1
|
Lv X, Liu S, Li X, Lv H, Shao K, Luo S, Zhao D, Yan C, Lin P. AAV-based TCAP delivery rescues mitochondria dislocation in limb-girdle muscular dystrophy R7. Brain 2025; 148:1680-1694. [PMID: 39468783 DOI: 10.1093/brain/awae351] [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: 09/23/2023] [Revised: 07/15/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Limb-girdle muscular dystrophy R7 is a rare genetic disease caused by homozygous or compound heterozygous variants in the titin-cap (TCAP) gene that results in the absence of the protein telethonin. The primary pathological features of limb-girdle muscular dystrophy R7 are fibre size variation, nuclear centralization and abnormal mitochondrial distribution. The mechanisms underlying this disease are unclear and there is currently no specific treatment for limb-girdle muscular dystrophy R7. This study established a Tcap-deficient mouse model to explore the disease mechanism of mitochondria dislocation and potential therapeutic strategies. We used methods such as proteomics, immunofluorescence, histopathological staining and western blotting to explore the mechanism of mitochondrial dislocation. Moreover, in the quest for a prospective therapeutic intervention for this disorder, the adeno-associated virus (AAV) serotype 2/9 was employed to deliver the Tcap gene into the muscles of these mice, facilitating preclinical experimentation. After 2 months and 7 months, the muscular phenotype was evaluated and selected mice were humanely euthanized for subsequent molecular and histological analysis. The phenotype of Tcap-/- mice mimicked that observed in individuals diagnosed with limb-girdle muscular dystrophy R7. This study elucidated the mechanism of mitochondrial dislocation in limb-girdle muscular dystrophy R7. Through our in vitro experiments, we discovered that telethonin aids in preserving the integrity of desmin by preventing truncation at the N-terminus. Additionally, telethonin combined with desmin and co-localized at the Z-disc. Research has shown that the Tcap gene plays a crucial role in controlling the desmin cytoskeleton organization. The absence of telethonin leads to a collapsed desmin cytoskeleton. This causes disorganization of the mitochondrial network, leading to mitochondrial dysfunction. In addition, the study investigated the efficacy of AAV-mediated Tcap replacement in Tcap-/- mice. By intramuscular delivery of AAV, we observed dramatic improvements in muscle phenotype, muscle pathology, CK levels, muscle MRI, mitochondrial network organization and mitochondrial function. The results of this study demonstrated that telethonin deficiency led to desmin cytoskeleton collapse that caused mitochondrial dislocation. AAV-mediated replacement therapy could be a promising safe and efficient treatment option for limb-girdle muscular dystrophy R7. The study highlights the potential of AAV-mediated replacement therapy for specific types of limb-girdle muscular dystrophy.
Collapse
Affiliation(s)
- Xiaoqing Lv
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong 250012, China
| | - Shuangwu Liu
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong 250012, China
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, China
| | - Xi Li
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Medical Genetics, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - He Lv
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Kai Shao
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266035, China
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, China
| | - Sushan Luo
- Department of Neurology and Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai 200040, China
| | - Dandan Zhao
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chuanzhu Yan
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong 250012, China
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266000, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266035, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Brain Science Research Institute, Shandong University, Jinan, Shandong 250000, China
| | - Pengfei Lin
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong 250012, China
| |
Collapse
|
2
|
Hu Y, Liu G, Yuan J, Yin D, Lin Y. Mutations in the TCAP gene may lead to restrictive phenotype hypertrophic cardiomyopathy with poor prognosis: case report. Eur Heart J Case Rep 2025; 9:ytaf180. [PMID: 40330574 PMCID: PMC12053251 DOI: 10.1093/ehjcr/ytaf180] [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: 11/03/2024] [Revised: 12/17/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025]
Abstract
Background Genetic disorders are a significant cause of cardiomyopathies. Mutations in the TCAP gene (OMIM #604488) encoding the Z-disc protein Telethonin associated with a mixed phenotype of hypertrophic and restrictive cardiomyopathy with poor prognosis have not yet been reported. Case summary A 47-year-old male presented with heart failure symptoms over a year, which had worsened in the past week. He has a familial history of cardiomyopathy, as his mother was diagnosed with restrictive cardiomyopathy (RCM). Transthoracic echocardiography and cardiac magnetic resonance imaging (CMR) revealed non-obstructive hypertrophic cardiomyopathy (HCM) with severe diastolic dysfunction, biatrial enlargement, preserved ejection fraction, and normal chamber size. Endomyocardial biopsy demonstrated cardiomyocyte hypertrophy and focal fibrosis. The patient was diagnosed with hypertrophic cardiomyopathy with a restrictive phenotype (RP-HCM). Whole exome sequencing identified a frameshift TCAP mutation producing a truncated product (p.Glu12fs) in the family. Despite interventions, the patient's cardiac function progressively deteriorated, leading to his placement on the heart transplant waiting list 1 year later. Discussion In conclusion, we report for the first time that a heterozygous TCAP frameshift mutation resulting in a truncated protein product may contribute to the development of RP-HCM, providing new insights into the genetic basis of cardiomyopathy with mixed phenotypes.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 1017 North Dongmen road, ShenZhen 518020, China
| | - Guangzhong Liu
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 1017 North Dongmen road, ShenZhen 518020, China
- Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen Key Medical Discipline (SZXK003), 1017 North Dongmen road, Shenzhen 518020, Guangdong, China
| | - Jie Yuan
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 1017 North Dongmen road, ShenZhen 518020, China
- Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen Key Medical Discipline (SZXK003), 1017 North Dongmen road, Shenzhen 518020, Guangdong, China
| | - Da Yin
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 1017 North Dongmen road, ShenZhen 518020, China
- Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen Key Medical Discipline (SZXK003), 1017 North Dongmen road, Shenzhen 518020, Guangdong, China
| | - Yaowang Lin
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), 1017 North Dongmen road, ShenZhen 518020, China
- Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen Key Medical Discipline (SZXK003), 1017 North Dongmen road, Shenzhen 518020, Guangdong, China
| |
Collapse
|
3
|
Zhou H, Chen X, Tao Y, Li Z, Wu H, Shi H, Wu X, Huang F. Corticosterone-Induced Myocardial Dysfunctions and the Cardioprotective Role of Tauroursodeoxycholic Acid: An Experimental Study in Mice. Clin Exp Pharmacol Physiol 2025; 52:e70027. [PMID: 39929210 DOI: 10.1111/1440-1681.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 12/02/2024] [Accepted: 01/11/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Major depressive disorder increases cardiovascular risk through stress-induced elevated cortisol levels. Tauroursodeoxycholic acid (TUDCA), a bile acid, has been reported to have anti-inflammatory, anti-depressive and cardioprotective effects. However, the effects of stress-induced myocardial dysfunctions remain unclear. Our study aims to investigate corticosterone-induced myocardial dysfunctions and the role of TUDCA in rescuing such dysfunctions. METHODS To achieve this, experiments were conducted on mice that had been exposed to corticosterone, with treatment involving TUDCA. We first evaluated depression-like behaviours using the open field test, forced swimming test and sucrose preference test, and assessed cardiac function using echocardiography. We then analysed the levels of norepinephrine (NE), adenosine triphosphate (ATP) and B-cell lymphoma-2 (Bcl-2)/Bcl-2 Associated X-protein (Bax) using liquid chromatography-mass spectrometry, enzyme-linked immunosorbent assay and Western blot, respectively. Finally, we investigated gene expression and signalling pathways through RNA-sequencing, which were further validated by qRT-PCR. RESULTS The results demonstrate that corticosterone administration induced depression-like behaviours in mice, including a significant increase in immobility time during the tail suspension test and a significant decrease in the sucrose preference rate. Additionally, it induced cardiac dysfunction in mice, including a decrease in ejection fraction and fractional shortening. Furthermore, corticosterone administration resulted in an increase in left ventricular volume-systolic and left ventricular end-systolic volume index in the mouse left ventricular myocardium. Moreover, it elevated the NE concentration in mouse serum and decreased ATP levels and the Bcl-2/Bax protein expression ratio in the mouse left ventricular tissue. Notably, these detrimental changes were rescued by TUDCA treatment. Additionally, corticosterone affected genes related to cardiac muscle contraction and mitochondrial function, while TUDCA countered this impact by modulating genes associated with muscle processes and ion transport, potentially alleviating myocardial contractile dysfunction. CONCLUSION Overall, our results suggest that corticosterone induces depression-like behaviours, cardiac dysfunction, elevated serum NE levels, reduced ATP and a decreased Bcl-2/Bax ratio, disrupting myocardial contraction and mitochondrial function. TUDCA effectively reversed these effects and modulated genes linked to muscle contraction and ion transport, highlighting its potential in mitigating corticosterone-induced behavioural and cardiac impairments.
Collapse
Affiliation(s)
- Houyuan Zhou
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Xiaoying Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Yanlin Tao
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Zikang Li
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
4
|
Parafati M, Thwin Z, Malany LK, Coen PM, Malany S. Microgravity Accelerates Skeletal Muscle Degeneration: Functional and Transcriptomic Insights from a Muscle Lab-on-Chip Model Onboard the ISS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.634580. [PMID: 39974935 PMCID: PMC11838239 DOI: 10.1101/2025.01.26.634580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Microgravity accelerates skeletal muscle degeneration, mimicking aging, yet its effects on human muscle cell function and signaling remain underexplored. Using a muscle lab-on-chip model onboard the International Space Station, we examined how microgravity and electrically stimulated contractions influence muscle biology and age-related muscle changes. Our 3D bioengineered muscle model, cultured for 21 days (12 days in microgravity), included myobundles from young, active and older, sedentary individuals, with and without electrically stimulated contraction. Real-time data collected within an autonomous Space Tango CubeLab™ showed reduced contraction magnitude in microgravity. Global transcriptomic analysis revealed increased gene expression and particularly mitochondrial-related gene expression in microgravity for the electrically stimulated younger myobundles, while the older myobundles were less responsive. Moreover, a comparative analysis using a skeletal muscle aging gene expression database revealed that certain age-induced genes showed changes in expression in myobundles from the younger cohort when exposed to microgravity, whereas these genes remained unchanged in myobundles from the older cohort. Younger, electrically stimulated myobundles in microgravity exhibited higher expression of 45 aging genes involved in key aging pathways related to inflammation and immune function, mitochondrial dysfunction, and cellular stress; and decreased expression of 41 aging genes associated with inflammation, and cell growth. This study highlights a unique age-related molecular signature in muscle cells exposed to microgravity and underscores electrical stimulation as a potential countermeasure. These insights advance understanding of skeletal muscle aging and microgravity-induced degeneration, informing strategies for mitigating age-related muscle atrophy in space and on Earth.
Collapse
Affiliation(s)
- Maddalena Parafati
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA
| | - Zon Thwin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA
| | | | - Paul M. Coen
- Translational Research Institute, AdventHealth, Orlando, Florida 32804, USA
| | - Siobhan Malany
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|
5
|
Grogan A, Brong A, Joca HC, Boyman L, Kaplan AD, Ward CW, Greiser M, Kontrogianni-Konstantopoulos A. Constitutive deletion of the obscurin-Ig58/59 domains induces atrial remodeling and Ca2+-based arrhythmogenesis. JCI Insight 2025; 10:e184202. [PMID: 39804820 PMCID: PMC11949006 DOI: 10.1172/jci.insight.184202] [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: 06/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Obscurin is a giant protein that coordinates diverse aspects of striated muscle physiology. Obscurin immunoglobulin domains 58/59 (Ig58/59) associate with essential sarcomeric and Ca2+ cycling proteins. To explore the pathophysiological significance of Ig58/59, we generated the Obscn-ΔIg58/59 mouse model, expressing obscurin constitutively lacking Ig58/59. Males in this line develop atrial fibrillation by 6 months, with atrial and ventricular dilation by 12 months. As Obscn-ΔIg58/59 left ventricles at 6 months exhibit no deficits in sarcomeric ultrastructure or Ca2+ signaling, we hypothesized that susceptibility to arrhythmia may emanate from the atria. Ultrastructural evaluation of male Obscn-ΔIg58/59 atria uncovered prominent Z-disk streaming by 6 months and further misalignment by 12 months. Relatedly, isolated Obscn-ΔIg58/59 atrial cardiomyocytes exhibited increased Ca2+ spark frequency and age-specific alterations in Ca2+ cycling dynamics, coinciding with arrhythmia onset and progression. Quantitative analysis of the transverse-axial tubule (TAT) network using super-resolution microscopy demonstrated significant TAT depletion in Obscn-ΔIg58/59 atria. These structural and Ca2+ signaling deficits were accompanied by age-specific alterations in the expression or phosphorylation of T-cap protein, which links transverse tubules to Z-disks, and junctophilin 2, which connects transverse tubules to the sarcoplasmic reticulum. Collectively, our work establishes the Obscn-ΔIg58/59 model as a reputable genetic model for atrial cardiomyopathy and provides mechanistic insights into atrial fibrillation and remodeling.
Collapse
Affiliation(s)
| | - Annie Brong
- Department of Biochemistry and Molecular Biology
| | | | - Liron Boyman
- Department of Physiology and Center for Biomedical Engineering and Technology
- Marlene and Stewart Greenebaum Comprehensive Cancer Center
| | - Aaron D. Kaplan
- Department of Physiology and Center for Biomedical Engineering and Technology
- Division of Cardiology, Department of Medicine; and
| | | | - Maura Greiser
- Department of Physiology and Center for Biomedical Engineering and Technology
- Claude D. Pepper Older Americans Independence Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
6
|
Loh YH, Lv J, Goh Y, Sun X, Zhu X, Muheyati M, Luan Y. Remodelling of T-Tubules and Associated Calcium Handling Dysfunction in Heart Failure: Mechanisms and Therapeutic Insights. Can J Cardiol 2024; 40:2569-2588. [PMID: 39455023 DOI: 10.1016/j.cjca.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
In cardiomyocytes, transverse tubules (T-tubules) are sarcolemmal invaginations that facilitate excitation-contraction coupling and diastolic function. The clinical significance of T-tubules has become evident in that their remodelling is recognised as a hallmark feature of heart failure (HF) and a key contributor to disrupted Ca2+ homeostasis, compromised cardiac function, and arrhythmogenesis. Further investigations have revealed that T-tubule remodelling is particularly pronounced in HF with reduced ejection fraction (HFrEF), but not in HF with preserved ejection fraction, implying that T-tubule remodelling may play a crucial pathophysiologic role in HFrEF. While research on the functional importance of T-tubules is ongoing, T-tubule remodelling has been found to be reversible. That finding has triggered a surge in studies aimed at identifying specific therapeutic approaches for HFrEF. This review discusses the functional importance of T-tubules and their microdomains, the pathophysiology of T-tubule remodelling, and the potential mechanisms of current HFrEF therapeutic approaches in reversing T-tubule alterations. We also highlight discrepancies regarding the roles of T-tubule proteins in the recovery process across studies to offer valuable insights for future research.
Collapse
Affiliation(s)
- Yi Hao Loh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Jingyi Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yenfang Goh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xiangjie Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xianfeng Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Critical Care Medicine, Hangzhou Ninth People's Hospital, China
| | - Muergen Muheyati
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; School of Medicine, Shaoxing University, China.
| |
Collapse
|
7
|
Zhao Y, Liang J, Liu X, Li H, Chang C, Gao P, Du F, Zhang R. Tcap deficiency impedes striated muscle function and heart regeneration with elevated ROS and autophagy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167485. [PMID: 39226992 DOI: 10.1016/j.bbadis.2024.167485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024]
Abstract
Telethonin/titin-cap (TCAP) encodes a Z-disc protein that plays important roles in sarcomere/T-tubule interactions, stretch-sensing and signaling. Mutations in TCAP are associated with muscular dystrophy and cardiomyopathy; however, the complete etiology and its roles in myocardial infarction and regeneration are not fully understood. Here, we generated tcap gene knockout zebrafish with CRISPR/Cas9 technology and observed muscular dystrophy-like phenotypes and abnormal mitochondria in skeletal muscles. The stretch-sensing ability was inhibited in tcap-/- mutants. Moreover, Tcap deficiency led to alterations in cardiac morphology and function as well as increases in reactive oxygen species (ROS) and mitophagy. In addition, the cardiac regeneration and cardiomyocyte proliferation ability of tcap-/- mutants were impaired, but these impairments could be rescued by supplementation with ROS scavengers or autophagy inhibitors. Overall, our study demonstrates the essential roles of Tcap in striated muscle function and heart regeneration. Additionally, elevations in ROS and autophagy may account for the phenotypes resulting from Tcap deficiency and could serve as novel therapeutic targets for muscular dystrophy and cardiomyopathy.
Collapse
Affiliation(s)
- Yan Zhao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China.
| | - Jieling Liang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Xuan Liu
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Huicong Li
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Cheng Chang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Peng Gao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Fen Du
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| | - Ruilin Zhang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
8
|
Ruud M, Frisk M, Melleby AO, Norseng PA, Mohamed BA, Li J, Aronsen JM, Setterberg IE, Jakubiczka J, van Hout I, Coffey S, Shen X, Nygård S, Lunde IG, Tønnessen T, Jones PP, Sjaastad I, Gullestad L, Toischer K, Dahl CP, Christensen G, Louch WE. Regulation of cardiomyocyte t-tubule structure by preload and afterload: Roles in cardiac compensation and decompensation. J Physiol 2024; 602:4487-4510. [PMID: 38686538 DOI: 10.1113/jp284566] [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: 02/22/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Mechanical load is a potent regulator of cardiac structure and function. Although high workload during heart failure is associated with disruption of cardiomyocyte t-tubules and Ca2+ homeostasis, it remains unclear whether changes in preload and afterload may promote adaptive t-tubule remodelling. We examined this issue by first investigating isolated effects of stepwise increases in load in cultured rat papillary muscles. Both preload and afterload increases produced a biphasic response, with the highest t-tubule densities observed at moderate loads, whereas excessively low and high loads resulted in low t-tubule levels. To determine the baseline position of the heart on this bell-shaped curve, mice were subjected to mildly elevated preload or afterload (1 week of aortic shunt or banding). Both interventions resulted in compensated cardiac function linked to increased t-tubule density, consistent with ascension up the rising limb of the curve. Similar t-tubule proliferation was observed in human patients with moderately increased preload or afterload (mitral valve regurgitation, aortic stenosis). T-tubule growth was associated with larger Ca2+ transients, linked to upregulation of L-type Ca2+ channels, Na+-Ca2+ exchanger, mechanosensors and regulators of t-tubule structure. By contrast, marked elevation of cardiac load in rodents and patients advanced the heart down the declining limb of the t-tubule-load relationship. This bell-shaped relationship was lost in the absence of electrical stimulation, indicating a key role of systolic stress in controlling t-tubule plasticity. In conclusion, modest augmentation of workload promotes compensatory increases in t-tubule density and Ca2+ cycling, whereas this adaptation is reversed in overloaded hearts during heart failure progression. KEY POINTS: Excised papillary muscle experiments demonstrated a bell-shaped relationship between cardiomyocyte t-tubule density and workload (preload or afterload), which was only present when muscles were electrically stimulated. The in vivo heart at baseline is positioned on the rising phase of this curve because moderate increases in preload (mice with brief aortic shunt surgery, patients with mitral valve regurgitation) resulted in t-tubule growth. Moderate increases in afterload (mice and patients with mild aortic banding/stenosis) similarly increased t-tubule density. T-tubule proliferation was associated with larger Ca2+ transients, with upregulation of the L-type Ca2+ channel, Na+-Ca2+ exchanger, mechanosensors and regulators of t-tubule structure. By contrast, marked elevation of cardiac load in rodents and patients placed the heart on the declining phase of the t-tubule-load relationship, promoting heart failure progression. The dependence of t-tubule structure on preload and afterload thus enables both compensatory and maladaptive remodelling, in rodents and humans.
Collapse
Affiliation(s)
- Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Joanna Jakubiczka
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Isabelle van Hout
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Sean Coffey
- Department of Medicine and HeartOtago, Dunedin School of Medicine, Dunedin Hospital, Dunedin, New Zealand
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ståle Nygård
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Peter P Jones
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Karl Toischer
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Cristen P Dahl
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Li C, Warren DT, Zhou C, De Silva S, Wilson DGS, Garcia-Maya M, Wheeler MA, Meinke P, Sawyer G, Ehler E, Wehnert M, Rao L, Zhang Q, Shanahan CM. Nesprin-2 is a novel scaffold protein for telethonin and FHL-2 in the cardiomyocyte sarcomere. J Biol Chem 2024; 300:107254. [PMID: 38569934 PMCID: PMC11078644 DOI: 10.1016/j.jbc.2024.107254] [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: 09/05/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.
Collapse
Affiliation(s)
- Chen Li
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK; Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Derek T Warren
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK; School of Pharmacy, University of East Anglia, Norwich, UK
| | - Can Zhou
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Shanelle De Silva
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Darren G S Wilson
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Mitla Garcia-Maya
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Matthew A Wheeler
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Peter Meinke
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Munich, Germany
| | - Greta Sawyer
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK
| | - Elisabeth Ehler
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK; Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Manfred Wehnert
- Institute of Human Genetics, University of Greifswald, Greifswald, Germany
| | - Li Rao
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiuping Zhang
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK.
| | - Catherine M Shanahan
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, UK.
| |
Collapse
|
10
|
Aboonabi A, McCauley MD. Myofilament dysfunction in diastolic heart failure. Heart Fail Rev 2024; 29:79-93. [PMID: 37837495 PMCID: PMC10904515 DOI: 10.1007/s10741-023-10352-z] [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: 09/18/2023] [Indexed: 10/16/2023]
Abstract
Diastolic heart failure (DHF), in which impaired ventricular filling leads to typical heart failure symptoms, represents over 50% of all heart failure cases and is linked with risk factors, including metabolic syndrome, hypertension, diabetes, and aging. A substantial proportion of patients with this disorder maintain normal left ventricular systolic function, as assessed by ejection fraction. Despite the high prevalence of DHF, no effective therapeutic agents are available to treat this condition, partially because the molecular mechanisms of diastolic dysfunction remain poorly understood. As such, by focusing on the underlying molecular and cellular processes contributing to DHF can yield new insights that can represent an exciting new avenue and propose a novel therapeutic approach for DHF treatment. This review discusses new developments from basic and clinical/translational research to highlight current knowledge gaps, help define molecular determinants of diastolic dysfunction, and clarify new targets for treatment.
Collapse
Affiliation(s)
- Anahita Aboonabi
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago, 840 S. Wood St., 920S (MC 715), Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
| | - Mark D McCauley
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago, 840 S. Wood St., 920S (MC 715), Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
11
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
12
|
Cisterna B, Boschi F, Lacavalla MA, Vattemi GNA, Zancanaro C, Malatesta M. Physical training promotes remodeling of the skeletal muscle extracellular matrix: An ultrastructural study in a murine model of Down syndrome. Microsc Res Tech 2023; 86:1517-1528. [PMID: 37381675 DOI: 10.1002/jemt.24379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/30/2023]
Abstract
Down syndrome (DS) is a genetically based disease caused by triplication of chromosome 21. DS is characterized by multi-systemic premature aging associated with deficit in motor coordination, balance, and postural control. Using a morphological, morphometrical, and immunocytochemical ultrastructural approach, this study investigated in vastus lateralis muscle of Ts65Dn mouse, a murine model of DS, the effect of an adapted physical training on the extracellular matrix (ECM) characteristics and whether the forecasted exercise-induced ECM remodeling impacts on sarcomere organization. Morphometry demonstrated thicker basement membrane and larger collagen bundles with larger interfibrillar spacing as well as irregularly arrayed myofibrils and lower telethonin density on Z-lines in trisomic versus euploid sedentary mice. In agreement with the multi-systemic premature aging described in DS, these ECM alterations were similar to those previously observed in skeletal muscle of aged mice. Adapted physical training induced remodeling of ECM in both trisomic and euploid mice, that is, enlargement of the collagen bundles associated with hypertrophy of collagen fibrils and reduction of the interfibrillar spacing. A re-alignment of the myofibrils and a higher telethonin density on Z-line was found in trisomic mice. Altogether, our findings suggest that physical training is an effective tool in limiting/counteracting the trisomy-associated musculoskeletal structural anomalies. The current findings constitute a solid experimental background for further study investigating the possible positive effect of physical training on skeletal muscle performance. RESEARCH HIGHLIGHTS: Vastus lateralis muscle of trisomic mice shows aging-like alterations of extracellular matrix. Training promotes extracellular matrix remodeling. Training may be an effective tool to counteract trisomy-associated alterations of skeletal muscle.
Collapse
Affiliation(s)
- Barbara Cisterna
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Maria Assunta Lacavalla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Carlo Zancanaro
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
13
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Moore J, Ewoldt J, Venturini G, Pereira AC, Padilha K, Lawton M, Lin W, Goel R, Luptak I, Perissi V, Seidman CE, Seidman J, Chin MT, Chen C, Emili A. Multi-Omics Profiling of Hypertrophic Cardiomyopathy Reveals Altered Mechanisms in Mitochondrial Dynamics and Excitation-Contraction Coupling. Int J Mol Sci 2023; 24:4724. [PMID: 36902152 PMCID: PMC10002553 DOI: 10.3390/ijms24054724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Hypertrophic cardiomyopathy is one of the most common inherited cardiomyopathies and a leading cause of sudden cardiac death in young adults. Despite profound insights into the genetics, there is imperfect correlation between mutation and clinical prognosis, suggesting complex molecular cascades driving pathogenesis. To investigate this, we performed an integrated quantitative multi-omics (proteomic, phosphoproteomic, and metabolomic) analysis to illuminate the early and direct consequences of mutations in myosin heavy chain in engineered human induced pluripotent stem-cell-derived cardiomyocytes relative to late-stage disease using patient myectomies. We captured hundreds of differential features, which map to distinct molecular mechanisms modulating mitochondrial homeostasis at the earliest stages of pathobiology, as well as stage-specific metabolic and excitation-coupling maladaptation. Collectively, this study fills in gaps from previous studies by expanding knowledge of the initial responses to mutations that protect cells against the early stress prior to contractile dysfunction and overt disease.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jourdan Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
| | | | | | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Sao Paulo 05508-000, Brazil
| | - Matthew Lawton
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Weiwei Lin
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Raghuveera Goel
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ivan Luptak
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA 02118, USA
| | - Valentina Perissi
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02145, USA
| | - Christopher Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andrew Emili
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
15
|
Yoo SH. Circadian regulation of cardiac muscle function and protein degradation. Chronobiol Int 2023; 40:4-12. [PMID: 34521283 PMCID: PMC8918439 DOI: 10.1080/07420528.2021.1957911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
The circadian clock plays a fundamental role in physiology. In particular, the heart is a target organ where the clock orchestrates various aspects of cardiac function. At the molecular level, the clock machinery governs daily rhythms of gene expression. Such circadian regulation is in tune with the dynamic nature of heart structure and function, and provides the foundation for chronotherapeutic applications in cardiovascular diseases. In comparison, a regulatory role of the clock in cardiac protein degradation is poorly documented. Sarcomere is the structural and functional unit responsible for cardiac muscle contraction, and sarcomere components are closely regulated by protein folding and proteolysis. Emerging evidence supports a role of the circadian clock in governing sarcomere integrity and function. Particularly, recent studies uncovered a circadian regulation of a core sarcomere component TCAP. It is possible that circadian regulation of the cardiac muscle protein turnover is a key regulatory mechanism underlying cardiac remodeling in response to physiological and environmental stimuli. While the detailed regulatory mechanisms and the molecular links to cardiac (patho)physiology remain to be further studied, therapeutic strategies targeting circadian control in the heart may markedly enhance intervention outcomes against cardiovascular disease.
Collapse
Affiliation(s)
- Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
16
|
Tanboon J, Nishino I. Autosomal Recessive Limb-Girdle Muscular Dystrophies. CURRENT CLINICAL NEUROLOGY 2023:93-121. [DOI: 10.1007/978-3-031-44009-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
17
|
Lim JY, Kim E, Douglas CM, Wirianto M, Han C, Ono K, Kim SY, Ji JH, Tran CK, Chen Z, Esser KA, Yoo SH. The circadian E3 ligase FBXL21 regulates myoblast differentiation and sarcomere architecture via MYOZ1 ubiquitination and NFAT signaling. PLoS Genet 2022; 18:e1010574. [PMID: 36574402 PMCID: PMC9829178 DOI: 10.1371/journal.pgen.1010574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/09/2023] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Numerous molecular and physiological processes in the skeletal muscle undergo circadian time-dependent oscillations in accordance with daily activity/rest cycles. The circadian regulatory mechanisms underlying these cyclic processes, especially at the post-transcriptional level, are not well defined. Previously, we reported that the circadian E3 ligase FBXL21 mediates rhythmic degradation of the sarcomere protein TCAP in conjunction with GSK-3β, and Psttm mice harboring an Fbxl21 hypomorph allele show reduced muscle fiber diameter and impaired muscle function. To further elucidate the regulatory function of FBXL21 in skeletal muscle, we investigated another sarcomere protein, Myozenin1 (MYOZ1), that we identified as an FBXL21-binding protein from yeast 2-hybrid screening. We show that FBXL21 binding to MYOZ1 led to ubiquitination-mediated proteasomal degradation. GSK-3β co-expression and inhibition were found to accelerate and decelerate FBXL21-mediated MYOZ1 degradation, respectively. Previously, MYOZ1 has been shown to inhibit calcineurin/NFAT signaling important for muscle differentiation. In accordance, Fbxl21 KO and MyoZ1 KO in C2C12 cells impaired and enhanced myogenic differentiation respectively compared with control C2C12 cells, concomitant with distinct effects on NFAT nuclear localization and NFAT target gene expression. Importantly, in Psttm mice, both the levels and diurnal rhythm of NFAT2 nuclear localization were significantly diminished relative to wild-type mice, and circadian expression of NFAT target genes associated with muscle differentiation was also markedly dampened. Furthermore, Psttm mice exhibited significant disruption of sarcomere structure with a considerable excess of MYOZ1 accumulation in the Z-line. Taken together, our study illustrates a pivotal role of FBXL21 in sarcomere structure and muscle differentiation by regulating MYOZ1 degradation and NFAT2 signaling.
Collapse
Affiliation(s)
- Ji Ye Lim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Collin M. Douglas
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Chorong Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kaori Ono
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Sun Young Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Justin H. Ji
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Celia K. Tran
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Karyn A. Esser
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
18
|
Müller D, Donath S, Brückner EG, Biswanath Devadas S, Daniel F, Gentemann L, Zweigerdt R, Heisterkamp A, Kalies SMK. How Localized Z-Disc Damage Affects Force Generation and Gene Expression in Cardiomyocytes. Bioengineering (Basel) 2021; 8:bioengineering8120213. [PMID: 34940366 PMCID: PMC8698600 DOI: 10.3390/bioengineering8120213] [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: 10/22/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 11/24/2022] Open
Abstract
The proper function of cardiomyocytes (CMs) is highly related to the Z-disc, which has a pivotal role in orchestrating the sarcomeric cytoskeletal function. To better understand Z-disc related cardiomyopathies, novel models of Z-disc damage have to be developed. Human pluripotent stem cell (hPSC)-derived CMs can serve as an in vitro model to better understand the sarcomeric cytoskeleton. A femtosecond laser system can be applied for localized and defined damage application within cells as single Z-discs can be removed. We have investigated the changes in force generation via traction force microscopy, and in gene expression after Z-disc manipulation in hPSC-derived CMs. We observed a significant weakening of force generation after removal of a Z-disc. However, no significant changes of the number of contractions after manipulation were detected. The stress related gene NF-kB was significantly upregulated. Additionally, α-actinin (ACTN2) and filamin-C (FLNc) were upregulated, pointing to remodeling of the Z-disc and the sarcomeric cytoskeleton. Ultimately, cardiac troponin I (TNNI3) and cardiac muscle troponin T (TNNT2) were significantly downregulated. Our results allow a better understanding of transcriptional coupling of Z-disc damage and the relation of damage to force generation and can therefore finally pave the way to novel therapies of sarcomeric disorders.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Sören Donath
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Emanuel Georg Brückner
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Santoshi Biswanath Devadas
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Fiene Daniel
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Lara Gentemann
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Robert Zweigerdt
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Stefan Michael Klaus Kalies
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (D.M.); (S.D.); (E.G.B.); (F.D.); (L.G.); (A.H.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany; (S.B.D.); (R.Z.)
- Lower Saxony Centre for Biomedical Engineering and Implant Research and Development (NIFE), 30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
19
|
Solís C, Russell B. Striated muscle proteins are regulated both by mechanical deformation and by chemical post-translational modification. Biophys Rev 2021; 13:679-695. [PMID: 34777614 PMCID: PMC8555064 DOI: 10.1007/s12551-021-00835-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023] Open
Abstract
All cells sense force and build their cytoskeleton to optimize function. How is this achieved? Two major systems are involved. The first is that load deforms specific protein structures in a proportional and orientation-dependent manner. The second is post-translational modification of proteins as a consequence of signaling pathway activation. These two processes work together in a complex way so that local subcellular assembly as well as overall cell function are controlled. This review discusses many cell types but focuses on striated muscle. Detailed information is provided on how load deforms the structure of proteins in the focal adhesions and filaments, using α-actinin, vinculin, talin, focal adhesion kinase, LIM domain-containing proteins, filamin, myosin, titin, and telethonin as examples. Second messenger signals arising from external triggers are distributed throughout the cell causing post-translational or chemical modifications of protein structures, with the actin capping protein CapZ and troponin as examples. There are numerous unanswered questions of how mechanical and chemical signals are integrated by muscle proteins to regulate sarcomere structure and function yet to be studied. Therefore, more research is needed to see how external triggers are integrated with local tension generated within the cell. Nonetheless, maintenance of tension in the sarcomere is the essential and dominant mechanism, leading to the well-known phrase in exercise physiology: "use it or lose it."
Collapse
Affiliation(s)
- Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
20
|
Doran S, Arif M, Lam S, Bayraktar A, Turkez H, Uhlen M, Boren J, Mardinoglu A. Multi-omics approaches for revealing the complexity of cardiovascular disease. Brief Bioinform 2021; 22:bbab061. [PMID: 33725119 PMCID: PMC8425417 DOI: 10.1093/bib/bbab061] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/20/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
The development and progression of cardiovascular disease (CVD) can mainly be attributed to the narrowing of blood vessels caused by atherosclerosis and thrombosis, which induces organ damage that will result in end-organ dysfunction characterized by events such as myocardial infarction or stroke. It is also essential to consider other contributory factors to CVD, including cardiac remodelling caused by cardiomyopathies and co-morbidities with other diseases such as chronic kidney disease. Besides, there is a growing amount of evidence linking the gut microbiota to CVD through several metabolic pathways. Hence, it is of utmost importance to decipher the underlying molecular mechanisms associated with these disease states to elucidate the development and progression of CVD. A wide array of systems biology approaches incorporating multi-omics data have emerged as an invaluable tool in establishing alterations in specific cell types and identifying modifications in signalling events that promote disease development. Here, we review recent studies that apply multi-omics approaches to further understand the underlying causes of CVD and provide possible treatment strategies by identifying novel drug targets and biomarkers. We also discuss very recent advances in gut microbiota research with an emphasis on how diet and microbial composition can impact the development of CVD. Finally, we present various biological network analyses and other independent studies that have been employed for providing mechanistic explanation and developing treatment strategies for end-stage CVD, namely myocardial infarction and stroke.
Collapse
Affiliation(s)
- Stephen Doran
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Simon Lam
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Abdulahad Bayraktar
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Boren
- Institute of Medicine, Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
21
|
The Role of GSK-3β in the Regulation of Protein Turnover, Myosin Phenotype, and Oxidative Capacity in Skeletal Muscle under Disuse Conditions. Int J Mol Sci 2021; 22:ijms22105081. [PMID: 34064895 PMCID: PMC8151958 DOI: 10.3390/ijms22105081] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscles, being one of the most abundant tissues in the body, are involved in many vital processes, such as locomotion, posture maintenance, respiration, glucose homeostasis, etc. Hence, the maintenance of skeletal muscle mass is crucial for overall health, prevention of various diseases, and contributes to an individual’s quality of life. Prolonged muscle inactivity/disuse (due to limb immobilization, mechanical ventilation, bedrest, spaceflight) represents one of the typical causes, leading to the loss of muscle mass and function. This disuse-induced muscle loss primarily results from repressed protein synthesis and increased proteolysis. Further, prolonged disuse results in slow-to-fast fiber-type transition, mitochondrial dysfunction and reduced oxidative capacity. Glycogen synthase kinase 3β (GSK-3β) is a key enzyme standing at the crossroads of various signaling pathways regulating a wide range of cellular processes. This review discusses various important roles of GSK-3β in the regulation of protein turnover, myosin phenotype, and oxidative capacity in skeletal muscles under disuse/unloading conditions and subsequent recovery. According to its vital functions, GSK-3β may represent a perspective therapeutic target in the treatment of muscle wasting induced by chronic disuse, aging, and a number of diseases.
Collapse
|
22
|
Wirianto M, Yang J, Kim E, Gao S, Paudel KR, Choi JM, Choe J, Gloston GF, Ademoji P, Parakramaweera R, Jin J, Esser KA, Jung SY, Geng YJ, Lee HK, Chen Z, Yoo SH. The GSK-3β-FBXL21 Axis Contributes to Circadian TCAP Degradation and Skeletal Muscle Function. Cell Rep 2021; 32:108140. [PMID: 32937135 PMCID: PMC8299398 DOI: 10.1016/j.celrep.2020.108140] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/04/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022] Open
Abstract
FBXL21 is a clock-controlled E3 ligase modulating circadian periodicity via subcellular-specific CRYPTOCHROME degradation. How FBXL21 regulates tissue-specific circadian physiology and what mechanism operates upstream is poorly understood. Here we report the sarcomere component TCAP as a cytoplasmic substrate of FBXL21. FBXL21 interacts with TCAP in a circadian manner antiphasic to TCAP accumulation in skeletal muscle, and circadian TCAP oscillation is disrupted in Psttm mice with an Fbxl21 hypomorph mutation. GSK-3β phosphorylates FBXL21 and TCAP to activate FBXL21-mediated, phosphodegron-dependent TCAP degradation. GSK-3β inhibition or knockdown diminishes FBXL21-Cul1 complex formation and delays FBXL21-mediated TCAP degradation. Finally, Psttm mice show significant skeletal muscle defects, including impaired fiber size, exercise tolerance, grip strength, and response to glucocorticoid-induced atrophy, in conjunction with cardiac dysfunction. These data highlight a circadian regulatory pathway where a GSK-3β-FBXL21 functional axis controls TCAP degradation via SCF complex formation and regulates skeletal muscle function. Wirianto et al. find that the circadian E3 ligase FBXL21 drives rhythmic degradation of the sarcomeric protein TCAP in skeletal muscle. GSK-3β co-phosphorylates FBXL21 and TCAP and promotes SCF complex formation and phosphodegron-dependent TCAP turnover. Psttm mice, expressing a hypomorphic Fbxl21 mutant, show dysregulated TCAP degradation and impaired muscle function.
Collapse
Affiliation(s)
- Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Jiah Yang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Song Gao
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Keshav Raj Paudel
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Jong Min Choi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jeehwan Choe
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Gabrielle F Gloston
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Precious Ademoji
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Randika Parakramaweera
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Jianping Jin
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, The University of Florida College of Medicine, Gainesville, FL 32610-0274, USA
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yong-Jian Geng
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Hyun Kyoung Lee
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA.
| |
Collapse
|
23
|
Feyen DAM, McKeithan WL, Bruyneel AAN, Spiering S, Hörmann L, Ulmer B, Zhang H, Briganti F, Schweizer M, Hegyi B, Liao Z, Pölönen RP, Ginsburg KS, Lam CK, Serrano R, Wahlquist C, Kreymerman A, Vu M, Amatya PL, Behrens CS, Ranjbarvaziri S, Maas RGC, Greenhaw M, Bernstein D, Wu JC, Bers DM, Eschenhagen T, Metallo CM, Mercola M. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep 2021; 32:107925. [PMID: 32697997 PMCID: PMC7437654 DOI: 10.1016/j.celrep.2020.107925] [Citation(s) in RCA: 232] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have enormous potential for the study of human cardiac disorders. However, their physiological immaturity severely limits their utility as a model system and their adoption for drug discovery. Here, we describe maturation media designed to provide oxidative substrates adapted to the metabolic needs of human iPSC (hiPSC)-CMs. Compared with conventionally cultured hiPSC-CMs, metabolically matured hiPSC-CMs contract with greater force and show an increased reliance on cardiac sodium (Na+) channels and sarcoplasmic reticulum calcium (Ca2+) cycling. The media enhance the function, long-term survival, and sarcomere structures in engineered heart tissues. Use of the maturation media made it possible to reliably model two genetic cardiac diseases: long QT syndrome type 3 due to a mutation in the cardiac Na+ channel SCN5A and dilated cardiomyopathy due to a mutation in the RNA splicing factor RBM20. The maturation media should increase the fidelity of hiPSC-CMs as disease models. Physiological immaturity of iPSC-derived cardiomyocytes limits their fidelity as disease models. Feyen et al. developed a low glucose, high oxidative substrate media that increase maturation of ventricular-like hiPSC-CMs in 2D and 3D cultures relative to standard protocols. Improved characteristics include a low resting Vm, rapid depolarization, and increased Ca2+ dependence and force generation.
Collapse
Affiliation(s)
- Dries A M Feyen
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wesley L McKeithan
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Arne A N Bruyneel
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sean Spiering
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Larissa Hörmann
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bärbel Ulmer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hui Zhang
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Francesca Briganti
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michaela Schweizer
- Electron Microscopy Unit, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bence Hegyi
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Zhandi Liao
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | | | - Kenneth S Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Chi Keung Lam
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Serrano
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Christine Wahlquist
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Alexander Kreymerman
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Vu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Prashila L Amatya
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Charlotta S Behrens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sara Ranjbarvaziri
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Renee G C Maas
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matthew Greenhaw
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel Bernstein
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Wu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
24
|
Solís C, Solaro RJ. Novel insights into sarcomere regulatory systems control of cardiac thin filament activation. J Gen Physiol 2021; 153:211903. [PMID: 33740037 PMCID: PMC7988513 DOI: 10.1085/jgp.202012777] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Our review focuses on sarcomere regulatory mechanisms with a discussion of cardiac-specific modifications to the three-state model of thin filament activation from a blocked to closed to open state. We discuss modulation of these thin filament transitions by Ca2+, by crossbridge interactions, and by thick filament–associated proteins, cardiac myosin–binding protein C (cMyBP-C), cardiac regulatory light chain (cRLC), and titin. Emerging evidence supports the idea that the cooperative activation of the thin filaments despite a single Ca2+ triggering regulatory site on troponin C (cTnC) cannot be considered in isolation of other functional domains of the sarcomere. We discuss long- and short-range interactions among these domains with the regulatory units of thin filaments, including proteins at the barbed end at the Z-disc and the pointed end near the M-band. Important to these discussions is the ever-increasing understanding of the role of cMyBP-C, cRLC, and titin filaments. Detailed knowledge of these control processes is critical to the understanding of mechanisms sustaining physiological cardiac state with varying hemodynamic load, to better defining genetic and acquired cardiac disorders, and to developing targets for therapies at the level of the sarcomeres.
Collapse
Affiliation(s)
- Christopher Solís
- University of Illinois at Chicago, College of Medicine, Department of Physiology and Biophysics and Center for Cardiovascular Research, Chicago, IL
| | - R John Solaro
- University of Illinois at Chicago, College of Medicine, Department of Physiology and Biophysics and Center for Cardiovascular Research, Chicago, IL
| |
Collapse
|
25
|
The Role of Z-disc Proteins in Myopathy and Cardiomyopathy. Int J Mol Sci 2021; 22:ijms22063058. [PMID: 33802723 PMCID: PMC8002584 DOI: 10.3390/ijms22063058] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The Z-disc acts as a protein-rich structure to tether thin filament in the contractile units, the sarcomeres, of striated muscle cells. Proteins found in the Z-disc are integral for maintaining the architecture of the sarcomere. They also enable it to function as a (bio-mechanical) signalling hub. Numerous proteins interact in the Z-disc to facilitate force transduction and intracellular signalling in both cardiac and skeletal muscle. This review will focus on six key Z-disc proteins: α-actinin 2, filamin C, myopalladin, myotilin, telethonin and Z-disc alternatively spliced PDZ-motif (ZASP), which have all been linked to myopathies and cardiomyopathies. We will summarise pathogenic variants identified in the six genes coding for these proteins and look at their involvement in myopathy and cardiomyopathy. Listing the Minor Allele Frequency (MAF) of these variants in the Genome Aggregation Database (GnomAD) version 3.1 will help to critically re-evaluate pathogenicity based on variant frequency in normal population cohorts.
Collapse
|
26
|
Jiang H, Hooper C, Kelly M, Steeples V, Simon JN, Beglov J, Azad AJ, Leinhos L, Bennett P, Ehler E, Kalisch-Smith JI, Sparrow DB, Fischer R, Heilig R, Isackson H, Ehsan M, Patone G, Huebner N, Davies B, Watkins H, Gehmlich K. Functional analysis of a gene-edited mouse model to gain insights into the disease mechanisms of a titin missense variant. Basic Res Cardiol 2021; 116:14. [PMID: 33637999 PMCID: PMC7910237 DOI: 10.1007/s00395-021-00853-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/10/2021] [Indexed: 11/03/2022]
Abstract
Titin truncating variants are a well-established cause of cardiomyopathy; however, the role of titin missense variants is less well understood. Here we describe the generation of a mouse model to investigate the underlying disease mechanism of a previously reported titin A178D missense variant identified in a family with non-compaction and dilated cardiomyopathy. Heterozygous and homozygous mice carrying the titin A178D missense variant were characterised in vivo by echocardiography. Heterozygous mice had no detectable phenotype at any time point investigated (up to 1 year). By contrast, homozygous mice developed dilated cardiomyopathy from 3 months. Chronic adrenergic stimulation aggravated the phenotype. Targeted transcript profiling revealed induction of the foetal gene programme and hypertrophic signalling pathways in homozygous mice, and these were confirmed at the protein level. Unsupervised proteomics identified downregulation of telethonin and four-and-a-half LIM domain 2, as well as the upregulation of heat shock proteins and myeloid leukaemia factor 1. Loss of telethonin from the cardiac Z-disc was accompanied by proteasomal degradation; however, unfolded telethonin accumulated in the cytoplasm, leading to a proteo-toxic response in the mice.We show that the titin A178D missense variant is pathogenic in homozygous mice, resulting in cardiomyopathy. We also provide evidence of the disease mechanism: because the titin A178D variant abolishes binding of telethonin, this leads to its abnormal cytoplasmic accumulation. Subsequent degradation of telethonin by the proteasome results in proteasomal overload, and activation of a proteo-toxic response. The latter appears to be a driving factor for the cardiomyopathy observed in the mouse model.
Collapse
Affiliation(s)
- He Jiang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Charlotte Hooper
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Matthew Kelly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Violetta Steeples
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Jillian N Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Julia Beglov
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Amar J Azad
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Lisa Leinhos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Pauline Bennett
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King's College London BHF Centre of Research Excellence, London, UK
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King's College London BHF Centre of Research Excellence, London, UK
| | | | - Duncan B Sparrow
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Roman Fischer
- Nuffield Department of Clinical Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Raphael Heilig
- Nuffield Department of Clinical Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Henrik Isackson
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
| | - Mehroz Ehsan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Giannino Patone
- Max Delbrueck Centre for Molecular Medicine, Berlin, Germany
| | - Norbert Huebner
- Max Delbrueck Centre for Molecular Medicine, Berlin, Germany
| | - Benjamin Davies
- Transgenic Core, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU, UK.
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
27
|
Russell B, Solís C. Mechanosignaling pathways alter muscle structure and function by post-translational modification of existing sarcomeric proteins to optimize energy usage. J Muscle Res Cell Motil 2021; 42:367-380. [PMID: 33595762 DOI: 10.1007/s10974-021-09596-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/04/2021] [Indexed: 12/29/2022]
Abstract
A transduced mechanical signal arriving at its destination in muscle alters sarcomeric structure and function. A major question addressed is how muscle mass and tension generation are optimized to match actual performance demands so that little energy is wasted. Three cases for improved energy efficiency are examined: the troponin complex for tuning force production, control of the myosin heads in a resting state, and the Z-disc proteins for sarcomere assembly. On arrival, the regulation of protein complexes is often controlled by post-translational modification (PTM), of which the most common are phosphorylation by kinases, deacetylation by histone deacetylases and ubiquitination by E3 ligases. Another branch of signals acts not through peptide covalent bonding but via ligand interactions (e.g. Ca2+ and phosphoinositide binding). The myosin head and the regulation of its binding to actin by the troponin complex is the best and earliest example of signal destinations that modify myofibrillar contractility. PTMs in the troponin complex regulate both the efficiency of the contractile function to match physiologic demand for work, and muscle mass via protein degradation. The regulation of sarcomere assembly by integration of incoming signaling pathways causing the same PTMs or ligand binding are discussed in response to mechanical loading and unloading by the Z-disc proteins CapZ, α-actinin, telethonin, titin N-termini, and others. Many human mutations that lead to cardiomyopathy and heart disease occur in the proteins discussed above, which often occur at their PTM or ligand binding sites.
Collapse
Affiliation(s)
- Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
28
|
Lewis HR, Eminaga S, Gautel M, Avkiran M. Phosphorylation at Serines 157 and 161 Is Necessary for Preserving Cardiac Expression Level and Functions of Sarcomeric Z-Disc Protein Telethonin. Front Physiol 2021; 12:732020. [PMID: 34566695 PMCID: PMC8455888 DOI: 10.3389/fphys.2021.732020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Aims: In cardiac myocytes, the sarcomeric Z-disc protein telethonin is constitutively bis-phosphorylated at C-terminal residues S157 and S161; however, the functional significance of this phosphorylation is not known. We sought to assess the significance of telethonin phosphorylation in vivo, using a novel knock-in (KI) mouse model generated to express non-phosphorylatable telethonin (Tcap S157/161A). Methods and Results: Tcap S157/161A and wild-type (WT) littermates were characterized by echocardiography at baseline and after sustained β-adrenergic stimulation via isoprenaline infusion. Heart tissues were collected for gravimetric, biochemical, and histological analyses. At baseline, Tcap S157/161A mice did not show any variances in cardiac structure or function compared with WT littermates and mutant telethonin remained localized to the Z-disc. Ablation of telethonin phosphorylation sites resulted in a gene-dosage dependent decrease in the cardiac telethonin protein expression level in mice carrying the S157/161A alleles, without any alteration in telethonin mRNA levels. The proteasome inhibitor MG132 significantly increased the expression level of S157/161A telethonin protein in myocytes from Tcap S157/161A mice, but not telethonin protein in myocytes from WT mice, indicating a role for the ubiquitin-proteasome system in the regulation of telethonin protein expression level. Tcap S157/161A mice challenged with sustained β-adrenergic stimulation via isoprenaline infusion developed cardiac hypertrophy accompanied by mild systolic dysfunction. Furthermore, the telethonin protein expression level was significantly increased in WT mice following isoprenaline stimulation but this response was blunted in Tcap S157/161A mice. Conclusion: Overall, these data reveal that telethonin protein turnover in vivo is regulated in a novel phosphorylation-dependent manner and suggest that C-terminal phosphorylation may protect telethonin against proteasomal degradation and preserve cardiac function during hemodynamic stress. Given that human telethonin C-terminal mutations have been associated with cardiac and skeletal myopathies, further research on their potential impact on phosphorylation-dependent regulation of telethonin protein expression could provide valuable mechanistic insight into those myopathies.
Collapse
Affiliation(s)
- Hannah R. Lewis
- School of Cardiovascular Medicine and Sciences, St Thomas’ Hospital, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Seda Eminaga
- School of Cardiovascular Medicine and Sciences, St Thomas’ Hospital, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Mathias Gautel
- School of Basic and Medical Biosciences, Guy’s Hospital, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Metin Avkiran
- School of Cardiovascular Medicine and Sciences, St Thomas’ Hospital, King’s College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
- *Correspondence: Metin Avkiran,
| |
Collapse
|
29
|
Tan Y, Wang L, Gao J, Ma J, Yu H, Zhang Y, Wang T, Han L. Multiomics Integrative Analysis for Discovering the Potential Mechanism of Dioscin against Hyperuricemia Mice. J Proteome Res 2020; 20:645-660. [PMID: 33107303 DOI: 10.1021/acs.jproteome.0c00584] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hyperuricemia is a well-known key risk factor for gout and can cause a variety of metabolic diseases. Several studies have shown that dioscin could improve metabolic symptoms and reduce the uric acid level in blood. However, there is no comprehensive metabolomic study on the anti-hyperuricemia effects of dioscin. A total of 29 adult male Kunming mice were divided into three groups: Normal (blank), PO (potassium oxonate-administrated, 200 mg/kg/day), and Dioscin (potassium oxonate + dioscin, potassium oxonate 200 mg/kg/day, dioscin 50 mg/kg/day). All mice were treated for 42 days via oral gavage. This paper implemented an untargeted metabolomics study based on 1H NMR and LC-MS to discover the comprehensive mechanism of dioscin. Furthermore, a targeted lipidomics was fulfilled to further analyze the lipid metabolism disorder. Finally, the metabolic pathway mediated by dioscin was verified at the gene level by means of transcriptomics. The results show 53 different metabolites were closely related to the improvement of dioscin in PO-induced hyperuricemia, and 19 of them were lipids. These metabolites are mainly involved in the tricarboxylic acid cycle, lipid metabolism, amino acid metabolism, and pyrimidine metabolism. According to the transcriptomics study, the levels of 89 genes were significantly changed in the PO group compared to the normal control. Among them, six gene levels were restored by the treatment of dioscin. The six changed genes (tx1b, Tsku, Tmem163, Psmc3ip, Tcap, Tbx15) are mainly involved in the cell cycle and energy metabolism. These metabolites and genes might provide useful information for further study of the therapeutic mechanism of dioscin.
Collapse
Affiliation(s)
- Yao Tan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| | - Liming Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| | - Jian Gao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| | - Junhong Ma
- Tianjin Hospital of ITCWM Nankai Hospital, Tianjin 300100, China
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| | - Yi Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| | - Tao Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| | - Lifeng Han
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, 10 Poyanghu Road, Jinghai District, Tianjin 301617, China
| |
Collapse
|
30
|
Peris-Moreno D, Taillandier D, Polge C. MuRF1/TRIM63, Master Regulator of Muscle Mass. Int J Mol Sci 2020; 21:ijms21186663. [PMID: 32933049 PMCID: PMC7555135 DOI: 10.3390/ijms21186663] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
The E3 ubiquitin ligase MuRF1/TRIM63 was identified 20 years ago and suspected to play important roles during skeletal muscle atrophy. Since then, numerous studies have been conducted to decipher the roles, molecular mechanisms and regulation of this enzyme. This revealed that MuRF1 is an important player in the skeletal muscle atrophy process occurring during catabolic states, making MuRF1 a prime candidate for pharmacological treatments against muscle wasting. Indeed, muscle wasting is an associated event of several diseases (e.g., cancer, sepsis, diabetes, renal failure, etc.) and negatively impacts the prognosis of patients, which has stimulated the search for MuRF1 inhibitory molecules. However, studies on MuRF1 cardiac functions revealed that MuRF1 is also cardioprotective, revealing a yin and yang role of MuRF1, being detrimental in skeletal muscle and beneficial in the heart. This review discusses data obtained on MuRF1, both in skeletal and cardiac muscles, over the past 20 years, regarding the structure, the regulation, the location and the different functions identified, and the first inhibitors reported, and aim to draw the picture of what is known about MuRF1. The review also discusses important MuRF1 characteristics to consider for the design of future drugs to maintain skeletal muscle mass in patients with different pathologies.
Collapse
|
31
|
Swist S, Unger A, Li Y, Vöge A, von Frieling-Salewsky M, Skärlén Å, Cacciani N, Braun T, Larsson L, Linke WA. Maintenance of sarcomeric integrity in adult muscle cells crucially depends on Z-disc anchored titin. Nat Commun 2020; 11:4479. [PMID: 32900999 PMCID: PMC7478974 DOI: 10.1038/s41467-020-18131-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
The giant protein titin is thought to be required for sarcomeric integrity in mature myocytes, but direct evidence for this hypothesis is limited. Here, we describe a mouse model in which Z-disc-anchored TTN is depleted in adult skeletal muscles. Inactivation of TTN causes sarcomere disassembly and Z-disc deformations, force impairment, myocyte de-stiffening, upregulation of TTN-binding mechanosensitive proteins and activation of protein quality-control pathways, concomitant with preferential loss of thick-filament proteins. Interestingly, expression of the myosin-bound Cronos-isoform of TTN, generated from an alternative promoter not affected by the targeting strategy, does not prevent deterioration of sarcomere formation and maintenance. Finally, we demonstrate that loss of Z-disc-anchored TTN recapitulates muscle remodeling in critical illness ‘myosinopathy’ patients, characterized by TTN-depletion and loss of thick filaments. We conclude that full-length TTN is required to integrate Z-disc and A-band proteins into the mature sarcomere, a function that is lost when TTN expression is pathologically lowered. Titin is considered an integrator of muscle cell proteins but direct evidence is limited. Here, titin is inactivated in adult mouse muscles, which causes sarcomere disassembly, protein mis-expression and force impairment, recapitulating key alterations in critical illness myopathy patient muscles.
Collapse
Affiliation(s)
- Sandra Swist
- Department of Systems Physiology, Ruhr University Bochum, D-44780, Bochum, Germany.
| | - Andreas Unger
- Institute of Physiology II, University of Munster, D-48149, Munster, Germany
| | - Yong Li
- Institute of Physiology II, University of Munster, D-48149, Munster, Germany
| | - Anja Vöge
- Department of Systems Physiology, Ruhr University Bochum, D-44780, Bochum, Germany
| | | | - Åsa Skärlén
- Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Nicola Cacciani
- Department of Physiology and Pharmacology, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231, Bad Nauheim, Germany
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Wolfgang A Linke
- Institute of Physiology II, University of Munster, D-48149, Munster, Germany.
| |
Collapse
|
32
|
Turker I, Makiyama T, Ueyama T, Shimizu A, Yamakawa M, Chen P, Vatta M, Horie M, Ai T. Telethonin
variants found in Brugada syndrome, J‐wave pattern ECG, and ARVC reduce peak Na
v
1.5 currents in HEK‐293 cells. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2020; 43:838-846. [DOI: 10.1111/pace.13996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/11/2020] [Accepted: 06/21/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Isik Turker
- Department of Medicine Krannert Institute of Cardiology Indiana University School of Medicine Indianapolis Indiana
| | - Takeru Makiyama
- Cardiovascular Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Takeshi Ueyama
- Department of Cardiology Yamaguchi University School of Medical Science Yamaguchi Japan
| | - Akihiko Shimizu
- Department of Cardiology Yamaguchi University School of Medical Science Yamaguchi Japan
| | - Masaru Yamakawa
- Department of Pediatrics Kobe City Medical Center General Hospital Kobe Japan
| | - Peng‐Sheng Chen
- Department of Medicine Krannert Institute of Cardiology Indiana University School of Medicine Indianapolis Indiana
| | - Matteo Vatta
- Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis Indiana
| | - Minoru Horie
- Department of Cardiovascular and Respiratory Medicine Shiga University of Medical Science Shiga Japan
| | - Tomohiko Ai
- Department of Medicine Krannert Institute of Cardiology Indiana University School of Medicine Indianapolis Indiana
- Department of Clinical Laboratory Medicine Juntendo University School of Medicine Tokyo Japan
| |
Collapse
|
33
|
Deficiency of nuclear receptor interaction protein leads to cardiomyopathy by disrupting sarcomere structure and mitochondrial respiration. J Mol Cell Cardiol 2019; 137:9-24. [PMID: 31629737 DOI: 10.1016/j.yjmcc.2019.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/15/2019] [Accepted: 09/17/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cardiomyopathy is a common and lethal complication in patients with limb-girdle muscular dystrophy (LGMD), one of the most prevalent forms of muscular dystrophy. The pathogenesis underlying LGMD-related cardiomyopathy remains unclear. NRIP (gene name DCAF6), a Ca2+-dependent calmodulin binding protein, was reduced in dystrophic muscles from LGMD patients. Mice lacking NRIP exhibit a myopathic phenotype resembling that in LGMD patients, making NRIP deficiency a potential culprit leading to cardiomyopathy. This study aimed to determine if NRIP deficiency leads to cardiomyopathy and to explore the underlying molecular mechanisms. METHODS AND RESULTS NRIP expression was reduced in both human and mouse failing hearts. Muscle-specific NRIP knockout (MCK-Cre::Dcaf6flox/flox) mouse heart and isolated cardiomyocytes exhibited markedly reduced contractility. Transmission electron microscopy revealed abnormal sarcomere structures and mitochondrial morphology in MCK-Cre::Dcaf6flox/flox hearts. Protein co-immunoprecipitation and confocal imaging revealed that NRIP interacts with α-actinin 2 (ACTN2) at the Z-disc. We found that NRIP facilitated ACTN2-mediated F-actin bundling, and that NRIP deficiency resulted in reduced binding between Z-disc proteins ACTN2 and Cap-Z. In addition, NRIP-deficiency led to increased mitochondrial ROS and impaired mitochondrial respiration/ATP production owing to elevated cellular NADH/NAD+ ratios. Treatment with mitochondria-directed antioxidant mitoTEMPO or NAD+ precursor nicotinic acid restored mitochondrial function and cardiac contractility in MCK-Cre::Dcaf6flox/flox mice. CONCLUSIONS NRIP is essential to maintain sarcomere structure and mitochondrial/contractile function in cardiomyocytes. Our results revealed a novel role for NRIP deficiency in the pathogenesis of LGMD and heart failure. Targeting NRIP, therefore, could be a powerful new approach to treat myocardial dysfunction in LGMD and heart failure patients.
Collapse
|
34
|
Sharlo K, Paramonova I, Turtikova O, Tyganov S, Shenkman B. Plantar mechanical stimulation prevents calcineurin-NFATc1 inactivation and slow-to-fast fiber type shift in rat soleus muscle under hindlimb unloading. J Appl Physiol (1985) 2019; 126:1769-1781. [PMID: 31046517 DOI: 10.1152/japplphysiol.00029.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The prevailing myosin isoform [myosin heavy chain (MyHC)] in a skeletal muscle determines contractile properties of the muscle. Under actual or simulated microgravity conditions such as human bed rest or rat hindlimb unloading, decrease in expression of MyHC of the slow type [MyHC I(β)] has been observed. It was demonstrated that increasing sensory input by performing plantar mechanical stimulation (PMS) on the soles of the feet results in an increase in neuromuscular activation of the lower limb muscles and may prevent slow-to-fast fiber type shift. The calcineurin-nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) signaling pathway is the main cascade regulating MyHC I(β) expression. The present study was aimed to analyze the states of the calcineurin-NFATc1 signaling cascade under conditions of PMS during rat hindlimb unloading. Male Wistar rats were randomly assigned to vivarium control groups and 1-day unloading (1HS), 3-day unloading (3HS), 1HS+PMS, and 3HS+PMS groups. We found that both 1 day and 3 days of unloading caused decrease in MyHC I(β) mRNA expression and decrease in glycogen synthase kinase-3β phosphorylation (Ser 9) that brought about the kinase activation, and these effects of unloading were prevented by PMS. Three days of unloading also caused increase in expression of calsarcin-2 (myozenin-I), which was found to be the endogenous calcineurin inhibitor. The level of calsarcin-2 expression in the 3HS+PMS group did not differ from the control group. Therefore, we conclude that PMS upregulates the calcineurin-NFATc1 signaling pathway and prevents unloading-induced MyHC I(β) decrease. NEW & NOTEWORTHY It is widely accepted that changes in the myosin phenotype during functional unloading (disuse) are determined by a decreased expression of the myosin heavy chain (MyHC) I(β) gene, and this decrease leads to changes of contractile and fatigue characteristics of soleus muscle. The calcineurin-nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) pathway is one of the most important signaling cascades regulating slow MyHC isoform expression. The present study is the first to show that plantar mechanical stimulation upregulates calcineurin-NFATc1 signaling in soleus muscles of hindlimb-unloaded rats.
Collapse
Affiliation(s)
- Kristina Sharlo
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Inna Paramonova
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Olga Turtikova
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Sergey Tyganov
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Boris Shenkman
- Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| |
Collapse
|
35
|
Müller D, Hagenah D, Biswanath S, Coffee M, Kampmann A, Zweigerdt R, Heisterkamp A, Kalies SMK. Femtosecond laser-based nanosurgery reveals the endogenous regeneration of single Z-discs including physiological consequences for cardiomyocytes. Sci Rep 2019; 9:3625. [PMID: 30842507 PMCID: PMC6403391 DOI: 10.1038/s41598-019-40308-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/13/2019] [Indexed: 11/24/2022] Open
Abstract
A highly organized cytoskeleton architecture is the basis for continuous and controlled contraction in cardiomyocytes (CMs). Abnormalities in cytoskeletal elements, like the Z-disc, are linked to several diseases. It is challenging to reveal the mechanisms of CM failure, endogenous repair, or mechanical homeostasis on the scale of single cytoskeletal elements. Here, we used a femtosecond (fs) laser to ablate single Z-discs in human pluripotent stem cells (hPSC) -derived CMs (hPSC-CM) and neonatal rat CMs. We show, that CM viability was unaffected by the loss of a single Z-disc. Furthermore, more than 40% of neonatal rat and 68% of hPSC-CMs recovered the Z-disc loss within 24 h. Significant differences to control cells, after the Z-disc loss, in terms of cell perimeter, x- and y-expansion and calcium homeostasis were not found. Only 14 days in vitro old hPSC-CMs reacted with a significant decrease in cell area, x- and y-expansion 24 h past nanosurgery. This demonstrates that CMs can compensate the loss of a single Z-disc and recover a regular sarcomeric pattern during spontaneous contraction. It also highlights the significant potential of fs laser-based nanosurgery to physically micro manipulate CMs to investigate cytoskeletal functions and organization of single elements.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany. .,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany. .,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.
| | - Dorian Hagenah
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Santoshi Biswanath
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Michelle Coffee
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Andreas Kampmann
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.,Clinic for Cranio-Maxillo-Facial Surgery, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Stefan M K Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
36
|
Freundt JK, Linke WA. Titin as a force-generating muscle protein under regulatory control. J Appl Physiol (1985) 2018; 126:1474-1482. [PMID: 30521425 DOI: 10.1152/japplphysiol.00865.2018] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Titin has long been recognized as a mechanical protein in muscle cells that has a main function as a molecular spring in the contractile units, the sarcomeres. Recent work suggests that the titin spring contributes to muscle contraction in a more active manner than previously thought. In this review, we highlight this property, specifically the ability of the immunoglobulin-like (Ig) domains of titin to undergo unfolding-refolding transitions when isolated titin molecules or skeletal myofibrils are held at physiological force levels. Folding of titin Ig domains under force is a hitherto unappreciated, putative source of work production in muscle cells, which could work in synergy with the actomyosin system to maximize the energy delivered by a stretched, actively contracting muscle. This review also focuses on the mechanisms shown to modulate titin-based viscoelastic forces in skeletal muscle cells, including chaperone binding, titin oxidation, phosphorylation, Ca2+ binding, and interaction with actin filaments. Along the way, we discuss which of these modulatory mechanisms might contribute to the phenomenon of residual force enhancement relevant for eccentric muscle contractions. Finally, a brief perspective is added on the potential for the alterations in titin-based force to dynamically alter mechano-chemical signaling pathways in the muscle cell. We conclude that titin from skeletal muscle is a determinant of both passive and active tension and a bona fide mechanosensor, whose stiffness is tuned by various independent mechanisms.
Collapse
Affiliation(s)
- Johanna K Freundt
- Institute of Physiology II, University of Muenster , Muenster , Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster , Muenster , Germany
| |
Collapse
|
37
|
UBE2E1 Is Preferentially Expressed in the Cytoplasm of Slow-Twitch Fibers and Protects Skeletal Muscles from Exacerbated Atrophy upon Dexamethasone Treatment. Cells 2018; 7:cells7110214. [PMID: 30453501 PMCID: PMC6262581 DOI: 10.3390/cells7110214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 12/27/2022] Open
Abstract
Skeletal muscle mass is reduced during many diseases or physiological situations (disuse, aging), which results in decreased strength and increased mortality. Muscle mass is mainly controlled by the ubiquitin-proteasome system (UPS), involving hundreds of ubiquitinating enzymes (E2s and E3s) that target their dedicated substrates for subsequent degradation. We recently demonstrated that MuRF1, an E3 ubiquitin ligase known to bind to sarcomeric proteins (telethonin, α-actin, myosins) during catabolic situations, interacts with 5 different E2 enzymes and that these E2-MuRF1 couples are able to target telethonin, a small sarcomeric protein, for degradation. Amongst the E2s interacting with MuRF1, E2E1 was peculiar as the presence of the substrate was necessary for optimal MuRF1-E2E1 interaction. In this work, we focused on the putative role of E2E1 during skeletal muscle atrophy. We found that E2E1 expression was restricted to type I and type IIA muscle fibers and was not detectable in type IIB fibers. This strongly suggests that E2E1 targets are fiber-specific and may be strongly linked to the contractile and metabolic properties of the skeletal muscle. However, E2E1 knockdown was not sufficient for preserving the protein content in C2C12 myotubes subjected to a catabolic state (dexamethasone treatment), suggesting that E2E1 is not involved in the development of muscle atrophy. By contrast, E2E1 knockdown aggravated the atrophying process in both catabolic C2C12 myotubes and the Tibialis anterior muscle of mice, suggesting that E2E1 has a protective effect on muscle mass.
Collapse
|
38
|
Urmaliya V, Franchelli G. A multidimensional sight on cardiac failure: uncovered from structural to molecular level. Heart Fail Rev 2018; 22:357-370. [PMID: 28474325 DOI: 10.1007/s10741-017-9610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Heart failure is one of the leading causes of death, with high mortality rate within 5 years after diagnosis. Treatment and prognosis options for heart failure primarily targeted on hemodynamic and neurohumoral components that drive progressive deterioration of the heart. However, given the multifactorial background that eventually leads to the "phenotype" named heart failure, better insight into the various components may lead to personalized treatment opportunities. Indeed, currently used criteria to diagnose and/or classify heart failure are possibly too focused on phenotypic improvement rather than the molecular driver of the disease and could therefore be further refined by integrating the leap of molecular and cellular knowledge. The ambiguity of the ejection fraction-based classification criteria became evident with development of advanced molecular techniques and the dawn of omics disciplines which introduced the idea that disease is caused by a myriad of cellular and molecular processes rather than a single event or pathway. The fact that different signaling pathways may underlie similar clinical manifestations calls for a more holistic study of heart failure. In this context, the systems biology approach can offer a better understanding of how different components of a system are altered during disease and how they interact with each other, potentially leading to improved diagnosis and classification of this condition. This review is aimed at addressing heart failure through a multilayer approach that covers individually some of the anatomical, morphological, functional, and tissue aspects, with focus on cellular and subcellular features as an alternative insight into new therapeutic opportunities.
Collapse
Affiliation(s)
- Vijay Urmaliya
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium.
| | | |
Collapse
|
39
|
Polge C, Cabantous S, Deval C, Claustre A, Hauvette A, Bouchenot C, Aniort J, Béchet D, Combaret L, Attaix D, Taillandier D. A muscle-specific MuRF1-E2 network requires stabilization of MuRF1-E2 complexes by telethonin, a newly identified substrate. J Cachexia Sarcopenia Muscle 2018; 9:129-145. [PMID: 29271608 PMCID: PMC5803617 DOI: 10.1002/jcsm.12249] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/21/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Muscle wasting is observed in the course of many diseases and also during physiological conditions (disuse, ageing). Skeletal muscle mass is largely controlled by the ubiquitin-proteasome system and thus by the ubiquitinating enzymes (E2s and E3s) that target substrates for subsequent degradation. MuRF1 is the only E3 ubiquitin ligase known to target contractile proteins (α-actin, myosins) during catabolic situations. However, MuRF1 depends on E2 ubiquitin-conjugating enzymes for ubiquitin chain formation on the substrates. MuRF1-E2 couples are therefore putative targets for preventing muscle wasting. METHODS We focused on 14 E2 enzymes that are either expressed in skeletal muscle or up-regulated during atrophying conditions. In this work, we demonstrated that only highly sensitive and complementary interactomic approaches (surface plasmon resonance, yeast three-hybrid, and split green fluorescent protein) allowed the identification of MuRF1 E2 partners. RESULTS Five E2 enzymes physically interacted with MuRF1, namely, E2E1, E2G1, E2J1, E2J2, and E2L3. Moreover, we demonstrated that MuRF1-E2E1 and MuRF1-E2J1 interactions are facilitated by telethonin, a newly identified MuRF1 substrate. We next showed that the five identified E2s functionally interacted with MuRF1 since, in contrast to the non-interacting E2D2, their co-expression in HEK293T cells with MuRF1 led to increased telethonin degradation. Finally, we showed that telethonin governed the affinity between MuRF1 and E2E1 or E2J1. CONCLUSIONS We report here the first MuRF1-E2s network, which may prove valuable for deciphering the precise mechanisms involved in the atrophying muscle programme and for proposing new therapeutical approaches.
Collapse
Affiliation(s)
- Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Stéphanie Cabantous
- Cancer Research Center of Toulouse, INSERM UMR 1037F‐31037ToulouseFrance
- Université de ToulouseF‐31062ToulouseFrance
| | - Christiane Deval
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Agnès Claustre
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Antoine Hauvette
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Catherine Bouchenot
- Cancer Research Center of Toulouse, INSERM UMR 1037F‐31037ToulouseFrance
- Université de ToulouseF‐31062ToulouseFrance
| | - Julien Aniort
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
- Service de Néphrologie Réanimation Médicale, Pôle Respiratoire, Endocrinologie‐Diabétologie, Urologie, Néphrologie‐Dialyse, Nutrition Clinique, InfectiologieRéanimation Médicale, Hygiène Hospitalière (REUNNIRH)F‐63000Clermont‐FerrandFrance
| | - Daniel Béchet
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Lydie Combaret
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Didier Attaix
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| | - Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition HumaineCRNH AuvergneF‐63000Clermont‐FerrandFrance
| |
Collapse
|
40
|
Taatjes DJ, Roth J. In focus in HCB. Histochem Cell Biol 2017; 148:473-475. [PMID: 28936549 DOI: 10.1007/s00418-017-1609-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA.
| | - Jürgen Roth
- University of Zurich, 8091, Zurich, Switzerland
| |
Collapse
|
41
|
Abstract
Hypertension is the leading risk factor for heart disease and stroke, and is estimated to cause 9.4 million deaths globally every year. The pathogenesis of hypertension is complex, but lifestyle factors such as diet are important contributors to the disease. High dietary intake of fruit and vegetables is associated with reduced blood pressure and lower cardiovascular mortality. A critical relationship between dietary intake and the composition of the gut microbiota has been described in the literature, and a growing body of evidence supports the role of the gut microbiota in the regulation of blood pressure. In this Review, we describe the mechanisms by which the gut microbiota and its metabolites, including short-chain fatty acids, trimethylamine N-oxide, and lipopolysaccharides, act on downstream cellular targets to prevent or contribute to the pathogenesis of hypertension. These effects have a direct influence on tissues such as the kidney, the endothelium, and the heart. Finally, we consider the role of the gut microbiota in resistant hypertension, the possible intergenerational effect of the gut microbiota on blood pressure regulation, and the promising therapeutic potential of gut microbiota modification to improve health and prevent disease.
Collapse
Affiliation(s)
- Francine Z Marques
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Department of Pharmacology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Wellington Road, Clayton Victoria 3800, Australia
| | - Charles R Mackay
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, 23 Innovation Walk, Clayton, Victoria 3800, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Heart Centre, Alfred Hospital, Philip Block, Level 3, 55 Commercial Road, Melbourne, Victoria 3004, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, 99 Commercial Road, Melbourne, Victoria 3004, Australia
| |
Collapse
|
42
|
Zuppinger C, Gibbons G, Dutta-Passecker P, Segiser A, Most H, Suter TM. Characterization of cytoskeleton features and maturation status of cultured human iPSC-derived cardiomyocytes. Eur J Histochem 2017; 61:2763. [PMID: 28735524 PMCID: PMC5484009 DOI: 10.4081/ejh.2017.2763] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 06/07/2017] [Accepted: 06/07/2017] [Indexed: 01/01/2023] Open
Abstract
Recent innovations in stem cell technologies and the availability of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have opened new possibilities for studies and drug testing on human cardiomyocytes in vitro. Still, there are concerns about the precise nature of such 'reprogrammed' cells. We have performed an investigation using immunocytochemistry and confocal microscopy on several cellular features using commercially available hiPSC-CMs. For some selected developmentally regulated or cardiac chamber-specific proteins, we have compared the results from hiPSC-derived cardiomyocytes with freshly isolated, ventricular cardiomyocytes from adult rats. The results show that all typical cardiac proteins are expressed in these hiPSC-CMs. Furthermore, intercalated disc-like structures, calcium cycling proteins, and myofibrils are present. However, some of these proteins are only known from early developmental stages of the ventricular myocardium or the diseased adult heart. A heterogeneous expression pattern in the cell population was noted for some muscle proteins, such as for myosin light chains, or incomplete organization in sarcomeres, such as for telethonin. These observations indicate that hiPSC-CMs can be considered genuine human cardiomyocytes of an early developmental state. The here described marker proteins of maturation may become instrumental in future studies attempting the improvement of cardiomyocyte in vitro models.
Collapse
|
43
|
Ikenberg E, Karin I, Ertl-Wagner B, Abicht A, Bulst S, Krause S, Schoser B, Reilich P, Walter MC. Rare diagnosis of telethoninopathy (LGMD2G) in a Turkish patient. Neuromuscul Disord 2017; 27:856-860. [PMID: 28666572 DOI: 10.1016/j.nmd.2017.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/01/2017] [Accepted: 05/30/2017] [Indexed: 10/19/2022]
Abstract
Telethoninopathy is one of the rarest forms of Limb-girdle muscular dystrophy (LGMD). So far, only a small number of LGMD type 2 G (LGMD2G) patients have been described, mostly patients from Brazil. Here we present a 35-year-old female patient of Turkish ethnicity with LGMD2G due to a novel homozygous frame-shift mutation c.90_91del (p.Ser31Hisfs*11) in the telethonin gene, probably leading to truncated protein or nonsense mediated decay. Myalgia and walking on tiptoes were the first symptoms starting in early childhood, around age 22 proximal, later distal leg muscles became affected. Muscle biopsy showed a degenerative myopathy with lobulated fibers, creatine kinase levels were elevated to 1200 U/l. No cardiomyopathy has been detected but ventricular extrasystoles were treated with verapamil. Even though telethoninopathy represents a rare condition, testing for LGMD2G should be included into the diagnostic work-up of mild myopathies with early toe walking and distal and proximal involvement.
Collapse
Affiliation(s)
- Elena Ikenberg
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Ivan Karin
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Birgit Ertl-Wagner
- Dept. of Radiology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Angela Abicht
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany; Medical Genetics Center - MGZ, Munich, Germany
| | | | - Sabine Krause
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Peter Reilich
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maggie C Walter
- Friedrich-Baur-Institute, Dept. of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany.
| |
Collapse
|
44
|
Genetic epidemiology of titin-truncating variants in the etiology of dilated cardiomyopathy. Biophys Rev 2017; 9:207-223. [PMID: 28510119 PMCID: PMC5498329 DOI: 10.1007/s12551-017-0265-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/10/2017] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome defined by the inability of the heart to pump enough blood to meet the body's metabolic demands. Major causes of HF are cardiomyopathies (diseases of the myocardium associated with mechanical and/or electrical dysfunction), among which the most common form is dilated cardiomyopathy (DCM). DCM is defined by ventricular chamber enlargement and systolic dysfunction with normal left ventricular wall thickness, which leads to progressive HF. Over 60 genes are linked to the etiology of DCM. Titin (TTN) is the largest known protein in biology, spanning half the cardiac sarcomere and, as such, is a basic structural and functional unit of striated muscles. It is essential for heart development as well as mechanical and regulatory functions of the sarcomere. Next-generation sequencing (NGS) in clinical DCM cohorts implicated truncating variants in titin (TTNtv) as major disease alleles, accounting for more than 25% of familial DCM cases, but these variants have also been identified in 2-3% of the general population, where these TTNtv blur diagnostic and clinical utility. Taking into account the published TTNtv and their association to DCM, it becomes clear that TTNtv harm the heart with position-dependent occurrence, being more harmful when present in the A-band TTN, presumably with dominant negative/gain-of-function mechanisms. However, these insights are challenged by the depiction of position-independent toxicity of TTNtv acting via haploinsufficient alleles, which are sufficient to induce cardiac pathology upon stress. In the current review, we provide an overview of TTN and discuss studies investigating various TTN mutations. We also present an overview of different mechanisms postulated or experimentally validated in the pathogenicity of TTNtv. DCM-causing genes are also discussed with respect to non-truncating mutations in the etiology of DCM. One way of understanding pathogenic variants is probably to understand the context in which they may or may not affect protein-protein interactions, changes in cell signaling, and substrate specificity. In this regard, we also provide a brief overview of TTN interactions in situ. Quantitative models in the risk assessment of TTNtv are also discussed. In summary, we highlight the importance of gene-environment interactions in the etiology of DCM and further mechanistic studies used to delineate the pathways which could be targeted in the management of DCM.
Collapse
|
45
|
Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann M, Tan JK, Kuruppu S, Rajapakse NW, El-Osta A, Mackay CR, Kaye DM. High-Fiber Diet and Acetate Supplementation Change the Gut Microbiota and Prevent the Development of Hypertension and Heart Failure in Hypertensive Mice. Circulation 2017; 135:964-977. [DOI: 10.1161/circulationaha.116.024545] [Citation(s) in RCA: 718] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/29/2016] [Indexed: 01/11/2023]
Abstract
Background:
Dietary intake of fruit and vegetables is associated with lower incidence of hypertension, but the mechanisms involved have not been elucidated. Here, we evaluated the effect of a high-fiber diet and supplementation with the short-chain fatty acid acetate on the gut microbiota and the prevention of cardiovascular disease.
Methods:
Gut microbiome, cardiorenal structure/function, and blood pressure were examined in sham and mineralocorticoid excess–treated mice with a control diet, high-fiber diet, or acetate supplementation. We also determined the renal and cardiac transcriptome of mice treated with the different diets.
Results:
We found that high consumption of fiber modified the gut microbiota populations and increased the abundance of acetate-producing bacteria independently of mineralocorticoid excess. Both fiber and acetate decreased gut dysbiosis, measured by the ratio of Firmicutes to Bacteroidetes, and increased the prevalence of
Bacteroides acidifaciens
. Compared with mineralocorticoid-excess mice fed a control diet, both high-fiber diet and acetate supplementation significantly reduced systolic and diastolic blood pressures, cardiac fibrosis, and left ventricular hypertrophy. Acetate had similar effects and markedly reduced renal fibrosis. Transcriptome analyses showed that the protective effects of high fiber and acetate were accompanied by the downregulation of cardiac and renal
Egr1
, a master cardiovascular regulator involved in cardiac hypertrophy, cardiorenal fibrosis, and inflammation. We also observed the upregulation of a network of genes involved in circadian rhythm in both tissues and downregulation of the renin-angiotensin system in the kidney and mitogen-activated protein kinase signaling in the heart.
Conclusions:
A diet high in fiber led to changes in the gut microbiota that played a protective role in the development of cardiovascular disease. The favorable effects of fiber may be explained by the generation and distribution of one of the main metabolites of the gut microbiota, the short-chain fatty acid acetate. Acetate effected several molecular changes associated with improved cardiovascular health and function.
Collapse
Affiliation(s)
- Francine Z. Marques
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Erin Nelson
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Po-Yin Chu
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Duncan Horlock
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - April Fiedler
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Mark Ziemann
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Jian K. Tan
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Sanjaya Kuruppu
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Niwanthi W. Rajapakse
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Assam El-Osta
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - Charles R. Mackay
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| | - David M. Kaye
- From Heart Failure Research Group (F.Z.M., E.N., P.-Y.C., D.H., A.F., N.W.R., D.M.K.) and Human Epigenetics Group (M.Z., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Australia; Central Clinical School, Faculty of Medicine Nursing and Health Sciences (F.Z.M., A.E.-O., D.M.K.), Infection and Immunity Program, Monash Biomedicine Discovery Institute (J.K.T., C.R.M.), Department of Biochemistry and Molecular Biology (J.K.T., S.K., C.R.M.), and Department of Physiology (N.W.R.), Monash
| |
Collapse
|
46
|
Gautel M, Djinović-Carugo K. The sarcomeric cytoskeleton: from molecules to motion. ACTA ACUST UNITED AC 2016; 219:135-45. [PMID: 26792323 DOI: 10.1242/jeb.124941] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Highly ordered organisation of striated muscle is the prerequisite for the fast and unidirectional development of force and motion during heart and skeletal muscle contraction. A group of proteins, summarised as the sarcomeric cytoskeleton, is essential for the ordered assembly of actin and myosin filaments into sarcomeres, by combining architectural, mechanical and signalling functions. This review discusses recent cell biological, biophysical and structural insight into the regulated assembly of sarcomeric cytoskeleton proteins and their roles in dissipating mechanical forces in order to maintain sarcomere integrity during passive extension and active contraction. α-Actinin crosslinks in the Z-disk show a pivot-and-rod structure that anchors both titin and actin filaments. In contrast, the myosin crosslinks formed by myomesin in the M-band are of a ball-and-spring type and may be crucial in providing stable yet elastic connections during active contractions, especially eccentric exercise.
Collapse
Affiliation(s)
- Mathias Gautel
- King's College London BHF Centre of Research Excellence, Randall Division for Cell and Molecular Biophysics, and Cardiovascular Division, New Hunt's House, London SE1 1UL, UK
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, Vienna A-1030, Austria Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Aškerčeva 5, Ljubljana 1000, Slovenia
| |
Collapse
|
47
|
Hastings R, de Villiers CP, Hooper C, Ormondroyd L, Pagnamenta A, Lise S, Salatino S, Knight SJL, Taylor JC, Thomson KL, Arnold L, Chatziefthimiou SD, Konarev PV, Wilmanns M, Ehler E, Ghisleni A, Gautel M, Blair E, Watkins H, Gehmlich K. Combination of Whole Genome Sequencing, Linkage, and Functional Studies Implicates a Missense Mutation in Titin as a Cause of Autosomal Dominant Cardiomyopathy With Features of Left Ventricular Noncompaction. ACTA ACUST UNITED AC 2016; 9:426-435. [PMID: 27625337 PMCID: PMC5068189 DOI: 10.1161/circgenetics.116.001431] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/31/2016] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Background— High throughput next-generation sequencing techniques have made whole genome sequencing accessible in clinical practice; however, the abundance of variation in the human genomes makes the identification of a disease-causing mutation on a background of benign rare variants challenging. Methods and Results— Here we combine whole genome sequencing with linkage analysis in a 3-generation family affected by cardiomyopathy with features of autosomal dominant left ventricular noncompaction cardiomyopathy. A missense mutation in the giant protein titin is the only plausible disease-causing variant that segregates with disease among the 7 surviving affected individuals, with interrogation of the entire genome excluding other potential causes. This A178D missense mutation, affecting a conserved residue in the second immunoglobulin-like domain of titin, was introduced in a bacterially expressed recombinant protein fragment and biophysically characterized in comparison to its wild-type counterpart. Multiple experiments, including size exclusion chromatography, small-angle x ray scattering, and circular dichroism spectroscopy suggest partial unfolding and domain destabilization in the presence of the mutation. Moreover, binding experiments in mammalian cells show that the mutation markedly impairs binding to the titin ligand telethonin. Conclusions— Here we present genetic and functional evidence implicating the novel A178D missense mutation in titin as the cause of a highly penetrant familial cardiomyopathy with features of left ventricular noncompaction. This expands the spectrum of titin’s roles in cardiomyopathies. It furthermore highlights that rare titin missense variants, currently often ignored or left uninterpreted, should be considered to be relevant for cardiomyopathies and can be identified by the approach presented here.
Collapse
|
48
|
Bang ML. Animal Models of Congenital Cardiomyopathies Associated With Mutations in Z-Line Proteins. J Cell Physiol 2016; 232:38-52. [PMID: 27171814 DOI: 10.1002/jcp.25424] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/10/2016] [Indexed: 01/15/2023]
Abstract
The cardiac Z-line at the boundary between sarcomeres is a multiprotein complex connecting the contractile apparatus with the cytoskeleton and the extracellular matrix. The Z-line is important for efficient force generation and transmission as well as the maintenance of structural stability and integrity. Furthermore, it is a nodal point for intracellular signaling, in particular mechanosensing and mechanotransduction. Mutations in various genes encoding Z-line proteins have been associated with different cardiomyopathies, including dilated cardiomyopathy, hypertrophic cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, restrictive cardiomyopathy, and left ventricular noncompaction, and mutations even within the same gene can cause widely different pathologies. Animal models have contributed to a great advancement in the understanding of the physiological function of Z-line proteins and the pathways leading from mutations in Z-line proteins to cardiomyopathy, although genotype-phenotype prediction remains a great challenge. This review presents an overview of the currently available animal models for Z-line and Z-line associated proteins involved in human cardiomyopathies with special emphasis on knock-in and transgenic mouse models recapitulating the clinical phenotypes of human cardiomyopathy patients carrying mutations in Z-line proteins. Pros and cons of mouse models will be discussed and a future outlook will be given. J. Cell. Physiol. 232: 38-52, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council and Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
49
|
Krüger M, Kötter S. Titin, a Central Mediator for Hypertrophic Signaling, Exercise-Induced Mechanosignaling and Skeletal Muscle Remodeling. Front Physiol 2016; 7:76. [PMID: 26973541 PMCID: PMC4771757 DOI: 10.3389/fphys.2016.00076] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/15/2016] [Indexed: 01/09/2023] Open
Abstract
Titin is a giant scaffold protein with multiple functions in striated muscle physiology. Due to the elastic I-band domains and the filament-like integration in the half-sarcomere titin is an important factor for sarcomere assembly and serves as an adaptable molecular spring that determines myofilament distensibility. Protein-interactions e.g., with muscle ankyrin repeat proteins or muscle LIM-protein link titin to hypertrophic signaling and via p62 and Muscle Ring Finger proteins to mechanisms that control protein quality control. This review summarizes our current knowledge on titin as a central node for exercise-induced mechanosignaling and remodeling and further highlights the pathophysiological implications.
Collapse
Affiliation(s)
- Martina Krüger
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf Düsseldorf, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Heinrich Heine University Düsseldorf Düsseldorf, Germany
| |
Collapse
|
50
|
Bodmer NK, Theisen KE, Dima RI. Molecular investigations into the mechanics of a muscle anchoring complex. Biophys J 2016; 108:2322-32. [PMID: 25954889 DOI: 10.1016/j.bpj.2015.03.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/16/2015] [Accepted: 03/13/2015] [Indexed: 11/15/2022] Open
Abstract
The titin-telethonin complex, essential for anchoring filaments in the Z-disk of the sarcomere, is composed of immunoglobulin domains. Surprisingly, atomic force microscopy experiments showed that it resists forces much higher than the typical immunoglobulin domain and that the force distribution is unusually broad. To investigate the origin of this behavior, we developed a multiscale simulation approach, combining minimalist and atomistic models (SOP-AT). By following the mechanical response of the complex on experimental timescales, we found that the mechanical stability of titin-telethonin is modulated primarily by the strength of contacts between telethonin and the two titin chains, and secondarily by the timescales of conformational excursions inside telethonin and the pulled titin domains. Importantly, the conformational transitions executed by telethonin in simulations support its proposed role in mechanosensing. Our SOP-AT computational approach thus provides a powerful tool for the exploration of the link between conformational diversity and the broadness of the mechanical response, which can be applied to other multidomain complexes.
Collapse
Affiliation(s)
| | - Kelly E Theisen
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio
| | - Ruxandra I Dima
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|