1
|
Long J, Liu W, Fan X, Yang Y, Yang X, Tang Z. A comprehensive atlas of pig RNA editome across 23 tissues reveals RNA editing affecting interaction mRNA-miRNAs. G3 (BETHESDA, MD.) 2024; 14:jkae178. [PMID: 39090686 PMCID: PMC11457091 DOI: 10.1093/g3journal/jkae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 04/30/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
RNA editing is a co-transcriptional/post-transcriptional modification that is mediated by the ADAR enzyme family. Profiling of RNA editing is very limited in pigs. In this study, we collated 3813 RNA-seq data from the public repositories across 23 tissues and carried out comprehensive profiling of RNA editing in pigs. In total, 127,927 A-to-I RNA-editing sites were detected. Our analysis showed that 98.2% of RNA-editing sites were located within repeat regions, primarily within the pig-specific SINE retrotransposon PRE-1/Pre0_SS elements. Subsequently, we focused on analyzing specific RNA-editing sites (SESs) in skeletal muscle tissues. Functional enrichment analyses suggested that they were enriched in signaling pathways associated with muscle cell differentiation, including DMD, MYOD1, and CAV1 genes. Furthermore, we discovered that RNA editing event in the 3'UTR of CFLAR mRNA influenced miR-708-5p binding in this region. In this study, the panoramic RNA-editing landscape of different tissues of pigs was systematically mapped, and RNA-editing sites and genes involved in muscle cell differentiation were identified. In summary, we identified modifications to pig RNA-editing sites and provided candidate targets for further validation.
Collapse
Affiliation(s)
- Jiajia Long
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning, Guangxi 530004, China
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Weiwei Liu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning, Guangxi 530004, China
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xinhao Fan
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yalan Yang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xiaogan Yang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Zhonglin Tang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science & Technology, Guangxi University, Nanning, Guangxi 530004, China
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
2
|
Miller MJ, Gries KJ, Marcotte GR, Ryan Z, Strub MD, Kunz HE, Arendt BK, Dasari S, Ebert SM, Adams CM, Lanza IR. Human myofiber-enriched aging-induced lncRNA FRAIL1 promotes loss of skeletal muscle function. Aging Cell 2024; 23:e14097. [PMID: 38297807 PMCID: PMC11019130 DOI: 10.1111/acel.14097] [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: 10/05/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 02/02/2024] Open
Abstract
The loss of skeletal muscle mass during aging is a significant health concern linked to adverse outcomes in older individuals. Understanding the molecular basis of age-related muscle loss is crucial for developing strategies to combat this debilitating condition. Long noncoding RNAs (lncRNAs) are a largely uncharacterized class of biomolecules that have been implicated in cellular homeostasis and dysfunction across a many tissues and cell types. To identify lncRNAs that might contribute to skeletal muscle aging, we screened for lncRNAs whose expression was altered in vastus lateralis muscle from older compared to young adults. We identified FRAIL1 as an aging-induced lncRNA with high abundance in human skeletal muscle. In healthy young and older adults, skeletal muscle FRAIL1 was increased with age in conjunction with lower muscle function. Forced expression of FRAIL1 in mouse tibialis anterior muscle elicits a dose-dependent reduction in skeletal muscle fiber size that is independent of changes in muscle fiber type. Furthermore, this reduction in muscle size is dependent on an intact region of FRAIL1 that is highly conserved across non-human primates. Unbiased transcriptional and proteomic profiling of the effects of FRAIL1 expression in mouse skeletal muscle revealed widespread changes in mRNA and protein abundance that recapitulate age-related changes in pathways and processes that are known to be altered in aging skeletal muscle. Taken together, these findings shed light on the intricate molecular mechanisms underlying skeletal muscle aging and implicate FRAIL1 in age-related skeletal muscle phenotypes.
Collapse
Affiliation(s)
- Matthew J. Miller
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- University of IowaIowa CityIowaUSA
| | | | - George R. Marcotte
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- University of IowaIowa CityIowaUSA
| | - Zachary Ryan
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | | | - Hawley E. Kunz
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | | | - Surendra Dasari
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Scott M. Ebert
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Emmyon, Inc.RochesterMinnesotaUSA
| | - Christopher M. Adams
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Emmyon, Inc.RochesterMinnesotaUSA
| | - Ian R. Lanza
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
3
|
Nie Z, Guo N, Peng Y, Gao Y, Cao H, Zhang S. Duality of the SVIL expression in bladder cancer and its correlation with immune infiltration. Sci Rep 2023; 13:14595. [PMID: 37670039 PMCID: PMC10480233 DOI: 10.1038/s41598-023-41759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023] Open
Abstract
SVIL is a member of the villin/gelsolin superfamily and is responsible for encoding supervillin. It has been reported to be closely related to the occurrence and development of various tumors. However, the mechanism of SVIL in bladder cancer has not been reported yet. In this research, we evaluated the relationship between SVIL expression and bladder cancer in public dataset and examined the expression of SVIL in bladder cancer cell lines, tissue microarrays and patients in our cohort. Our work determined that the expression of SVIL in bladder cancer tissue was significantly lower than that in normal tissue. However, in bladder cancer tissues, the high expression of SVIL is significantly associated with poor prognosis. This kind of duality is very novel and has great research value. The expression level of SVIL can well predict the survival time of bladder cancer patients, and is an independent risk factor of bladder cancer patients. The expression of SVIL is also closely related to the immune tumor microenvironment of bladder cancer. Our research provides a basis for personalized therapeutic targets for bladder cancer.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China.
| |
Collapse
|
4
|
Chumakova OS, Baulina NM. Advanced searching for hypertrophic cardiomyopathy heritability in real practice tomorrow. Front Cardiovasc Med 2023; 10:1236539. [PMID: 37583586 PMCID: PMC10425241 DOI: 10.3389/fcvm.2023.1236539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease associated with morbidity and mortality at any age. As studies in recent decades have shown, the genetic architecture of HCM is quite complex both in the entire population and in each patient. In the rapidly advancing era of gene therapy, we have to provide a detailed molecular diagnosis to our patients to give them the chance for better and more personalized treatment. In addition to emphasizing the importance of genetic testing in routine practice, this review aims to discuss the possibility to go a step further and create an expanded genetic panel that contains not only variants in core genes but also new candidate genes, including those located in deep intron regions, as well as structural variations. It also highlights the benefits of calculating polygenic risk scores based on a combination of rare and common genetic variants for each patient and of using non-genetic HCM markers, such as microRNAs that can enhance stratification of risk for HCM in unselected populations alongside rare genetic variants and clinical factors. While this review is focusing on HCM, the discussed issues are relevant to other cardiomyopathies.
Collapse
Affiliation(s)
- Olga S. Chumakova
- Laboratory of Functional Genomics of Cardiovascular Diseases, National Medical Research Centre of Cardiology Named After E.I. Chazov, Moscow, Russia
| | | |
Collapse
|
5
|
Gibertini S, Ruggieri A, Cheli M, Maggi L. Protein Aggregates and Aggrephagy in Myopathies. Int J Mol Sci 2023; 24:ijms24098456. [PMID: 37176163 PMCID: PMC10179229 DOI: 10.3390/ijms24098456] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
A number of muscular disorders are hallmarked by the aggregation of misfolded proteins within muscle fibers. A specialized form of macroautophagy, termed aggrephagy, is designated to remove and degrade protein aggregates. This review aims to summarize what has been studied so far about the direct involvement of aggrephagy and the activation of the key players, among others, p62, NBR1, Alfy, Tollip, Optineurin, TAX1BP1 and CCT2 in muscular diseases. In the first part of the review, we describe the aggrephagy pathway with the involved proteins; then, we illustrate the muscular disorder histologically characterized by protein aggregates, highlighting the role of aggrephagy pathway abnormalities in these muscular disorders.
Collapse
Affiliation(s)
- Sara Gibertini
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Alessandra Ruggieri
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Marta Cheli
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", 20133 Milan, Italy
| |
Collapse
|
6
|
Tadros R, Zheng SL, Grace C, Jordà P, Francis C, Jurgens SJ, Thomson KL, Harper AR, Ormondroyd E, West DM, Xu X, Theotokis PI, Buchan RJ, McGurk KA, Mazzarotto F, Boschi B, Pelo E, Lee M, Noseda M, Varnava A, Vermeer AM, Walsh R, Amin AS, van Slegtenhorst MA, Roslin N, Strug LJ, Salvi E, Lanzani C, de Marvao A, Roberts JD, Tremblay-Gravel M, Giraldeau G, Cadrin-Tourigny J, L'Allier PL, Garceau P, Talajic M, Pinto YM, Rakowski H, Pantazis A, Baksi J, Halliday BP, Prasad SK, Barton PJ, O'Regan DP, Cook SA, de Boer RA, Christiaans I, Michels M, Kramer CM, Ho CY, Neubauer S, Matthews PM, Wilde AA, Tardif JC, Olivotto I, Adler A, Goel A, Ware JS, Bezzina CR, Watkins H. Large scale genome-wide association analyses identify novel genetic loci and mechanisms in hypertrophic cardiomyopathy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.28.23285147. [PMID: 36778260 PMCID: PMC9915807 DOI: 10.1101/2023.01.28.23285147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is an important cause of morbidity and mortality with both monogenic and polygenic components. We here report results from the largest HCM genome-wide association study (GWAS) and multi-trait analysis (MTAG) including 5,900 HCM cases, 68,359 controls, and 36,083 UK Biobank (UKB) participants with cardiac magnetic resonance (CMR) imaging. We identified a total of 70 loci (50 novel) associated with HCM, and 62 loci (32 novel) associated with relevant left ventricular (LV) structural or functional traits. Amongst the common variant HCM loci, we identify a novel HCM disease gene, SVIL, which encodes the actin-binding protein supervillin, showing that rare truncating SVIL variants cause HCM. Mendelian randomization analyses support a causal role of increased LV contractility in both obstructive and non-obstructive forms of HCM, suggesting common disease mechanisms and anticipating shared response to therapy. Taken together, the findings significantly increase our understanding of the genetic basis and molecular mechanisms of HCM, with potential implications for disease management.
Collapse
Affiliation(s)
- Rafik Tadros
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Sean L Zheng
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Christopher Grace
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Paloma Jordà
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Catherine Francis
- National Heart & Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kate L Thomson
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Oxford Genetics Laboratories, Churchill Hospital, Oxford, UK
| | - Andrew R Harper
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elizabeth Ormondroyd
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Dominique M West
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Xiao Xu
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Pantazis I Theotokis
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Rachel J Buchan
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Kathryn A McGurk
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Francesco Mazzarotto
- National Heart & Lung Institute, Imperial College London, London, UK
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | - Michael Lee
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Michela Noseda
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Amanda Varnava
- National Heart & Lung Institute, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Alexa Mc Vermeer
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Genetics, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, (ERN GUARD-HEART; https://guardheart.ern-net.eu)
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, (ERN GUARD-HEART; https://guardheart.ern-net.eu)
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Nicole Roslin
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lisa J Strug
- Departments of Statistical Sciences and Computer Science, Data Sciences Institute, University of Toronto, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Ontario Regional Centre, Canadian Statistical Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Lanzani
- Genomics of Renal Diseases and Hypertension Unit, Nephrology Operative Unit, IRCCS San Raffaele Hospital, Milan, Italy
- Chair of Nephrology, Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio de Marvao
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada
| | - Maxime Tremblay-Gravel
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Genevieve Giraldeau
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Philippe L L'Allier
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Patrick Garceau
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Mario Talajic
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, (ERN GUARD-HEART; https://guardheart.ern-net.eu)
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Antonis Pantazis
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - John Baksi
- National Heart & Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Brian P Halliday
- National Heart & Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sanjay K Prasad
- National Heart & Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul Jr Barton
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Declan P O'Regan
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Stuart A Cook
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- National Heart Centre Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore
| | - Rudolf A de Boer
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Imke Christiaans
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michelle Michels
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, (ERN GUARD-HEART; https://guardheart.ern-net.eu)
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Christopher M Kramer
- Department of Medicine, Cardiovascular Division, University of Virginia Health, Charlottesville, VA, USA
| | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, NIHR Oxford Health Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Paul M Matthews
- Department of Brain Sciences and UK Dementia Research Institute, Imperial College London, London, UK
| | - Arthur A Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, (ERN GUARD-HEART; https://guardheart.ern-net.eu)
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- ECGen, Cardiogenetics Focus Group of EHRA, France
| | - Jean-Claude Tardif
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, Meyer Children Hospital, University of Florence, Florence, Italy
| | - Arnon Adler
- Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anuj Goel
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - James S Ware
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Program in Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, (ERN GUARD-HEART; https://guardheart.ern-net.eu)
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Zhao M, Banhos Danneskiold-Samsøe N, Ulicna L, Nguyen Q, Voilquin L, Lee DE, White JP, Jiang Z, Cuthbert N, Paramasivam S, Bielczyk-Maczynska E, Van Rechem C, Svensson KJ. Phosphoproteomic mapping reveals distinct signaling actions and activation of muscle protein synthesis by Isthmin-1. eLife 2022; 11:e80014. [PMID: 36169399 PMCID: PMC9592085 DOI: 10.7554/elife.80014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
The secreted protein isthmin-1 (Ism1) mitigates diabetes by increasing adipocyte and skeletal muscle glucose uptake by activating the PI3K-Akt pathway. However, while both Ism1 and insulin converge on these common targets, Ism1 has distinct cellular actions suggesting divergence in downstream intracellular signaling pathways. To understand the biological complexity of Ism1 signaling, we performed phosphoproteomic analysis after acute exposure, revealing overlapping and distinct pathways of Ism1 and insulin. We identify a 53% overlap between Ism1 and insulin signaling and Ism1-mediated phosphoproteome-wide alterations in ~450 proteins that are not shared with insulin. Interestingly, we find several unknown phosphorylation sites on proteins related to protein translation, mTOR pathway, and, unexpectedly, muscle function in the Ism1 signaling network. Physiologically, Ism1 ablation in mice results in altered proteostasis, including lower muscle protein levels under fed and fasted conditions, reduced amino acid incorporation into proteins, and reduced phosphorylation of the key protein synthesis effectors Akt and downstream mTORC1 targets. As metabolic disorders such as diabetes are associated with accelerated loss of skeletal muscle protein content, these studies define a non-canonical mechanism by which this antidiabetic circulating protein controls muscle biology.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| | | | - Livia Ulicna
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Quennie Nguyen
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Laetitia Voilquin
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| | - David E Lee
- Duke Molecular Physiology Institute, Duke University School of MedicineDurhamUnited States
- Department of Medicine, Duke University School of MedicineDurhamUnited States
| | - James P White
- Duke Molecular Physiology Institute, Duke University School of MedicineDurhamUnited States
- Department of Medicine, Duke University School of MedicineDurhamUnited States
- Duke Center for the Study of Aging and Human Development, Duke University School of MedicineDurhamUnited States
| | - Zewen Jiang
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Department of Laboratory Medicine, University of California, San FranciscoSan FranciscoUnited States
- Diabetes Center, University of California, San FranciscoSan FranciscoUnited States
| | - Nickeisha Cuthbert
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Shrika Paramasivam
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Ewa Bielczyk-Maczynska
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of MedicineStanfordUnited States
| | - Capucine Van Rechem
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
8
|
Zhang J, Wen H, Qi X, Zhang Y, Dong X, Zhang K, Zhang M, Li J, Li Y. Morphological and Molecular Responses of Lateolabrax maculatus Skeletal Muscle Cells to Different Temperatures. Int J Mol Sci 2022; 23:ijms23179812. [PMID: 36077203 PMCID: PMC9456278 DOI: 10.3390/ijms23179812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
Temperature strongly modulates muscle development and growth in ectothermic teleosts; however, the underlying mechanisms remain largely unknown. In this study, primary cultures of skeletal muscle cells of Lateolabrax maculatus were conducted and reared at different temperatures (21, 25, and 28 °C) in both the proliferation and differentiation stages. CCK-8, EdU, wound scratch and nuclear fusion index assays revealed that the proliferation, myogenic differentiation, and migration processes of skeletal muscle cells were significantly accelerated as the temperature raises. Based on the GO, GSEA, and WGCNA, higher temperature (28 °C) induced genes involved in HSF1 activation, DNA replication, and ECM organization processes at the proliferation stage, as well as HSF1 activation, calcium activity regulation, myogenic differentiation, and myoblast fusion, and sarcomere assembly processes at the differentiation stage. In contrast, lower temperature (21 °C) increased the expression levels of genes associated with DNA damage, DNA repair and apoptosis processes at the proliferation stage, and cytokine signaling and neutrophil degranulation processes at the differentiation stage. Additionally, we screened several hub genes regulating myogenesis processes. Our results could facilitate the understanding of the regulatory mechanism of temperature on fish skeletal muscle growth and further contribute to utilizing rational management strategies and promoting organism growth and development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yun Li
- Correspondence: ; Tel.: +86-0532-82-031-792
| |
Collapse
|
9
|
Damoiseaux J, Mammen AL, Piette Y, Benveniste O, Allenbach Y. 256 th ENMC international workshop: Myositis specific and associated autoantibodies (MSA-ab): Amsterdam, The Netherlands, 8-10 October 2021. Neuromuscul Disord 2022; 32:594-608. [PMID: 35644723 DOI: 10.1016/j.nmd.2022.05.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Andrew L Mammen
- Muscle Disease Unit. Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Yves Piette
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium; Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical Immunology, Sorbonne Université, INSERM, UMR974, Pitié-Salpêtrière University Hospital, Paris, France
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Sorbonne Université, INSERM, UMR974, Pitié-Salpêtrière University Hospital, Paris, France
| |
Collapse
|
10
|
Aung N, Vargas JD, Yang C, Fung K, Sanghvi MM, Piechnik SK, Neubauer S, Manichaikul A, Rotter JI, Taylor KD, Lima JAC, Bluemke DA, Kawut SM, Petersen SE, Munroe PB. Genome-wide association analysis reveals insights into the genetic architecture of right ventricular structure and function. Nat Genet 2022; 54:783-791. [PMID: 35697868 DOI: 10.1038/s41588-022-01083-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/22/2022] [Indexed: 01/03/2023]
Abstract
Right ventricular (RV) structure and function influence the morbidity and mortality from coronary artery disease (CAD), dilated cardiomyopathy (DCM), pulmonary hypertension and heart failure. Little is known about the genetic basis of RV measurements. Here we perform genome-wide association analyses of four clinically relevant RV phenotypes (RV end-diastolic volume, RV end-systolic volume, RV stroke volume, RV ejection fraction) from cardiovascular magnetic resonance images, using a state-of-the-art deep learning algorithm in 29,506 UK Biobank participants. We identify 25 unique loci associated with at least one RV phenotype at P < 2.27 ×10-8, 17 of which are validated in a combined meta-analysis (n = 41,830). Several candidate genes overlap with Mendelian cardiomyopathy genes and are involved in cardiac muscle contraction and cellular adhesion. The RV polygenic risk scores (PRSs) are associated with DCM and CAD. The findings substantially advance our understanding of the genetic underpinning of RV measurements.
Collapse
Affiliation(s)
- Nay Aung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Jose D Vargas
- Veterans Affairs Medical Center, Washington, DC, USA.,Georgetown University, Washington, DC, USA
| | - Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Kenneth Fung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Mihir M Sanghvi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Steffen E Petersen
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK. .,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK. .,Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK.
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK. .,National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.
| |
Collapse
|
11
|
Stachowski-Doll MJ, Papadaki M, Martin TG, Ma W, Gong HM, Shao S, Shen S, Muntu NA, Kumar M, Perez E, Martin JL, Moravec CS, Sadayappan S, Campbell SG, Irving T, Kirk JA. GSK-3β Localizes to the Cardiac Z-Disc to Maintain Length Dependent Activation. Circ Res 2022; 130:871-886. [PMID: 35168370 PMCID: PMC8930626 DOI: 10.1161/circresaha.121.319491] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 02/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Altered kinase localization is gaining appreciation as a mechanism of cardiovascular disease. Previous work suggests GSK-3β (glycogen synthase kinase 3β) localizes to and regulates contractile function of the myofilament. We aimed to discover GSK-3β's in vivo role in regulating myofilament function, the mechanisms involved, and the translational relevance. METHODS Inducible cardiomyocyte-specific GSK-3β knockout mice and left ventricular myocardium from nonfailing and failing human hearts were studied. RESULTS Skinned cardiomyocytes from knockout mice failed to exhibit calcium sensitization with stretch indicating a loss of length-dependent activation (LDA), the mechanism underlying the Frank-Starling Law. Titin acts as a length sensor for LDA, and knockout mice had decreased titin stiffness compared with control mice, explaining the lack of LDA. Knockout mice exhibited no changes in titin isoforms, titin phosphorylation, or other thin filament phosphorylation sites known to affect passive tension or LDA. Mass spectrometry identified several z-disc proteins as myofilament phospho-substrates of GSK-3β. Agreeing with the localization of its targets, GSK-3β that is phosphorylated at Y216 binds to the z-disc. We showed pY216 was necessary and sufficient for z-disc binding using adenoviruses for wild-type, Y216F, and Y216E GSK-3β in neonatal rat ventricular cardiomyocytes. One of GSK-3β's z-disc targets, abLIM-1 (actin-binding LIM protein 1), binds to the z-disc domains of titin that are important for maintaining passive tension. Genetic knockdown of abLIM-1 via siRNA in human engineered heart tissues resulted in enhancement of LDA, indicating abLIM-1 may act as a negative regulator that is modulated by GSK-3β. Last, GSK-3β myofilament localization was reduced in left ventricular myocardium from failing human hearts, which correlated with depressed LDA. CONCLUSIONS We identified a novel mechanism by which GSK-3β localizes to the myofilament to modulate LDA. Importantly, z-disc GSK-3β levels were reduced in patients with heart failure, indicating z-disc localized GSK-3β is a possible therapeutic target to restore the Frank-Starling mechanism in patients with heart failure.
Collapse
Affiliation(s)
- Marisa J Stachowski-Doll
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Thomas G Martin
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Weikang Ma
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Henry M Gong
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Stephanie Shao
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
| | - Shi Shen
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
| | - Nitha Aima Muntu
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung, and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Edith Perez
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Jody L Martin
- Department of Pharmacology, Cardiovascular Research Institute, UC Davis School of Medicine, CA (J.L.M.)
| | - Christine S Moravec
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, OH (C.S.M.)
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung, and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Stuart G Campbell
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT (S.G.C.)
| | - Thomas Irving
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| |
Collapse
|
12
|
Smith TC, Vasilakos G, Shaffer SA, Puglise JM, Chou CH, Barton ER, Luna EJ. Novel γ-sarcoglycan interactors in murine muscle membranes. Skelet Muscle 2022; 12:2. [PMID: 35065666 PMCID: PMC8783446 DOI: 10.1186/s13395-021-00285-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The sarcoglycan complex (SC) is part of a network that links the striated muscle cytoskeleton to the basal lamina across the sarcolemma. The SC coordinates changes in phosphorylation and Ca++-flux during mechanical deformation, and these processes are disrupted with loss-of-function mutations in gamma-sarcoglycan (Sgcg) that cause Limb girdle muscular dystrophy 2C/R5. METHODS To gain insight into how the SC mediates mechano-signaling in muscle, we utilized LC-MS/MS proteomics of SC-associated proteins in immunoprecipitates from enriched sarcolemmal fractions. Criteria for inclusion were co-immunoprecipitation with anti-Sgcg from C57BL/6 control muscle and under-representation in parallel experiments with Sgcg-null muscle and with non-specific IgG. Validation of interaction was performed in co-expression experiments in human RH30 rhabdomyosarcoma cells. RESULTS We identified 19 candidates as direct or indirect interactors for Sgcg, including the other 3 SC proteins. Novel potential interactors included protein-phosphatase-1-catalytic-subunit-beta (Ppp1cb, PP1b) and Na+-K+-Cl--co-transporter NKCC1 (SLC12A2). NKCC1 co-localized with Sgcg after co-expression in human RH30 rhabdomyosarcoma cells, and its cytosolic domains depleted Sgcg from cell lysates upon immunoprecipitation and co-localized with Sgcg after detergent permeabilization. NKCC1 localized in proximity to the dystrophin complex at costameres in vivo. Bumetanide inhibition of NKCC1 cotransporter activity in isolated muscles reduced SC-dependent, strain-induced increases in phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). In silico analysis suggests that candidate SC interactors may cross-talk with survival signaling pathways, including p53, estrogen receptor, and TRIM25. CONCLUSIONS Results support that NKCC1 is a new SC-associated signaling protein. Moreover, the identities of other candidate SC interactors suggest ways by which the SC and NKCC1, along with other Sgcg interactors such as the membrane-cytoskeleton linker archvillin, may regulate kinase- and Ca++-mediated survival signaling in skeletal muscle.
Collapse
Affiliation(s)
- Tara C Smith
- Department of Radiology, Division of Cell Biology & Imaging, University of Massachusetts Medical School, Worcester, MA, USA
| | - Georgios Vasilakos
- Applied Physiology & Kinesiology, College of Health & Human Performance, University of Florida, Gainesville, FL, USA
| | - Scott A Shaffer
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA.,Mass Spectrometry Facility, University of Massachusetts Medical School, Shrewsbury, MA, USA
| | - Jason M Puglise
- Applied Physiology & Kinesiology, College of Health & Human Performance, University of Florida, Gainesville, FL, USA
| | - Chih-Hsuan Chou
- Applied Physiology & Kinesiology, College of Health & Human Performance, University of Florida, Gainesville, FL, USA
| | - Elisabeth R Barton
- Applied Physiology & Kinesiology, College of Health & Human Performance, University of Florida, Gainesville, FL, USA.
| | - Elizabeth J Luna
- Department of Radiology, Division of Cell Biology & Imaging, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
13
|
Quental R, Gonçalves D, Rodrigues E, Serrano Gonçalves E, Oliveira J, Parente Freixo J, Leão M. Congenital heart defects associated with pathogenic variants in WAC gene: Expanding the phenotypic and genotypic spectrum of DeSanto-Shinawi syndrome. Am J Med Genet A 2022; 188:1311-1316. [PMID: 34997803 DOI: 10.1002/ajmg.a.62636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 11/10/2022]
Abstract
WAC-related intellectual disability, also known as DeSanto-Shinawi syndrome, is a rare autosomal dominant genetic disorder caused by pathogenic variants in WAC gene. This syndrome is characterized by developmental delay, intellectual disability, behavioral abnormalities, and dysmorphic facial features, including deep-set eyes, flat nasal bridge, bulbous nasal tip, and synophrys. Chromosomal deletions at 10p12p11 encompassing WAC gene have been described in patients with a similar phenotype, presenting with developmental delay, intellectual disability, visual impairments, abnormal behavior, and dysmorphic features. An important clinical difference between the two groups of patients, is that those with large deletions frequently present with congenital cardiac defects, which were rarely reported in patients with pathogenic variants in WAC. The genes underlying heart defects in patients with the deletion have not yet been fully clarified. Here, we describe two unrelated Portuguese patients with de novo pathogenic variants in WAC gene, previously unreported in the literature. Both patients present with microcephaly, developmental delay, intellectual disability, behavioral problems, and facial dysmorphisms. Interestingly, the youngest patient has a severe congenital cardiac malformation, showing that intragenic pathogenic WAC variants can also be associated with heart defects. Therefore, this report expands the phenotypic and genotypic spectrum of this rare syndrome and provides deeper insights by comparing the clinical features of our patients with previously reported cases.
Collapse
Affiliation(s)
- Rita Quental
- Department of Medical Genetics, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
| | - Daniel Gonçalves
- Neurodevelopment Unit, Department of Pediatrics, CHUSJ, Porto, Portugal
| | | | | | - Jorge Oliveira
- CGPP - Centro de Genética Preditiva e Preventiva, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - João Parente Freixo
- CGPP - Centro de Genética Preditiva e Preventiva, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Miguel Leão
- Department of Medical Genetics, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
| |
Collapse
|
14
|
Shin JJ, Kim B, Kang J, Choi J, Moon BJ, Ryu DS, Yoon SH, Chin DK, Lee JK, Kim KN, Ha Y. Clinical, Radiographic, and Genetic Analyses in a Population-Based Cohort of Adult Spinal Deformity in the Older Population. Neurospine 2021; 18:608-617. [PMID: 34610692 PMCID: PMC8497247 DOI: 10.14245/ns.2142544.272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/27/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to identify the sagittal parameters associated with health-related quality of life and genetic variations that increase the risk of adult spinal deformity (ASD) onset in the older population. METHODS We recruited 120 participants who had a sagittal vertical axis > 50 mm in a sagittal imbalance study. Sagittal radiographic parameters, cross-sectional area, and intramuscular fatty infiltration using the Goutallier classification in the paraspinal lumbar muscles were evaluated. Functional scales included the self-reported Oswestry Disability Index (ODI), 36-item Short Form Health Survey (SF-36), and visual analogue scales (VAS) for back and leg pain. We performed whole-exome sequencing and an exome-wide association study using the 100 control subjects and 63 individuals with severe phenotypes of sagittal imbalance. RESULTS Pelvic incidence minus lumbar lordosis (PI-LL) mismatch was negatively associated with the SF-36 and positively correlated with ODI and VAS for back and leg pain. PI-LL was related to the quality and size of the paraspinal muscles, especially the multifidus muscle. We identified common individual variants that reached exome-wide significance using single-variant analysis. The most significant single-nucleotide polymorphism was rs78773460, situated in an exon of the SVIL gene (odds ratio, 9.61; p = 1.15 × 10-9). CONCLUSION Older age, higher body mass index, and a more significant PI-LL mismatch were associated with unfavorable results on functional scales. We found a genetic variation in the SVIL gene, which has been associated with the integrity of the cytoskeleton and the development of skeletal muscles, in severe ASD phenotypes. Our results help to elucidate the pathogenesis of ASD.
Collapse
Affiliation(s)
- Jun Jae Shin
- Department of Neurosurgery, Yongin Severance Hospital, Yonsei University School of Medicine, Yongin, Korea
| | - Byeongwoo Kim
- Department of Neurosurgery, Champodonamu Hospital, Seoul, Korea
| | - Juwon Kang
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Bong Ju Moon
- Department of Neurosurgery, Chonnam National University Medical School & Research Institute of Medical Sciences, Gwangju, Korea
| | - Dal Sung Ryu
- Department of Neurosurgery, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Seung Hwan Yoon
- Department of Neurosurgery, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Dong Kyu Chin
- Department of Neurosurgery, Spine and Spinal Cord Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Kil Lee
- Department of Neurosurgery, Chonnam National University Medical School & Research Institute of Medical Sciences, Gwangju, Korea
| | - Keung Nyun Kim
- Department of Neurosurgery, Spine and Spinal Cord Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Ha
- Department of Neurosurgery, Spine and Spinal Cord Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,POSTECH Biotech Center, Pohang University of Science and Technology, Pohang, Korea
| |
Collapse
|
15
|
Biallelic loss-of-function variants in WDR11 are associated with microcephaly and intellectual disability. Eur J Hum Genet 2021; 29:1663-1668. [PMID: 34413497 PMCID: PMC8560748 DOI: 10.1038/s41431-021-00943-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/09/2021] [Accepted: 08/05/2021] [Indexed: 11/08/2022] Open
Abstract
Heterozygous missense variants in the WD repeat domain 11 (WDR11) gene are associated with hypogonadotropic hypogonadism in humans. In contrast, knockout of both alleles of Wdr11 in mice results in a more severe phenotype with growth and developmental delay, features of holoprosencephaly, heart defects and reproductive disorders. Similar developmental defects known to be associated with aberrant hedgehog signaling and ciliogenesis have been found in zebrafish after Wdr11 knockdown. We here report biallelic loss-of-function variants in the WDR11 gene in six patients from three independent families with intellectual disability, microcephaly and short stature. The findings suggest that biallelic WDR11 variants in humans result in an overlapping but milder phenotype compared to Wdr11-deficient animals. However, the observed human phenotype differs significantly from dominantly inherited variants leading to hypogonadotropic hypogonadism, suggesting that recessive WDR11 variants result in a clinically distinct entity.
Collapse
|
16
|
246th ENMC International Workshop: Protein aggregate myopathies 24-26 May 2019, Hoofddorp, The Netherlands. Neuromuscul Disord 2020; 31:158-166. [PMID: 33303357 DOI: 10.1016/j.nmd.2020.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022]
|