, RBM20 presents clinically in a highly gender-specific manner, with male patients suffering from an earlier disease onset and a more severe disease progression. Further research on RBM20, and treatment of RBM20 cardiomyopathy, will need to consider both the multitude and relative contribution of the different splicing targets and related pathways, as well as gender differences.
Collapse
|
29
|
Campostrini G, Windt LM, van Meer BJ, Bellin M, Mummery CL. Cardiac Tissues From Stem Cells: New Routes to Maturation and Cardiac Regeneration. Circ Res 2021; 128:775-801. [PMID: 33734815 PMCID: PMC8410091 DOI: 10.1161/circresaha.121.318183] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability of human pluripotent stem cells to form all cells of the body has provided many opportunities to study disease and produce cells that can be used for therapy in regenerative medicine. Even though beating cardiomyocytes were among the first cell types to be differentiated from human pluripotent stem cell, cardiac applications have advanced more slowly than those, for example, for the brain, eye, and pancreas. This is, in part, because simple 2-dimensional human pluripotent stem cell cardiomyocyte cultures appear to need crucial functional cues normally present in the 3-dimensional heart structure. Recent tissue engineering approaches combined with new insights into the dialogue between noncardiomyocytes and cardiomyocytes have addressed and provided solutions to issues such as cardiomyocyte immaturity and inability to recapitulate adult heart values for features like contraction force, electrophysiology, or metabolism. Three-dimensional bioengineered heart tissues are thus poised to contribute significantly to disease modeling, drug discovery, and safety pharmacology, as well as provide new modalities for heart repair. Here, we review the current status of 3-dimensional engineered heart tissues.
Collapse
Affiliation(s)
- Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
| | - Laura M. Windt
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
- MESA+ Institute (B.J.v.M.), University of Twente, Enschede, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
- Department of Biology, University of Padua, Italy (M.B.)
- Veneto Institute of Molecular Medicine, Padua, Padua, Italy (M.B.)
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands (G.C., L.M.W., B.J.v.M., M.B., C.L.M.)
- Department of Applied Stem Cell Technologies (C.L.M.), University of Twente, Enschede, the Netherlands
| |
Collapse
|
30
|
Genetic Cardiomyopathies: The Lesson Learned from hiPSCs. J Clin Med 2021; 10:jcm10051149. [PMID: 33803477 PMCID: PMC7967174 DOI: 10.3390/jcm10051149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Genetic cardiomyopathies represent a wide spectrum of inherited diseases and constitute an important cause of morbidity and mortality among young people, which can manifest with heart failure, arrhythmias, and/or sudden cardiac death. Multiple underlying genetic variants and molecular pathways have been discovered in recent years; however, assessing the pathogenicity of new variants often needs in-depth characterization in order to ascertain a causal role in the disease. The application of human induced pluripotent stem cells has greatly helped to advance our knowledge in this field and enabled to obtain numerous in vitro patient-specific cellular models useful to study the underlying molecular mechanisms and test new therapeutic strategies. A milestone in the research of genetically determined heart disease was the introduction of genomic technologies that provided unparalleled opportunities to explore the genetic architecture of cardiomyopathies, thanks to the generation of isogenic pairs. The aim of this review is to provide an overview of the main research that helped elucidate the pathophysiology of the most common genetic cardiomyopathies: hypertrophic, dilated, arrhythmogenic, and left ventricular noncompaction cardiomyopathies. A special focus is provided on the application of gene-editing techniques in understanding key disease characteristics and on the therapeutic approaches that have been tested.
Collapse
|
31
|
Wang Y, Lei W, Yang J, Ni X, Ye L, Shen Z, Hu S. The updated view on induced pluripotent stem cells for cardiovascular precision medicine. Pflugers Arch 2021; 473:1137-1149. [DOI: 10.1007/s00424-021-02530-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
|
32
|
Tijsen AJ, Cócera Ortega L, Reckman YJ, Zhang X, van der Made I, Aufiero S, Li J, Kamps SC, van den Bout A, Devalla HD, van Spaendonck-Zwarts KY, Engelhardt S, Gepstein L, Ware JS, Pinto YM. Titin Circular RNAs Create a Back-Splice Motif Essential for SRSF10 Splicing. Circulation 2021; 143:1502-1512. [PMID: 33583186 PMCID: PMC8032209 DOI: 10.1161/circulationaha.120.050455] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Supplemental Digital Content is available in the text. Background: TTN (Titin), the largest protein in humans, forms the molecular spring that spans half of the sarcomere to provide passive elasticity to the cardiomyocyte. Mutations that disrupt the TTN transcript are the most frequent cause of hereditary heart failure. We showed before that TTN produces a class of circular RNAs (circRNAs) that depend on RBM20 to be formed. In this study, we show that the back-splice junction formed by this class of circRNAs creates a unique motif that binds SRSF10 to enable it to regulate splicing. Furthermore, we show that one of these circRNAs (cTTN1) distorts both localization of and splicing by RBM20. Methods: We calculated genetic constraint of the identified motif in 125 748 exomes collected from the gnomAD database. Furthermore, we focused on the highest expressed RBM20-dependent circRNA in the human heart, which we named cTTN1. We used shRNAs directed to the back-splice junction to induce selective loss of cTTN1 in human induced pluripotent stem cell–derived cardiomyocytes. Results: Human genetics suggests reduced genetic tolerance of the generated motif, indicating that mutations in this motif might lead to disease. RNA immunoprecipitation confirmed binding of circRNAs with this motif to SRSF10. Selective loss of cTTN1 in human induced pluripotent stem cell–derived cardiomyocytes induced structural abnormalities, apoptosis, and reduced contractile force in engineered heart tissue. In line with its SRSF10 binding, loss of cTTN1 caused abnormal splicing of important cardiomyocyte SRSF10 targets such as MEF2A and CASQ2. Strikingly, loss of cTTN1 also caused abnormal splicing of TTN itself. Mechanistically, we show that loss of cTTN1 distorts both localization of and splicing by RBM20. Conclusions: We demonstrate that circRNAs formed from the TTN transcript are essential for normal splicing of key muscle genes by enabling splice regulators RBM20 and SRSF10. This shows that the TTN transcript also has regulatory roles, besides its well-known signaling and structural function. In addition, we demonstrate that the specific sequence created by the back-splice junction of these circRNAs has important functions. This highlights the existence of functionally important sequences that cannot be recognized as such in the human genome but provides an as-yet unrecognized source for functional sequence variation.
Collapse
Affiliation(s)
- Anke J Tijsen
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Lucía Cócera Ortega
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Yolan J Reckman
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Xiaolei Zhang
- Imperial College London, South Kensington Campus, London, UK (X.Z., J.S.W.)
| | - Ingeborg van der Made
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Simona Aufiero
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Jiuru Li
- Medical Biology, Amsterdam Cardiovascular Sciences (J.L., H.D.D.), Amsterdam, The Netherlands
| | - Selina C Kamps
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Anouk van den Bout
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| | - Harsha D Devalla
- Medical Biology, Amsterdam Cardiovascular Sciences (J.L., H.D.D.), Amsterdam, The Netherlands
| | | | - Stefan Engelhardt
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.E.).,Institut für Pharmakologie und Toxikologie, Technische Universität München, Munich, Germany (S.E.)
| | - Lior Gepstein
- The Sohnis Family Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion-Institute of Technology, Haifa, Israel (L.G.)
| | - James S Ware
- Imperial College London, South Kensington Campus, London, UK (X.Z., J.S.W.)
| | - Yigal M Pinto
- Amsterdam UMC, University of Amsterdam, Departments of Experimental Cardiology, Amsterdam Cardiovascular Sciences (A.J.T., L.C.O., Y.J.R., I.v.d.M., S.A., S.C.K., A.v.d.B., Y.M.P.), Amsterdam, The Netherlands
| |
Collapse
|
33
|
Malignant Arrhythmogenic Role Associated with RBM20: A Comprehensive Interpretation Focused on a Personalized Approach. J Pers Med 2021; 11:jpm11020130. [PMID: 33671899 PMCID: PMC7918949 DOI: 10.3390/jpm11020130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
The RBM20 gene encodes the muscle-specific splicing factor RNA-binding motif 20, a regulator of heart-specific alternative splicing. Nearly 40 potentially deleterious variants in RBM20 have been reported in the last ten years, being found to be associated with highly arrhythmogenic events in familial dilated cardiomyopathy. Frequently, malignant arrhythmias can be a primary manifestation of disease. The early recognition of arrhythmic genotypes is crucial in avoiding lethal episodes, as it may have an impact on the adoption of personalized preventive measures. Our study performs a comprehensive update of data concerning rare variants in RBM20 that are associated with malignant arrhythmogenic phenotypes with a focus on personalized medicine.
Collapse
|
34
|
Microelectrode Arrays: A Valuable Tool to Analyze Stem Cell-Derived Cardiomyocytes. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Show Comparable Functionality to Their Autologous Origin. Cells 2020; 10:cells10010033. [PMID: 33379312 PMCID: PMC7823915 DOI: 10.3390/cells10010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 01/12/2023] Open
Abstract
A multimodal therapeutic approach involving radiotherapy is required when treating head and neck squamous cell carcinoma. However, radiotherapy is restricted due to its high risk for damages to the surrounding healthy tissue of the treated area. Tissue regeneration and wound healing is promoted by the survival and regenerative capacities of tissue-resident or invading stem cells. Mesenchymal stem cells (MSCs) exhibit a promising therapeutic potential in the field of cell-based tissue engineering and regenerative medicine due to their immunomodulatory properties and differentiation capacity. However, the generation of MSCs for therapeutic applications is still a major challenge. We aimed to produce highly homogeneous induced pluripotent stem cell-derived mesenchymal stem cells (iP-MSCs) in an autologous manner from initially isolated human mucosa mesenchymal stem cells (mMSCs) of the upper respiratory tract. Therefore, mMSCs were reprogrammed into induced pluripotent stem cells (iPSCs) by non-integrative chromosomal technologies and differentiated into corresponding iP-MSCs. We demonstrated that mMSCs and iP-MSCs show similar cell characteristics in terms of morphology, clonogenic potential, differentiation, and surface phenotype. Moreover, iP-MSCs demonstrated related immunosuppressive capacity as mMSCs including the secretion of cytokines, and T cell inhibition. Therefore, generating iP-MSCs in an autologous manner may be a novel personalized treatment option in regenerative medicine.
Collapse
|
36
|
Gaertner A, Klauke B, Felski E, Kassner A, Brodehl A, Gerdes D, Stanasiuk C, Ebbinghaus H, Schulz U, Dubowy KO, Tiesmeier J, Laser KT, Bante H, Bergau L, Sommer P, Fox H, Morshuis M, Gummert J, Milting H. Cardiomyopathy-associated mutations in the RS domain affect nuclear localization of RBM20. Hum Mutat 2020; 41:1931-1943. [PMID: 32840935 DOI: 10.1002/humu.24096] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 08/06/2020] [Accepted: 08/19/2020] [Indexed: 12/30/2022]
Abstract
Mutations in RBM20 encoding the RNA-binding motif protein 20 (RBM20) are associated with an early onset and clinically severe forms of cardiomyopathies. Transcriptome analyses revealed RBM20 as an important regulator of cardiac alternative splicing. RBM20 mutations are especially localized in exons 9 and 11 including the highly conserved arginine and serine-rich domain (RS domain). Here, we investigated in several cardiomyopathy patients, the previously described RBM20-mutation p.Pro638Leu localized within the RS domain. In addition, we identified in a patient the novel mutation p.Val914Ala localized in the (glutamate-rich) Glu-rich domain of RBM20 encoded by exon 11. Its impact on the disease was investigated with a novel TTN- and RYR2-splicing assay based on the patients' cardiac messenger RNA. Furthermore, we showed in cell culture and in human cardiac tissue that mutant RBM20-p.Pro638Leu is not localized in the nuclei but causes an abnormal cytoplasmic localization of the protein. In contrast the splicing deficient RBM20-p.Val914Ala has no influence on the intracellular localization. These results indicate that disease-associated variants in RBM20 lead to aberrant splicing through different pathomechanisms dependent on the localization of the mutation. This might have an impact on the future development of therapeutic strategies for the treatment of RBM20-induced cardiomyopathies.
Collapse
Affiliation(s)
- Anna Gaertner
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Baerbel Klauke
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Elina Felski
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Astrid Kassner
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Andreas Brodehl
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Désirée Gerdes
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Caroline Stanasiuk
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Hans Ebbinghaus
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Uwe Schulz
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Karl-Otto Dubowy
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Kinderherzzentrum und Zentrum für Angeborene Herzfehler, Bad Oeynhausen, Germany
| | - Jens Tiesmeier
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Kai-Thorsten Laser
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Kinderherzzentrum und Zentrum für Angeborene Herzfehler, Bad Oeynhausen, Germany
| | - Hendrik Bante
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Elektrophysiologie/Rhythmologie, Bad Oeynhausen, Germany
| | - Leonard Bergau
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Elektrophysiologie/Rhythmologie, Bad Oeynhausen, Germany
| | - Philipp Sommer
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Elektrophysiologie/Rhythmologie, Bad Oeynhausen, Germany
| | - Henrik Fox
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Michiel Morshuis
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Jan Gummert
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| | - Hendrik Milting
- Herz- und Diabeteszentrum NRW, Universitätsklinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Klinik für Thorax- und Kardiovaskularchirurgie, Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Bad Oeynhausen, Germany
| |
Collapse
|
37
|
A Novel Recessive Mutation in SPEG Causes Early Onset Dilated Cardiomyopathy. PLoS Genet 2020; 16:e1009000. [PMID: 32925938 PMCID: PMC7571691 DOI: 10.1371/journal.pgen.1009000] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 10/19/2020] [Accepted: 07/21/2020] [Indexed: 01/05/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common cause of heart failure and sudden cardiac death. It has been estimated that up to half of DCM cases are hereditary. Mutations in more than 50 genes, primarily autosomal dominant, have been reported. Although rare, recessive mutations are thought to contribute considerably to DCM, especially in young children. Here we identified a novel recessive mutation in the striated muscle enriched protein kinase (SPEG, p. E1680K) gene in a family with nonsyndromic, early onset DCM. To ascertain the pathogenicity of this mutation, we generated SPEG E1680K homozygous mutant human induced pluripotent stem cell derived cardiomyocytes (iPSC-CMs) using CRISPR/Cas9-mediated genome editing. Functional studies in mutant iPSC-CMs showed aberrant calcium homeostasis, impaired contractility, and sarcomeric disorganization, recapitulating the hallmarks of DCM. By combining genetic analysis with human iPSCs, genome editing, and functional assays, we identified SPEG E1680K as a novel mutation associated with early onset DCM and provide evidence for its pathogenicity in vitro. Our study provides a conceptual paradigm for establishing genotype-phenotype associations in DCM with autosomal recessive inheritance.
Collapse
|
38
|
Ito M, Nomura S, Morita H, Komuro I. Trends and Limitations in the Assessment of the Contractile Properties of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes From Patients With Dilated Cardiomyopathy. Front Cardiovasc Med 2020; 7:154. [PMID: 33102534 PMCID: PMC7494730 DOI: 10.3389/fcvm.2020.00154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022] Open
Abstract
The application of human induced pluripotent stem cell-derived cardiomyocytes (hiPSCMs) from patients is expected in disease modeling and drug screening in vitro. Dilated cardiomyopathy (DCM) is an intractable disease characterized by the impairment of systolic function and leads to severe heart failure. A number of researchers have focused on disease modeling of DCM and reproduced its pathologic phenotypes in hiPSCMs, but a robust method to evaluate the contractile properties of cardiomyocytes in vitro has not been standardized. In addition, it is unknown whether the throughput of measurements and analyses could be increased sufficiently for compound screening. Here, we reviewed the articles in which the contractile abnormalities of DCM hiPSCMs were recapitulated and assessed the trends and problems in sample preparation and evaluation. We found that single-cell level analysis was ineffective in some cases, and a tissue engineering approach has become dominant recently because of its increased efficiency in reproducing impaired contractility. We also examined two commercially available automated measurement devices with moderate throughput for motion analysis using two-dimensional hiPSCM sheets composed of originally established DCM hiPSCMs. As a result, both of the tested devices, an impedance analyzer and a video image-based cell motion analyzer, were not effective in detecting the expected reduction of contractility in the DCM clone. These findings collectively suggest that a tissue engineering approach could expand the potential of disease modeling with hiPSCMs, and so far, appropriate methods for in vitro force measurement with sufficient throughput, but without sacrificing physiological fidelity, are awaited.
Collapse
Affiliation(s)
- Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
39
|
Modifications of Titin Contribute to the Progression of Cardiomyopathy and Represent a Therapeutic Target for Treatment of Heart Failure. J Clin Med 2020; 9:jcm9092770. [PMID: 32859027 PMCID: PMC7564493 DOI: 10.3390/jcm9092770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Titin is the largest human protein and an essential component of the cardiac sarcomere. With multiple immunoglobulin(Ig)-like domains that serve as molecular springs, titin contributes significantly to the passive tension, systolic function, and diastolic function of the heart. Mutations leading to early termination of titin are the most common genetic cause of dilated cardiomyopathy. Modifications of titin, which change protein length, and relative stiffness affect resting tension of the ventricle and are associated with acquired forms of heart failure. Transcriptional and post-translational changes that increase titin’s length and extensibility, making the sarcomere longer and softer, are associated with systolic dysfunction and left ventricular dilation. Modifications of titin that decrease its length and extensibility, making the sarcomere shorter and stiffer, are associated with diastolic dysfunction in animal models. There has been significant progress in understanding the mechanisms by which titin is modified. As molecular pathways that modify titin’s mechanical properties are elucidated, they represent therapeutic targets for treatment of both systolic and diastolic dysfunction. In this article, we review titin’s contribution to normal cardiac physiology, the pathophysiology of titin truncation variations leading to dilated cardiomyopathy, and transcriptional and post-translational modifications of titin. Emphasis is on how modification of titin can be utilized as a therapeutic target for treatment of heart failure.
Collapse
|
40
|
Rebs S, Sedaghat-Hamedani F, Kayvanpour E, Meder B, Streckfuss-Bömeke K. Generation of pluripotent stem cell lines and CRISPR/Cas9 modified isogenic controls from a patient with dilated cardiomyopathy harboring a RBM20 p.R634W mutation. Stem Cell Res 2020; 47:101901. [PMID: 32674065 DOI: 10.1016/j.scr.2020.101901] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/20/2020] [Accepted: 06/27/2020] [Indexed: 11/16/2022] Open
Abstract
RNA binding motif protein 20 (RBM20) is an alternative splicing factor and highly expressed in cardiac tissue. Mutations in the RS domain of RBM20 have been shown to cause different cardiomyopathies. Here, we generated induced pluripotent stem cells (iPSCs) from a dilated cardiomyopathy patient harboring the heterozygous RBM20 mutation p.R634W and consecutively produced isogenic control line using CRISPR/Cas9 genome editing. Patient-specific RBM20 iPSCs and isogenic control line maintained full pluripotency, genomic integrity, and in vitro differentiation capacity. All iPSC-lines were able to differentiate into pure cardiomyocytes, thus providing a valuable tool for studying the pathogenesis of human RBM20-mediated cardiac disease.
Collapse
Affiliation(s)
- Sabine Rebs
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany; German Center of Cardiovascular Research (DZHK), Germany
| | - Farbod Sedaghat-Hamedani
- German Center of Cardiovascular Research (DZHK), Germany; Department of Cardiology, Angiology and Pneumology, University Medical Center Heidelberg, Germany
| | - Elham Kayvanpour
- German Center of Cardiovascular Research (DZHK), Germany; Department of Cardiology, Angiology and Pneumology, University Medical Center Heidelberg, Germany
| | - Benjamin Meder
- German Center of Cardiovascular Research (DZHK), Germany; Department of Cardiology, Angiology and Pneumology, University Medical Center Heidelberg, Germany; Stanford Genome Technology Center, Department of Genetics, Stanford, CA, USA
| | - Katrin Streckfuss-Bömeke
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany; German Center of Cardiovascular Research (DZHK), Germany.
| |
Collapse
|
41
|
Pioner JM, Fornaro A, Coppini R, Ceschia N, Sacconi L, Donati MA, Favilli S, Poggesi C, Olivotto I, Ferrantini C. Advances in Stem Cell Modeling of Dystrophin-Associated Disease: Implications for the Wider World of Dilated Cardiomyopathy. Front Physiol 2020; 11:368. [PMID: 32477154 PMCID: PMC7235370 DOI: 10.3389/fphys.2020.00368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Familial dilated cardiomyopathy (DCM) is mostly caused by mutations in genes encoding cytoskeletal and sarcomeric proteins. In the pediatric population, DCM is the predominant type of primitive myocardial disease. A severe form of DCM is associated with mutations in the DMD gene encoding dystrophin, which are the cause of Duchenne Muscular Dystrophy (DMD). DMD-associated cardiomyopathy is still poorly understood and orphan of a specific therapy. In the last 5 years, a rise of interest in disease models using human induced pluripotent stem cells (hiPSCs) has led to more than 50 original studies on DCM models. In this review paper, we provide a comprehensive overview on the advances in DMD cardiomyopathy disease modeling and highlight the most remarkable findings obtained from cardiomyocytes differentiated from hiPSCs of DMD patients. We will also describe how hiPSCs based studies have contributed to the identification of specific myocardial disease mechanisms that may be relevant in the pathogenesis of DCM, representing novel potential therapeutic targets.
Collapse
Affiliation(s)
- Josè Manuel Pioner
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | | | - Raffaele Coppini
- Department of NeuroFarBa, Università degli Studi di Firenze, Florence, Italy
| | - Nicole Ceschia
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Sacconi
- LENS, Università degli Studi di Firenze and National Institute of Optics (INO-CNR), Florence, Italy
| | | | - Silvia Favilli
- Pediatric Cardiology, Meyer Children's Hospital, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
42
|
Modeling Cardiovascular Diseases with hiPSC-Derived Cardiomyocytes in 2D and 3D Cultures. Int J Mol Sci 2020; 21:ijms21093404. [PMID: 32403456 PMCID: PMC7246991 DOI: 10.3390/ijms21093404] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the generation of cardiac disease models based on human-induced pluripotent stem cells (hiPSCs) has become of common use, providing new opportunities to overcome the lack of appropriate cardiac models. Although much progress has been made toward the generation of hiPSC-derived cardiomyocytes (hiPS-CMs), several lines of evidence indicate that two-dimensional (2D) cell culturing presents significant limitations, including hiPS-CMs immaturity and the absence of interaction between different cell types and the extracellular matrix. More recently, new advances in bioengineering and co-culture systems have allowed the generation of three-dimensional (3D) constructs based on hiPSC-derived cells. Within these systems, biochemical and physical stimuli influence the maturation of hiPS-CMs, which can show structural and functional properties more similar to those present in adult cardiomyocytes. In this review, we describe the latest advances in 2D- and 3D-hiPSC technology for cardiac disease mechanisms investigation, drug development, and therapeutic studies.
Collapse
|
43
|
Fochi S, Lorenzi P, Galasso M, Stefani C, Trabetti E, Zipeto D, Romanelli MG. The Emerging Role of the RBM20 and PTBP1 Ribonucleoproteins in Heart Development and Cardiovascular Diseases. Genes (Basel) 2020; 11:genes11040402. [PMID: 32276354 PMCID: PMC7230170 DOI: 10.3390/genes11040402] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022] Open
Abstract
Alternative splicing is a regulatory mechanism essential for cell differentiation and tissue organization. More than 90% of human genes are regulated by alternative splicing events, which participate in cell fate determination. The general mechanisms of splicing events are well known, whereas only recently have deep-sequencing, high throughput analyses and animal models provided novel information on the network of functionally coordinated, tissue-specific, alternatively spliced exons. Heart development and cardiac tissue differentiation require thoroughly regulated splicing events. The ribonucleoprotein RBM20 is a key regulator of the alternative splicing events required for functional and structural heart properties, such as the expression of TTN isoforms. Recently, the polypyrimidine tract-binding protein PTBP1 has been demonstrated to participate with RBM20 in regulating splicing events. In this review, we summarize the updated knowledge relative to RBM20 and PTBP1 structure and molecular function; their role in alternative splicing mechanisms involved in the heart development and function; RBM20 mutations associated with idiopathic dilated cardiovascular disease (DCM); and the consequences of RBM20-altered expression or dysfunction. Furthermore, we discuss the possible application of targeting RBM20 in new approaches in heart therapies.
Collapse
|
44
|
Lange S, Pinotsis N, Agarkova I, Ehler E. The M-band: The underestimated part of the sarcomere. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118440. [PMID: 30738787 PMCID: PMC7023976 DOI: 10.1016/j.bbamcr.2019.02.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/16/2019] [Accepted: 02/05/2019] [Indexed: 12/20/2022]
Abstract
The sarcomere is the basic unit of the myofibrils, which mediate skeletal and cardiac Muscle contraction. Two transverse structures, the Z-disc and the M-band, anchor the thin (actin and associated proteins) and thick (myosin and associated proteins) filaments to the elastic filament system composed of titin. A plethora of proteins are known to be integral or associated proteins of the Z-disc and its structural and signalling role in muscle is better understood, while the molecular constituents of the M-band and its function are less well defined. Evidence discussed here suggests that the M-band is important for managing force imbalances during active muscle contraction. Its molecular composition is fine-tuned, especially as far as the structural linkers encoded by members of the myomesin family are concerned and depends on the specific mechanical characteristics of each particular muscle fibre type. Muscle activity signals from the M-band to the nucleus and affects transcription of sarcomeric genes, especially via serum response factor (SRF). Due to its important role as shock absorber in contracting muscle, the M-band is also more and more recognised as a contributor to muscle disease.
Collapse
Affiliation(s)
- Stephan Lange
- Biomedical Research Facility 2, School of Medicine, University of California, San Diego, Medical Sciences Research Bldg, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA; University of Gothenburg, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, UK
| | - Irina Agarkova
- InSphero, Wagistrasse 27, CH-8952 Schlieren, Switzerland
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK; School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Centre, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
45
|
Poetsch MS, Guan K. iPSCs for modeling of sarcomeric cardiomyopathies. RECENT ADVANCES IN IPSC DISEASE MODELING, VOLUME 1 2020:237-273. [DOI: 10.1016/b978-0-12-822227-0.00012-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
46
|
Binnewerg B, Schubert M, Voronkina A, Muzychka L, Wysokowski M, Petrenko I, Djurović M, Kovalchuk V, Tsurkan M, Martinovic R, Bechmann N, Fursov A, Ivanenko VN, Tabachnick KR, Smolii OB, Joseph Y, Giovine M, Bornstein SR, Stelling AL, Tunger A, Schmitz M, Taniya OS, Kovalev IS, Zyryanov GV, Guan K, Ehrlich H. Marine biomaterials: Biomimetic and pharmacological potential of cultivated Aplysina aerophoba marine demosponge. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 109:110566. [PMID: 32228987 DOI: 10.1016/j.msec.2019.110566] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/28/2019] [Accepted: 12/15/2019] [Indexed: 12/31/2022]
Abstract
Marine demosponges of the Verongiida order are considered a gold-mine for bioinspired materials science and marine pharmacology. The aim of this work was to simultaneously isolate selected bromotyrosines and unique chitinous structures from A. aerophoba and to propose these molecules and biomaterials for possible application as antibacterial and antitumor compounds and as ready-to-use scaffolds for cultivation of cardiomyocytes, respectively. Among the extracted bromotyrosines, the attention has been focused on aeroplysinin-1 that showed interesting unexpected growth inhibition properties for some Gram-negative clinical multi-resistant bacterial strains, such as A. baumannii and K. pneumoniae, and on aeroplysinin-1 and on isofistularin-3 for their anti-tumorigenic activity. For both compounds, the effects are cell line dependent, with significant growth inhibition activity on the neuroblastoma cell line SH-SY5Y by aeroplysinin-1 and on breast cancer cell line MCF-7 by isofistularin-3. In this study, we also compared the cultivation of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) on the A. aerophoba chitinous scaffolds, in comparison to chitin structures that were pre-coated with Geltrex™, an extracellular matrix mimetic which is used to enhance iPSC-CM adhesion. The iPSC-CMs on uncoated and pure chitin structures started contracting 24 h after seeding, with comparable behaviour observed on Geltrex-coated cell culture plates, confirming the biocompatibility of the sponge biomaterial with this cell type. The advantage of A. aerophoba is that this source organism does not need to be collected in large quantities to supply the necessary amount for further pre-clinical studies before chemical synthesis of the active compounds will be available. A preliminary analysis of marine sponge bioeconomy as a perspective direction for application of biomaterials and secondary bioactive metabolites has been finally performed for the first time.
Collapse
Affiliation(s)
- Björn Binnewerg
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden 01307, Germany
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden 01307, Germany
| | - Alona Voronkina
- Department of Pharmacy, National Pirogov Memorial Medical University, Vinnytsya 21018, Ukraine
| | - Liubov Muzychka
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, National Academy of Sciences of Ukraine, Kyiv 02094, Ukraine
| | - Marcin Wysokowski
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Poznan 60-965, Poland; Institute of Electronics and Sensor Materials, Technische Universität Bergakademie Freiberg, Freiberg 09599, Germany.
| | - Iaroslav Petrenko
- Institute of Electronics and Sensor Materials, Technische Universität Bergakademie Freiberg, Freiberg 09599, Germany
| | - Mirko Djurović
- Institute of Marine Biology, University of Montenegro, Kotor 85330, Montenegro
| | - Valentine Kovalchuk
- Department of Microbiology, National Pirogov Memorial Medical University, Vinnytsya 21018, Ukraine
| | - Mikhail Tsurkan
- Leibniz Institute of Polymer Research Dresden, Dresden 01069, Germany
| | - Rajko Martinovic
- Institute of Marine Biology, University of Montenegro, Kotor 85330, Montenegro
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Andriy Fursov
- Institute of Electronics and Sensor Materials, Technische Universität Bergakademie Freiberg, Freiberg 09599, Germany
| | - Viatcheslav N Ivanenko
- Department of Invertebrate Zoology, Biological Faculty, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Konstantin R Tabachnick
- P.P. Shirshov Institute of Oceanology, Russian Academy of Sciences, Moscow 117997, Russia; International Institute of Biomineralogy GmbH, Freiberg 09599, Germany
| | - Oleg B Smolii
- V.P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, National Academy of Sciences of Ukraine, Kyiv 02094, Ukraine
| | - Yvonne Joseph
- Institute of Electronics and Sensor Materials, Technische Universität Bergakademie Freiberg, Freiberg 09599, Germany
| | - Marco Giovine
- Department of Sciences of Earth, Environment and Life, University of Genoa, Genova 16132, Italy
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Diabetes and Nutritional Sciences Division, King's College London, London WC2R 2LS, UK
| | - Allison L Stelling
- Duke University Medical Center, Department of Biochemistry, Durham, NC, USA
| | - Antje Tunger
- National Center for Tumor Diseases, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Marc Schmitz
- National Center for Tumor Diseases, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Olga S Taniya
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg 620002, Russia; Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, Yekaterinburg 620219, Russia
| | - Igor S Kovalev
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg 620002, Russia
| | - Grigory V Zyryanov
- Department of Organic and Biomolecular Chemistry, Chemical Engineering Institute, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg 620002, Russia; Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, Yekaterinburg 620219, Russia
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden 01307, Germany.
| | - Hermann Ehrlich
- Institute of Electronics and Sensor Materials, Technische Universität Bergakademie Freiberg, Freiberg 09599, Germany.
| |
Collapse
|
47
|
Tharp CA, Haywood ME, Sbaizero O, Taylor MRG, Mestroni L. The Giant Protein Titin's Role in Cardiomyopathy: Genetic, Transcriptional, and Post-translational Modifications of TTN and Their Contribution to Cardiac Disease. Front Physiol 2019; 10:1436. [PMID: 31849696 PMCID: PMC6892752 DOI: 10.3389/fphys.2019.01436] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, sudden cardiac death and heart transplant. DCM is inherited in approximately 50% of cases, in which the most frequent genetic defects are truncation variants of the titin gene (TTNtv). TTN encodes titin, which is the largest protein in the body and is an essential component of the sarcomere. Titin serves as a biological spring, spanning half of the sarcomere and connecting the Z-disk to the M-line, with scaffold and signaling functions. Truncations of titin are believed to lead to either haploinsufficiency and loss-of-function, or to a “poison peptide” effect. However, other titin mechanisms are postulated to influence cardiac function including post-translational modifications, in particular changes in titin phosphorylation that alters the stiffness of the protein, and diversity of alternative splicing that generates different titin isoforms. In this article, we review the role of TTN mutations in development of DCM, how differential expression of titin isoforms relate to DCM pathophysiology, and discuss how post-translational modifications of titin can affect cardiomyocyte function. Current research efforts aim to elucidate the contribution of titin to myofibril assembly, stability, and signal transduction, and how mutant titin leads to cardiac dysfunction and human disease. Future research will need to translate this knowledge toward novel therapeutic approaches that can modulate titin transcriptional and post-translational defects to treat DCM and heart failure.
Collapse
Affiliation(s)
- Charles A Tharp
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mary E Haywood
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Luisa Mestroni
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
48
|
Kussauer S, David R, Lemcke H. hiPSCs Derived Cardiac Cells for Drug and Toxicity Screening and Disease Modeling: What Micro- Electrode-Array Analyses Can Tell Us. Cells 2019; 8:E1331. [PMID: 31661896 PMCID: PMC6912416 DOI: 10.3390/cells8111331] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) have been intensively used in drug development and disease modeling. Since iPSC-cardiomyocyte (CM) was first generated, their characterization has become a major focus of research. Multi-/micro-electrode array (MEA) systems provide a non-invasive user-friendly platform for detailed electrophysiological analysis of iPSC cardiomyocytes including drug testing to identify potential targets and the assessment of proarrhythmic risk. Here, we provide a systematical overview about the physiological and technical background of micro-electrode array measurements of iPSC-CM. We introduce the similarities and differences between action- and field potential and the advantages and drawbacks of MEA technology. In addition, we present current studies focusing on proarrhythmic side effects of novel and established compounds combining MEA systems and iPSC-CM. MEA technology will help to open a new gateway for novel therapies in cardiovascular diseases while reducing animal experiments at the same time.
Collapse
Affiliation(s)
- Sophie Kussauer
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| | - Robert David
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| | - Heiko Lemcke
- Department Cardiac Surgery, Medical Center, University of Rostock, 18057 Rostock, Germany.
| |
Collapse
|
49
|
Schubert M, Binnewerg B, Voronkina A, Muzychka L, Wysokowski M, Petrenko I, Kovalchuk V, Tsurkan M, Martinovic R, Bechmann N, Ivanenko VN, Fursov A, Smolii OB, Fromont J, Joseph Y, Bornstein SR, Giovine M, Erpenbeck D, Guan K, Ehrlich H. Naturally Prefabricated Marine Biomaterials: Isolation and Applications of Flat Chitinous 3D Scaffolds from Ianthella labyrinthus (Demospongiae: Verongiida). Int J Mol Sci 2019; 20:E5105. [PMID: 31618840 PMCID: PMC6829448 DOI: 10.3390/ijms20205105] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/27/2019] [Accepted: 10/14/2019] [Indexed: 12/16/2022] Open
Abstract
Marine sponges remain representative of a unique source of renewable biological materials. The demosponges of the family Ianthellidae possess chitin-based skeletons with high biomimetic potential. These three-dimensional (3D) constructs can potentially be used in tissue engineering and regenerative medicine. In this study, we focus our attention, for the first time, on the marine sponge Ianthella labyrinthus Bergquist & Kelly-Borges, 1995 (Demospongiae: Verongida: Ianthellidae) as a novel potential source of naturally prestructured bandage-like 3D scaffolds which can be isolated simultaneously with biologically active bromotyrosines. Specifically, translucent and elastic flat chitinous scaffolds have been obtained after bromotyrosine extraction and chemical treatments of the sponge skeleton with alternate alkaline and acidic solutions. For the first time, cardiomyocytes differentiated from human induced pluripotent stem cells (iPSC-CMs) have been used to test the suitability of I. labyrinthus chitinous skeleton as ready-to-use scaffold for their cell culture. Results reveal a comparable attachment and growth on isolated chitin-skeleton, compared to scaffolds coated with extracellular matrix mimetic Geltrex®. Thus, the natural, unmodified I. labyrinthus cleaned sponge skeleton can be used to culture iPSC-CMs and 3D tissue engineering. In addition, I. labyrinthus chitin-based scaffolds demonstrate strong and efficient capability to absorb blood deep into the microtubes due to their excellent capillary effect. These findings are suggestive of the future development of new sponge chitin-based absorbable hemostats as alternatives to already well recognized cellulose-based fabrics.
Collapse
Affiliation(s)
- Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Björn Binnewerg
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Alona Voronkina
- Department of Pharmacy, National Pirogov Memorial Medical University, Vinnytsya, 21018 Vinnytsia, Ukraine.
| | - Lyubov Muzychka
- V.P Kukhar Institute of Bioorganic Chemistry and Petrochemistry, National Academy of Science of Ukraine, Murmanska Str. 1, 02094 Kyiv, Ukraine.
| | - Marcin Wysokowski
- Faculty of Chemical Technology, Institute of Chemical Technology and Engineering, Poznan University of Technology, Berdychowo 4, 60-965 Poznan, Poland.
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner str. 3, 09599 Freiberg, Germany.
| | - Iaroslav Petrenko
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner str. 3, 09599 Freiberg, Germany.
| | - Valentine Kovalchuk
- Department of Microbiology, National Pirogov Memorial Medical University, Vinnytsya, 21018 Vinnytsia, Ukraine.
| | - Mikhail Tsurkan
- Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany.
| | - Rajko Martinovic
- Institute of Marine Biology, University of Montenegro, 85330 Kotor, Montenegro.
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Viatcheslav N Ivanenko
- Department of Invertebrate Zoology, Biological Faculty, Lomonosov Moscow State University, 119992 Moscow, Russia.
| | - Andriy Fursov
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner str. 3, 09599 Freiberg, Germany.
| | - Oleg B Smolii
- V.P Kukhar Institute of Bioorganic Chemistry and Petrochemistry, National Academy of Science of Ukraine, Murmanska Str. 1, 02094 Kyiv, Ukraine.
| | - Jane Fromont
- Aquatic Zoology Department, Western Australian Museum, Locked Bag 49, Welshpool DC, WA 6986, Australia.
| | - Yvonne Joseph
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner str. 3, 09599 Freiberg, Germany.
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- Diabetes and Nutritional Sciences Division, King's College London, London WC2R 2LS, UK.
| | - Marco Giovine
- Department of Sciences of Earth, Environment and Life, University of Genoa, Corso Europa 26, 16132 Genova, Italy.
| | - Dirk Erpenbeck
- Department of Earth and Environmental Sciences & GeoBio-Center, Ludwig-Maximilians-Universität München, Richard-Wagner-Str. 10, 80333 Munich, Germany.
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Hermann Ehrlich
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner str. 3, 09599 Freiberg, Germany.
| |
Collapse
|
50
|
Cresci S, Pereira NL, Ahmad F, Byku M, de las Fuentes L, Lanfear DE, Reilly CM, Owens AT, Wolf MJ. Heart Failure in the Era of Precision Medicine: A Scientific Statement From the American Heart Association. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2019; 12:458-485. [PMID: 31510778 DOI: 10.1161/hcg.0000000000000058] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of 5 people will develop heart failure over his or her lifetime. Early diagnosis and better understanding of the pathophysiology of this disease are critical to optimal treatment. The "omics"-genomics, pharmacogenomics, epigenomics, proteomics, metabolomics, and microbiomics- of heart failure represent rapidly expanding fields of science that have, to date, not been integrated into a single body of work. The goals of this statement are to provide a comprehensive overview of the current state of these omics as they relate to the development and progression of heart failure and to consider the current and potential future applications of these data for precision medicine with respect to prevention, diagnosis, and therapy.
Collapse
|