1
|
Shewale B, Ebrahim T, Samal A, Dubois N. Molecular Regulation of Cardiomyocyte Maturation. Curr Cardiol Rep 2025; 27:32. [PMID: 39836238 DOI: 10.1007/s11886-024-02189-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE OF THE REVIEW This review aims to discuss the process of cardiomyocyte maturation, with a focus on the underlying molecular mechanisms required to form a fully functional heart. We examine both long-standing concepts associated with cardiac maturation and recent developments, and the overall complexity of molecularly integrating all the processes that lead to a mature heart. RECENT FINDINGS Cardiac maturation, defined here as the sequential changes that occurring before the heart reaches full maturity, has been a subject of investigation for decades. Recently, there has been a renewed, highly focused interest in this process, driven by clinically motivated research areas where enhancing maturation may lead to improved therapeutic opportunities. These include using pluripotent stem cell models for cell therapy and disease modeling, as well as recent advancements in adult cardiac regeneration approaches. We highlight key processes underlying maturation of the heart, including cellular and organ growth, and electrophysiological, metabolic, and contractile maturation. We further discuss how these processes integrate and interact to contribute to the overall complexity of the developing heart. Finally, we emphasize the transformative potential for translating relevant maturation concepts to emerging models of heart disease and regeneration.
Collapse
Affiliation(s)
- Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tasneem Ebrahim
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arushi Samal
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
2
|
Verkerk L, Verkerk AO, Wilders R. Zebrafish as a Model System for Brugada Syndrome. Rev Cardiovasc Med 2024; 25:313. [PMID: 39355588 PMCID: PMC11440409 DOI: 10.31083/j.rcm2509313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 10/03/2024] Open
Abstract
Brugada syndrome (BrS) is an inheritable cardiac arrhythmogenic disease, associated with an increased risk of sudden cardiac death. It is most common in males around the age of 40 and the prevalence is higher in Asia than in Europe and the United States. The pathophysiology underlying BrS is not completely understood, but several hypotheses have been proposed. So far, the best effective treatment is the implantation of an implantable cardioverter-defibrillator (ICD), but device-related complications are not uncommon. Therefore, there is an urgent need to improve diagnosis and risk stratification and to find new treatment options. To this end, research should further elucidate the genetic basis and pathophysiological mechanisms of BrS. Several experimental models are being used to gain insight into these aspects. The zebrafish (Danio rerio) is a widely used animal model for the study of cardiac arrhythmias, as its cardiac electrophysiology shows interesting similarities to humans. However, zebrafish have only been used in a limited number of studies on BrS, and the potential role of zebrafish in studying the mechanisms of BrS has not been reviewed. Therefore, the present review aims to evaluate zebrafish as an animal model for BrS. We conclude that zebrafish can be considered as a valuable experimental model for BrS research, not only for gene editing technologies, but also for screening potential BrS drugs.
Collapse
Affiliation(s)
- Leonie Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
3
|
Jiang M, Tang C, Luo X, Zhou X, Chen M, Chi Y, Lai L, Zou Q. Generation of a homozygous RANGRF knockout hiPSC line by CRISPR/Cas9 system. Stem Cell Res 2023; 71:103136. [PMID: 37327621 DOI: 10.1016/j.scr.2023.103136] [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: 02/09/2023] [Revised: 05/19/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023] Open
Abstract
The RAN Guanine Nucleotide Release Factor (RANGRF) gene encodes the protein MOG1, which binds to Nav1.5 and facilitates its transport to the cell membrane. Nav1.5 mutations have been linked to various cardiac arrhythmias and cardiomyopathy. To investigate the role of RANGRF in this process, we utilized the CRISPR/Cas9 gene editing system to generate a homozygous RANGRF knockout hiPSC line. The availability of the cell line will prove to be an invaluable asset in the study of disease mechanisms and the testing of gene therapies for cardiomyopathy.
Collapse
Affiliation(s)
- Meiling Jiang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Chengcheng Tang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Xian Luo
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Xiaoqing Zhou
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Min Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Yue Chi
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Liangxue Lai
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China; China-New Zealand Joint Laboratory on Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou 510530, China; Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya 572000, China.
| | - Qingjian Zou
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Science, Wuyi University, Jiangmen, China.
| |
Collapse
|
4
|
Hu CC, Wei X, Liu JM, Han LL, Xia CK, Wu J, You T, Zhu AF, Yao SL, Yuan SY, Xu HD, Xia ZY, Wang TT, Mao WK. Cardiac-targeted PIASy gene silencing mediates deSUMOylation of caveolin-3 and prevents ischemia/reperfusion-induced Na v1.5 downregulation and ventricular arrhythmias. Mil Med Res 2022; 9:58. [PMID: 36229865 PMCID: PMC9563440 DOI: 10.1186/s40779-022-00415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 09/07/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Abnormal myocardial Nav1.5 expression and function cause lethal ventricular arrhythmias during myocardial ischemia-reperfusion (I/R). Protein inhibitor of activated STAT Y (PIASy)-mediated caveolin-3 (Cav-3) SUMO modification affects Cav-3 binding to the voltage-gated sodium channel 1.5 (Nav1.5). PIASy activity is increased after myocardial I/R, but it is unclear whether this is attributable to plasma membrane Nav1.5 downregulation and ventricular arrhythmias. METHODS Using recombinant adeno-associated virus subtype 9 (AAV9), rat cardiac PIASy was silenced using intraventricular injection of PIASy short hairpin RNA (shRNA). After two weeks, rat hearts were subjected to I/R and electrocardiography was performed to assess malignant arrhythmias. Tissues from peri-infarct areas of the left ventricle were collected for molecular biological measurements. RESULTS PIASy was upregulated by I/R (P < 0.01), with increased SUMO2/3 modification of Cav-3 and reduced membrane Nav1.5 density (P < 0.01). AAV9-PIASy shRNA intraventricular injection into the rat heart downregulated PIASy after I/R, at both mRNA and protein levels (P < 0.05 vs. Scramble-shRNA + I/R group), decreased SUMO-modified Cav-3 levels, enhanced Cav-3 binding to Nav1.5, and prevented I/R-induced decrease of Nav1.5 and Cav-3 co-localization in the intercalated disc and lateral membrane. PIASy silencing in rat hearts reduced I/R-induced fatal arrhythmias, which was reflected by a modest decrease in the duration of ventricular fibrillation (VF; P < 0.05 vs. Scramble-shRNA + I/R group) and a significantly reduced arrhythmia score (P < 0.01 vs. Scramble-shRNA + I/R group). The anti-arrhythmic effects of PIASy silencing were also evidenced by decreased episodes of ventricular tachycardia (VT), sustained VT and VF, especially at the time 5-10 min after ischemia (P < 0.05 vs. Scramble-shRNA + IR group). Using in vitro human embryonic kidney 293 T (HEK293T) cells and isolated adult rat cardiomyocyte models exposed to hypoxia/reoxygenation (H/R), we confirmed that increased PIASy promoted Cav-3 modification by SUMO2/3 and Nav1.5/Cav-3 dissociation after H/R. Mutation of SUMO consensus lysine sites in Cav-3 (K38R or K144R) altered the membrane expression levels of Nav1.5 and Cav-3 before and after H/R in HEK293T cells. CONCLUSIONS I/R-induced cardiac PIASy activation increased Cav-3 SUMOylation by SUMO2/3 and dysregulated Nav1.5-related ventricular arrhythmias. Cardiac-targeted PIASy silencing mediated Cav-3 deSUMOylation and partially prevented I/R-induced Nav1.5 downregulation in the plasma membrane of cardiomyocytes, and subsequent ventricular arrhythmias in rats. PIASy was identified as a potential therapeutic target for life-threatening arrhythmias in patients with ischemic heart diseases.
Collapse
Affiliation(s)
- Chen-Chen Hu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Wei
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jin-Min Liu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin-Lin Han
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng-Kun Xia
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Wu
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao You
- Department of Cardiology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - A-Fang Zhu
- Department of Anesthesiology, Peking Union Medical College Hospital, CAMS and PUMC, Beijing, 100730, China
| | - Shang-Long Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shi-Ying Yuan
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hao-Dong Xu
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Zheng-Yuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, 999077, China.,Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Ting-Ting Wang
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wei-Ke Mao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Zhao M, Han M, Liang L, Song Q, Li X, Du Y, Hu D, Cheng Y, Wang QK, Ke T. Mog1 deficiency promotes cardiac contractile dysfunction and isoproterenol-induced arrhythmias associated with cardiac fibrosis and Cx43 remodeling. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166429. [PMID: 35533905 DOI: 10.1016/j.bbadis.2022.166429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Our earlier studies identified MOG1 as a Nav1.5-binding protein that promotes Nav1.5 intracellular trafficking to plasma membranes. Genetic studies have identified MOG1 variants responsible for cardiac arrhythmias. However, the physiological functions of MOG1 in vivo remain incompletely characterized. In this study, we generated Mog1 knockout (Mog1-/-) mice. Mog1-/- mice did not develop spontaneous arrhythmias at the baseline, but exhibited a prolongation of QRS duration. Mog1-/- mice treated with isoproterenol (ISO), but not with flecainide, exhibited an increased risk of arrhythmias and even sudden death. Mog1-/- mice had normal cardiac morphology, however, LV systolic dysfunction was identified and associated with an increase in ventricular fibrosis. Whole-cell patch-clamping and Western blotting analysis clearly demonstrated the normal cardiac expression and function of Nav1.5 in Mog1-/- mice. Further RNA-seq and iTRAQ analysis identified critical pathways and genes, including extracellular matrix (Mmp2), gap junction (Gja1), and mitochondrial components that were dysregulated in Mog1-/- mice. RT-qPCR, Western blotting, and immunofluorescence assays revealed reduced cardiac expression of Gja1 in Mog1-/- mice. Dye transfer assays confirmed impairment of gap-junction function; Cx43 gap-junction enhancer ZP123 decreased arrhythmia inducibility in ISO-treated Mog1-/- mice. Transmission electron microscopy analysis revealed abnormal sarcomere ultrastructure and altered mitochondrial morphology in Mog1-/- mice. Mitochondrial dynamics was found to be disturbed, and associated with a trend toward increased mitochondrial fusion in Mog1-/- mice. Meanwhile, the level of ATP supply was increased in the hearts of Mog1-/- mice. These results indicate that MOG1 plays an important role in cardiac electrophysiology and cardiac contractile function.
Collapse
Affiliation(s)
- Miao Zhao
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Meng Han
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, Hubei Province, PR China
| | - Lina Liang
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Qixue Song
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055 Shenzhen, PR China
| | - Xia Li
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Yimei Du
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, PR China
| | - Dongping Hu
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Yu Cheng
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Qing K Wang
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China.
| | - Tie Ke
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China.
| |
Collapse
|
6
|
Daimi H, Lozano-Velasco E, Aranega A, Franco D. Genomic and Non-Genomic Regulatory Mechanisms of the Cardiac Sodium Channel in Cardiac Arrhythmias. Int J Mol Sci 2022; 23:1381. [PMID: 35163304 PMCID: PMC8835759 DOI: 10.3390/ijms23031381] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022] Open
Abstract
Nav1.5 is the predominant cardiac sodium channel subtype, encoded by the SCN5A gene, which is involved in the initiation and conduction of action potentials throughout the heart. Along its biosynthesis process, Nav1.5 undergoes strict genomic and non-genomic regulatory and quality control steps that allow only newly synthesized channels to reach their final membrane destination and carry out their electrophysiological role. These regulatory pathways are ensured by distinct interacting proteins that accompany the nascent Nav1.5 protein along with different subcellular organelles. Defects on a large number of these pathways have a tremendous impact on Nav1.5 functionality and are thus intimately linked to cardiac arrhythmias. In the present review, we provide current state-of-the-art information on the molecular events that regulate SCN5A/Nav1.5 and the cardiac channelopathies associated with defects in these pathways.
Collapse
Affiliation(s)
- Houria Daimi
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Estefanía Lozano-Velasco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| |
Collapse
|
7
|
Xiong H, Bai X, Quan Z, Yu D, Zhang H, Zhang C, Liang L, Yao Y, Yang Q, Wang Z, Wang L, Huang Y, Li H, Ren X, Tu X, Ke T, Xu C, Wang QK. Mechanistic insights into the interaction of cardiac sodium channel Na v1.5 with MOG1 and a new molecular mechanism for Brugada syndrome. Heart Rhythm 2021; 19:478-489. [PMID: 34843967 DOI: 10.1016/j.hrthm.2021.11.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/14/2021] [Accepted: 11/20/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Mutations in cardiac sodium channel Nav1.5 cause Brugada syndrome (BrS). MOG1 is a chaperone that binds to Nav1.5, facilitates Nav1.5 trafficking to the cell surface, and enhances the amplitude of sodium current INa. OBJECTIVE The purpose of this study was to identify structural elements involved in MOG1-Nav1.5 interaction and their relevance to the pathogenesis of BrS. METHODS Systematic analyses of large deletions, microdeletions, and point mutations, and glutathione S-transferases pull-down, co-immunoprecipitation, cell surface protein quantification, and patch-clamping of INa were performed. RESULTS Large deletion analysis defined the MOG1-Nav1.5 interaction domain to amino acids S476-H585 of Nav1.5 Loop I connecting transmembrane domains I and II. Microdeletion and point mutation analyses further defined the domain to F530T531F532R533R534R535. Mutations F530A, F532A, R533A, and R534A, but not T531A and R535A, significantly reduced MOG1-Nav1.5 interaction and eliminated MOG1-enhanced INa. Mutagenesis analysis identified D24, E36, D44, E53, and E101A of MOG1 as critical residues for interaction with Nav1.5 Loop I. We then characterized 3 mutations at the MOG1-Nav1.5 interaction domain: p.F530V, p.F532C, and p.R535Q reported from patients with long QT syndrome and BrS. We found that p.F532C reduced MOG1-Nav1.5 interaction and eliminated MOG1 function on INa; p.R535Q is also a loss-of-function mutation that reduces INa amplitude in a MOG1-independent manner, whereas p.F530V is benign as it does not have an apparent effect on MOG1 and INa. CONCLUSION Our findings define the MOG1-Nav1.5 interaction domain to a 5-amino-acid motif of F530T531F532R533R534 in Loop I. Mutation p.F532C associated with BrS abolishes Nav1.5 interaction with MOG1 and reduces MOG1-enhanced INa density, thereby uncovering a novel molecular mechanism for the pathogenesis of BrS.
Collapse
Affiliation(s)
- Hongbo Xiong
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Xuemei Bai
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Zhuang Quan
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Dong Yu
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Hongfu Zhang
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Chi Zhang
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Lina Liang
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Yufeng Yao
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Schoolof Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Zhijie Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Longfei Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, P. R. China
| | - Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, P. R. China
| | - Hui Li
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Xiang Ren
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Xin Tu
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China
| | - Tie Ke
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China.
| | - Chengqi Xu
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China.
| | - Qing K Wang
- Center for Human Genome Research, Department of Biological Sciences, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
8
|
Chatterjee D, Mahabir S, Chatterjee D, Gerlai R. Lasting effects of mild embryonic ethanol exposure on voltage-gated ion channels in adult zebrafish brain. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110327. [PMID: 33864849 DOI: 10.1016/j.pnpbp.2021.110327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/28/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
The zebrafish is increasingly well utilized in alcohol research, particularly in modeling human fetal alcohol spectrum disorders (FASD). FASD results from alcohol reaching the developing fetus intra utero, a completely preventable yet prevalent and devastating life-long disorder. The hope with animal models, including the zebrafish, is to discover the mechanisms underlying this disease, which may aid treatment and diagnosis. In the past, we developed an embryonic alcohol exposure regimen that is aimed at mimicking the milder, and most prevalent, forms of FASD in zebrafish. We have found numerous lasting alterations in behavior, neurochemistry, neuronal markers and glial cell phenotypes in this zebrafish FASD model. Using the same model (2 h long bath immersion of 24 h post-fertilization old zebrafish eggs into 1% vol/vol ethanol), here we conduct a proof of concept analysis of voltage-gated cation channels, investigating potential embryonic alcohol induced changes in L-, T- and N- type Ca++ and the SCN1A Na+ channels using Western blot followed by immunohistochemical analysis of the same channels in the pallium and cerebellum of the zebrafish brain. We report significant reduction of expression in all four channel proteins using both methods. We conclude that reduced voltage-gated cation channel expression induced by short and low dose exposure to alcohol during embryonic development of zebrafish may contribute to the previously demonstrated lasting behavioral and neurobiological changes.
Collapse
Affiliation(s)
| | - Samantha Mahabir
- Department of Psychology, University of Toronto, Mississauga, Ontario, Canada
| | | | - Robert Gerlai
- Department of Psychology, University of Toronto, Mississauga, Ontario, Canada.
| |
Collapse
|
9
|
Gou D, Zhou J, Song Q, Wang Z, Bai X, Zhang Y, Zuo M, Wang F, Chen A, Yousaf M, Yang Z, Peng H, Li K, Xie W, Tang J, Yao Y, Han M, Ke T, Chen Q, Xu C, Wang Q. Mog1 knockout causes cardiac hypertrophy and heart failure by downregulating tbx5-cryab-hspb2 signalling in zebrafish. Acta Physiol (Oxf) 2021; 231:e13567. [PMID: 33032360 DOI: 10.1111/apha.13567] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/09/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
AIMS MOG1 is a small protein that can bind to small GTPase RAN and regulate transport of RNA and proteins between the cytoplasm and nucleus. However, the in vivo physiological role of mog1 in the heart needs to be fully defined. METHODS Mog1 knockout zebrafish was generated by TALEN. Echocardiography, histological analysis, and electrocardiograms were used to examine cardiac structure and function. RNA sequencing and real-time RT-PCR were used to elucidate the molecular mechanism and to analyse the gene expression. Isoproterenol was used to induce cardiac hypertrophy. Whole-mount in situ hybridization was used to observe cardiac morphogenesis. RESULTS Mog1 knockout zebrafish developed cardiac hypertrophy and heart failure (enlarged pericardium, increased nppa and nppb expression and ventricular wall thickness, and reduced ejection fraction), which was aggravated by isoproterenol. RNAseq and KEGG pathway analyses revealed the effect of mog1 knockout on the pathways of cardiac hypertrophy, dilatation and contraction. Mechanistic studies revealed that mog1 knockout decreased expression of tbx5, which reduced expression of cryab and hspb2, resulting in cardiac hypertrophy and heart failure. Overexpression of cryab, hspb2 and tbx5 rescued the cardiac oedema phenotype of mog1 KO zebrafish. Telemetry electrocardiogram monitoring showed QRS and QTc prolongation and a reduced heart rate in mog1 knockout zebrafish, which was associated with reduced scn1b expression. Moreover, mog1 knockout resulted in abnormal cardiac looping during embryogenesis because of the reduced expression of nkx2.5, gata4 and hand2. CONCLUSION Our data identified an important molecular determinant for cardiac hypertrophy and heart failure, and rhythm maintenance of the heart.
Collapse
Affiliation(s)
- Dongzhi Gou
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Juan Zhou
- School of Basic Medicine Gannan Medical University Ganzhou P. R. China
| | - Qixue Song
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Zhijie Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Xuemei Bai
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Yidan Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Mengxia Zuo
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Fan Wang
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH USA
- Department of Molecular Medicine Cleveland Clinic Lerner College of Medicine of CaseWestern Reserve University Cleveland OH USA
| | - Ailan Chen
- Department of Cardiology Guangzhou Medical University Guangzhou P. R. China
| | - Muhammad Yousaf
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Huixing Peng
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Ke Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Wen Xie
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Jingluo Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Meng Han
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Tie Ke
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH USA
- Department of Molecular Medicine Cleveland Clinic Lerner College of Medicine of CaseWestern Reserve University Cleveland OH USA
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education College of Life Science and Technology and Center for Human Genome Research Huazhong University of Science and Technology Wuhan P. R. China
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH USA
- Department of Molecular Medicine Cleveland Clinic Lerner College of Medicine of CaseWestern Reserve University Cleveland OH USA
- Department of Genetics and Genome Science Case Western Reserve University School of Medicine Cleveland OH USA
| |
Collapse
|
10
|
Sieliwonczyk E, Matchkov VV, Vandendriessche B, Alaerts M, Bakkers J, Loeys B, Schepers D. Inherited Ventricular Arrhythmia in Zebrafish: Genetic Models and Phenotyping Tools. Rev Physiol Biochem Pharmacol 2021; 184:33-68. [PMID: 34533615 DOI: 10.1007/112_2021_65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In the last years, the field of inheritable ventricular arrhythmia disease modelling has changed significantly with a push towards the use of novel cellular cardiomyocyte based models. However, there is a growing need for new in vivo models to study the disease pathology at the tissue and organ level. Zebrafish provide an excellent opportunity for in vivo modelling of inheritable ventricular arrhythmia syndromes due to the remarkable similarity between their cardiac electrophysiology and that of humans. Additionally, many state-of-the-art methods in gene editing and electrophysiological phenotyping are available for zebrafish research. In this review, we give a comprehensive overview of the published zebrafish genetic models for primary electrical disorders and arrhythmogenic cardiomyopathy. We summarise and discuss the strengths and weaknesses of the different technical approaches for the generation of genetically modified zebrafish disease models, as well as the electrophysiological approaches in zebrafish phenotyping. By providing this detailed overview, we aim to draw attention to the potential of the zebrafish model for studying arrhythmia syndromes at the organ level and as a platform for personalised medicine and drug testing.
Collapse
Affiliation(s)
- Ewa Sieliwonczyk
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
| | - Vladimir V Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Bert Vandendriessche
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Maaike Alaerts
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Jeroen Bakkers
- Hubrecht Institute for Developmental and Stem Cell Biology, Utrecht, The Netherlands
| | - Bart Loeys
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Dorien Schepers
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.,Laboratory for Molecular, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
Belbachir N, Portero V, Al Sayed ZR, Gourraud JB, Dilasser F, Jesel L, Guo H, Wu H, Gaborit N, Guilluy C, Girardeau A, Bonnaud S, Simonet F, Karakachoff M, Pattier S, Scott C, Burel S, Marionneau C, Chariau C, Gaignerie A, David L, Genin E, Deleuze JF, Dina C, Sauzeau V, Loirand G, Baró I, Schott JJ, Probst V, Wu JC, Redon R, Charpentier F, Le Scouarnec S. RRAD mutation causes electrical and cytoskeletal defects in cardiomyocytes derived from a familial case of Brugada syndrome. Eur Heart J 2020; 40:3081-3094. [PMID: 31114854 DOI: 10.1093/eurheartj/ehz308] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/13/2018] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS The Brugada syndrome (BrS) is an inherited cardiac disorder predisposing to ventricular arrhythmias. Despite considerable efforts, its genetic basis and cellular mechanisms remain largely unknown. The objective of this study was to identify a new susceptibility gene for BrS through familial investigation. METHODS AND RESULTS Whole-exome sequencing performed in a three-generation pedigree with five affected members allowed the identification of one rare non-synonymous substitution (p.R211H) in RRAD, the gene encoding the RAD GTPase, carried by all affected members of the family. Three additional rare missense variants were found in 3/186 unrelated index cases. We detected higher levels of RRAD transcripts in subepicardium than in subendocardium in human heart, and in the right ventricle outflow tract compared to the other cardiac compartments in mice. The p.R211H variant was then subjected to electrophysiological and structural investigations in human cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs). Cardiomyocytes derived from induced pluripotent stem cells from two affected family members exhibited reduced action potential upstroke velocity, prolonged action potentials and increased incidence of early afterdepolarizations, with decreased Na+ peak current amplitude and increased Na+ persistent current amplitude, as well as abnormal distribution of actin and less focal adhesions, compared with intra-familial control iPSC-CMs Insertion of p.R211H-RRAD variant in control iPSCs by genome editing confirmed these results. In addition, iPSC-CMs from affected patients exhibited a decreased L-type Ca2+ current amplitude. CONCLUSION This study identified a potential new BrS-susceptibility gene, RRAD. Cardiomyocytes derived from induced pluripotent stem cells expressing RRAD variant recapitulated single-cell electrophysiological features of BrS, including altered Na+ current, as well as cytoskeleton disturbances.
Collapse
Affiliation(s)
- Nadjet Belbachir
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vincent Portero
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Zeina R Al Sayed
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Jean-Baptiste Gourraud
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Florian Dilasser
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Laurence Jesel
- CHU Strasbourg, Service de Cardiologie, Strasbourg, France
| | - Hongchao Guo
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Haodi Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathalie Gaborit
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | | | - Aurore Girardeau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Stephanie Bonnaud
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Floriane Simonet
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Matilde Karakachoff
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | | | - Carol Scott
- The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Sophie Burel
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Céline Marionneau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Caroline Chariau
- INSERM, CNRS, UNIV Nantes, CHU Nantes, SFR François Bonamy, iPSC core facility, Nantes, France
| | - Anne Gaignerie
- INSERM, CNRS, UNIV Nantes, CHU Nantes, SFR François Bonamy, iPSC core facility, Nantes, France
| | - Laurent David
- INSERM, CNRS, UNIV Nantes, CHU Nantes, SFR François Bonamy, iPSC core facility, Nantes, France.,Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, UNIV Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | | | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Institut de Génomique, CEA, Evry, France
| | - Christian Dina
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Vincent Sauzeau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Gervaise Loirand
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Isabelle Baró
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Jean-Jacques Schott
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Vincent Probst
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Joseph C Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard Redon
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Flavien Charpentier
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Solena Le Scouarnec
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| |
Collapse
|
12
|
Marchal GA, Verkerk AO, Mohan RA, Wolswinkel R, Boukens BJD, Remme CA. The sodium channel Na V 1.5 impacts on early murine embryonic cardiac development, structure and function in a non-electrogenic manner. Acta Physiol (Oxf) 2020; 230:e13493. [PMID: 32386467 PMCID: PMC7539970 DOI: 10.1111/apha.13493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/15/2020] [Accepted: 05/01/2020] [Indexed: 12/19/2022]
Abstract
AIM The voltage-gated sodium channel NaV 1.5, encoded by SCN5A, is essential for cardiac excitability and ensures proper electrical conduction. Early embryonic death has been observed in several murine models carrying homozygous Scn5amutations. We investigated when sodium current (INa ) becomes functionally relevant in the murine embryonic heart and how Scn5a/NaV 1.5 dysfunction impacts on cardiac development. METHODS Involvement of NaV 1.5-generated INa in murine cardiac electrical function was assessed by optical mapping in wild type (WT) embryos (embryonic day (E)9.5 and E10.5) in the absence and presence of the sodium channel blocker tetrodotoxin (30 µmol/L). INa was assessed by patch-clamp analysis in cardiomyocytes isolated from WT embryos (E9.5-17.5). In addition, cardiac morphology and electrical function was assessed in Scn5a-1798insD-/- embryos (E9.5-10.5) and their WT littermates. RESULTS In WT embryos, tetrodotoxin did not affect cardiac activation at E9.5, but slowed activation at E10.5. Accordingly, patch-clamp measurements revealed that INa was virtually absent at E9.5 but robustly present at E10.5. Scn5a-1798insD-/- embryos died in utero around E10.5, displaying severely affected cardiac activation and morphology. Strikingly, altered ventricular activation was observed in Scn5a-1798insD-/- E9.5 embryos before the onset of INa , in addition to reduced cardiac tissue volume compared to WT littermates. CONCLUSION We here demonstrate that NaV 1.5 is involved in cardiac electrical function from E10.5 onwards. Scn5a-1798insD-/- embryos displayed cardiac structural abnormalities at E9.5, indicating that NaV 1.5 dysfunction impacts on embryonic cardiac development in a non-electrogenic manner. These findings are potentially relevant for understanding structural defects observed in relation to NaV 1.5 dysfunction.
Collapse
Affiliation(s)
- Gerard A. Marchal
- Department of Experimental Cardiology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
| | - Arie O. Verkerk
- Department of Experimental Cardiology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
- Department of Medical Biology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
| | - Rajiv A. Mohan
- Department of Experimental Cardiology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
- Department of Medical Biology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
| | - Rianne Wolswinkel
- Department of Experimental Cardiology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
| | - Bastiaan J. D. Boukens
- Department of Medical Biology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology Amsterdam UMC (location Academic Medical Center) Amsterdam the Netherlands
| |
Collapse
|
13
|
Wang X, Li J, Yang Z, Wang L, Li L, Deng W, Zhou J, Wang L, Xu C, Chen Q, Wang QK. phlda3 overexpression impairs specification of hemangioblasts and vascular development. FEBS J 2018; 285:4071-4081. [PMID: 30188605 PMCID: PMC6218282 DOI: 10.1111/febs.14653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 08/20/2018] [Accepted: 09/04/2018] [Indexed: 01/25/2023]
Abstract
The phlda3 gene encodes a small, 127-amino acid protein with only a PH domain, and is involved in tumor suppression, proliferation of islet β-cells, insulin secretion, glucose tolerance, and liver injury. However, the role of phlda3 in vascular development is unknown. Here, we show that phlda3 overexpression decreases the expression levels of hemangioblast markers scl, fli1, and etsrp and intersegmental vessel (ISV) markers flk1 and cdh5, and disrupts ISV development in tg(flk1:GFP) and tg(fli1:GFP) zebrafish. Moreover, phlda3 overexpression inhibits the activation of protein kinase B (AKT) in zebrafish embryos, and the developmental defects of ISVs by phlda3 overexpression were reversed by the expression of a constitutively active form of AKT. These data suggest that phlda3 is a negative regulator of hemangioblast specification and ISV development via AKT signaling.
Collapse
Affiliation(s)
- Xiaojing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Jia Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Li Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Lei Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Wenqing Deng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Juan Zhou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Longfei Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic; Department of Molecular Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic; Department of Molecular Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
14
|
Oliete-Calvo P, Serrano-Quílez J, Nuño-Cabanes C, Pérez-Martínez ME, Soares LM, Dichtl B, Buratowski S, Pérez-Ortín JE, Rodríguez-Navarro S. A role for Mog1 in H2Bub1 and H3K4me3 regulation affecting RNAPII transcription and mRNA export. EMBO Rep 2018; 19:embr.201845992. [PMID: 30249596 DOI: 10.15252/embr.201845992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/28/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022] Open
Abstract
Monoubiquitination of histone H2B (to H2Bub1) is required for downstream events including histone H3 methylation, transcription, and mRNA export. The mechanisms and players regulating these events have not yet been completely delineated. Here, we show that the conserved Ran-binding protein Mog1 is required to sustain normal levels of H2Bub1 and H3K4me3 in Saccharomyces cerevisiae Mog1 is needed for gene body recruitment of Rad6, Bre1, and Rtf1 that are involved in H2B ubiquitination and genetically interacts with these factors. We provide evidence that the absence of MOG1 impacts on cellular processes such as transcription, DNA replication, and mRNA export, which are linked to H2Bub1. Importantly, the mRNA export defect in mog1Δ strains is exacerbated by the absence of factors that decrease H2Bub1 levels. Consistent with a role in sustaining H2Bub and H3K4me3 levels, Mog1 co-precipitates with components that participate in these modifications such as Bre1, Rtf1, and the COMPASS-associated factors Shg1 and Sdc1. These results reveal a novel role for Mog1 in H2B ubiquitination, transcription, and mRNA biogenesis.
Collapse
Affiliation(s)
- Paula Oliete-Calvo
- Gene expression and mRNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Joan Serrano-Quílez
- Gene expression and mRNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.,Gene expression and mRNA Metabolism Laboratory, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - Carme Nuño-Cabanes
- Gene expression and mRNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.,Gene expression and mRNA Metabolism Laboratory, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - María E Pérez-Martínez
- Departamento de Bioquímica y Biología Molecular and E.R.I. Biotecmed, Facultad de Biología, Universitat de València, Burjassot, Spain
| | - Luis M Soares
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Bernhard Dichtl
- School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Centre for Cellular and Molecular Biology, Deakin University, Geelong, Vic., Australia
| | - Stephen Buratowski
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - José E Pérez-Ortín
- Departamento de Bioquímica y Biología Molecular and E.R.I. Biotecmed, Facultad de Biología, Universitat de València, Burjassot, Spain
| | - Susana Rodríguez-Navarro
- Gene expression and mRNA Metabolism Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain .,Gene expression and mRNA Metabolism Laboratory, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| |
Collapse
|
15
|
Wang L, Wang X, Wang L, Yousaf M, Li J, Zuo M, Yang Z, Gou D, Bao B, Li L, Xiang N, Jia H, Xu C, Chen Q, Wang QK. Identification of a new adtrp1-tfpi regulatory axis for the specification of primitive myelopoiesis and definitive hematopoiesis. FASEB J 2017; 32:183-194. [PMID: 28877957 DOI: 10.1096/fj.201700166rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022]
Abstract
A genomic variant in the human ADTRP [androgen-dependent tissue factor (TF) pathway inhibitor (TFPI) regulating protein] gene increases the risk of coronary artery disease, the leading cause of death worldwide. TFPI is the TF pathway inhibitor that is involved in coagulation. Here, we report that adtrp and tfpi form a regulatory axis that specifies primitive myelopoiesis and definitive hematopoiesis, but not primitive erythropoiesis or vasculogenesis. In zebrafish, there are 2 paralogues for adtrp (i.e., adtrp1 and adtrp2). Knockdown of adtrp1 expression inhibits the specification of hemangioblasts, as shown by decreased expression of the hemangioblast markers, etsrp, fli1a, and scl; blocks primitive hematopoiesis, as shown by decreased expression of pu.1, mpo, and l-plastin; and disrupts the specification of hematopoietic stem cells (definitive hematopoiesis), as shown by decreased expression of runx1 and c-myb However, adtrp1 knockdown does not affect erythropoiesis during primitive hematopoiesis (no effect on gata1 or h-bae1) or vasculogenesis (no effect on kdrl, ephb2a, notch3, dab2, or flt4). Knockdown of adtrp2 expression does not have apparent effects on all markers tested. Knockdown of adtrp1 reduced the expression of tfpi, and hematopoietic defects in adtrp1 morphants were rescued by tfpi overexpression. These data suggest that the regulation of tfpi expression is one potential mechanism by which adtrp1 regulates primitive myelopoiesis and definitive hematopoiesis.-Wang, L., Wang, X., Wang, L., Yousaf, M., Li, J., Zuo, M., Yang, Z., Gou, D., Bao, B., Li, L., Xiang, N., Jia, H., Xu, C., Chen, Q., Wang, Q. K. Identification of a new adtrp1-tfpi regulatory axis for the specification of primitive myelopoiesis and definitive hematopoiesis.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Longfei Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Muhammad Yousaf
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxia Zuo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Dongzhi Gou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Binghao Bao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Xiang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haibo Jia
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; .,Department of Molecular Medicine, Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Qing Kenneth Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China; .,Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Up-regulation of miR-95-3p in hepatocellular carcinoma promotes tumorigenesis by targeting p21 expression. Sci Rep 2016; 6:34034. [PMID: 27698442 PMCID: PMC5048429 DOI: 10.1038/srep34034] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/05/2016] [Indexed: 01/16/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant cancers. To elucidate new regulatory mechanisms for heptocarcinogenesis, we investigated the regulation of p21, a cyclin-dependent kinase (CDK) inhibitor encoded by CDKN1A, in HCC. The expression level of p21 is decreased with the progression of HCC. Luciferase assays with a luciferase-p21-3' UTR reporter and its serial deletions identified a 15-bp repressor element at the 3'-UTR of CDKN1A, which contains a binding site for miR-95-3p. Mutation of the binding site eliminated the regulatory effect of miR-95-3p on p21 expression. Posttranscriptional regulation of p21 expression by miR-95-3p is mainly on the protein level (suppression of translation). Overexpression of miR-95-3p in two different HCC cell lines, HepG2 and SMMC7721, significantly promoted cell proliferation, cell cycle progression and cell migration, whereas a miR-95-3p specific inhibitor decreased cell proliferation, cell cycle progression and cell migration. The effects of miR-95-3p on cellular functions were rescued by overexpression of p21. Overexpression of miR-95-3p promoted cell proliferation and tumor growth in HCC xenograft mouse models. Expression of miR-95-3p was significantly higher in HCC samples than in adjacent non-cancerous samples. These results demonstrate that miR-95-3p is a potential new marker for HCC and regulates hepatocarcinogenesis by directly targeting CDKN1A/p21 expression.
Collapse
|