1
|
Zabalegui F, Castañeda SL, Amin G, Belli C, Miriuka SG, Moro LN. Derivation of two human induced pluripotent stem cell lines carrying a missense mutation in FHL1 (c.377G > A, p.C126Y) linked to familial muscular dystrophy. Stem Cell Res 2024; 75:103307. [PMID: 38244535 DOI: 10.1016/j.scr.2024.103307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
FHL1 gene locates in the Xq26 region and encodes for four and half LIM domain protein 1. It plays a crucial role in muscle cells and mutations in FHL1 are related to muscular dystrophy (MD). Peripheral blood mononuclear cells (PBMCs) were obtained from 2 family patients with MD that carry a pathogenic missense mutation in FHL1 (c.377G > A, p.C126Y). Induced pluripotent stem cells (iPSCs) were generated by PBMCs reprogramming using the lentiviral-hSTEMCCA-loxP vector, obtaining FHL1-T and FHL1-V iPSCs lines from patients. FHL1 genotype was maintained, and stemness and pluripotency were confirmed in both iPSCs lines.
Collapse
Affiliation(s)
- Federico Zabalegui
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Instituto de Neurociencias (INEU), CONICET, Buenos Aires, Argentina
| | - Sheila Lucia Castañeda
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Instituto de Neurociencias (INEU), CONICET, Buenos Aires, Argentina
| | - Guadalupe Amin
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Instituto de Neurociencias (INEU), CONICET, Buenos Aires, Argentina
| | - Carolina Belli
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Santiago Gabriel Miriuka
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Instituto de Neurociencias (INEU), CONICET, Buenos Aires, Argentina
| | - Lucía Natalia Moro
- Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Instituto de Neurociencias (INEU), CONICET, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Doh CY, Schmidt AV, Chinthalapudi K, Stelzer JE. Bringing into focus the central domains C3-C6 of myosin binding protein C. Front Physiol 2024; 15:1370539. [PMID: 38487262 PMCID: PMC10937550 DOI: 10.3389/fphys.2024.1370539] [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: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Myosin binding protein C (MyBPC) is a multi-domain protein with each region having a distinct functional role in muscle contraction. The central domains of MyBPC have often been overlooked due to their unclear roles. However, recent research shows promise in understanding their potential structural and regulatory functions. Understanding the central region of MyBPC is important because it may have specialized function that can be used as drug targets or for disease-specific therapies. In this review, we provide a brief overview of the evolution of our understanding of the central domains of MyBPC in regard to its domain structures, arrangement and dynamics, interaction partners, hypothesized functions, disease-causing mutations, and post-translational modifications. We highlight key research studies that have helped advance our understanding of the central region. Lastly, we discuss gaps in our current understanding and potential avenues to further research and discovery.
Collapse
Affiliation(s)
- Chang Yoon Doh
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Alexandra V. Schmidt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
3
|
Qi T, Zhang J, Zhang K, Zhang W, Song Y, Lian K, Kan C, Han F, Hou N, Sun X. Unraveling the role of the FHL family in cardiac diseases: Mechanisms, implications, and future directions. Biochem Biophys Res Commun 2024; 694:149468. [PMID: 38183876 DOI: 10.1016/j.bbrc.2024.149468] [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: 11/07/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Heart diseases are a major cause of morbidity and mortality worldwide. Understanding the molecular mechanisms underlying these diseases is essential for the development of effective diagnostic and therapeutic strategies. The FHL family consists of five members: FHL1, FHL2, FHL3, FHL4, and FHL5/Act. These members exhibit different expression patterns in various tissues including the heart. FHL family proteins are implicated in cardiac remodeling, regulation of metabolic enzymes, and cardiac biomechanical stress perception. A large number of studies have explored the link between FHL family proteins and cardiac disease, skeletal muscle disease, and ovarian metabolism, but a comprehensive and in-depth understanding of the specific molecular mechanisms targeting FHL on cardiac disease is lacking. The aim of this review is to explore the structure and function of FHL family members, to comprehensively elucidate the mechanisms by which they regulate the heart, and to explore in depth the changes in FHL family members observed in different cardiac disorders, as well as the effects of mutations in FHL proteins on heart health.
Collapse
Affiliation(s)
- Tongbing Qi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Wenqiang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Yixin Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Kexin Lian
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
4
|
Li D, Chen F, Tian Y, Su Y. Transcriptome analysis of the gene expression of M . iliotibialis lateralis affected by dietary methionine restriction. Front Physiol 2023; 14:1184651. [PMID: 37284544 PMCID: PMC10240061 DOI: 10.3389/fphys.2023.1184651] [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: 03/12/2023] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction: Methionine (Met) is an important amino acid related to the development of skeletal muscle. This study investigated the effects of dietary Met restriction on the gene expression of M. iliotibialis lateralis. Methods: A total of 84 day-old broiler chicks (Zhuanghe Dagu) with a similar initial body weight (207.62 ± 8.54 g) were used in this study. All birds were divided into two groups (CON; L-Met) based on the initial body weight. Each group consisted of six replicates with seven birds per replicate. The experimental period was 63 days (phase 1, days 1-21; phase 2, days 22-63). According to the nutritional requirements of Zhuanghe Dagu chickens, we provided a basal diet (0.39% Met levels during phase 1 and 0.35% Met levels during phase 2, as-fed basis) to the birds in the CON group, while we provided a Met-restricted diet (0.31% Met levels during phase 1 and 0.28% Met levels during phase 2, as-fed basis) to the birds in the L-Met group. The growth performance of broiler chicks and their M. iliotibialis lateralis development parameters were measured on days 21 and 63. Results and Discussion: In this study, dietary Met restriction did not affect the growth performance of broiler chicks but hindered the development of M. iliotibialis lateralis at both sampling timepoints. On the final day, three birds selected from each group (three from CON and three from L-Met) were used to obtain M. iliotibialis lateralis samples from leg muscle for further transcriptome analysis. Transcriptome analysis revealed that dietary Met restriction significantly upregulated 247 differentially expressed genes (DEGs) and downregulated 173 DEGs. Additionally, DEGs were mainly enriched in 10 pathways. Among DEGs, we observed that dietary Met restriction downregulated the expression of CSRP3, KY, FHL1, LMCD1, and MYOZ2 in M. iliotibialis lateralis. Therefore, we considered that dietary Met restriction had negative effects on the development of M. iliotibialis lateralis, and CSRP3, KY, FHL1, LMCD1, and MYOZ2 may serve as potential functional genes involved in this process.
Collapse
Affiliation(s)
- Desheng Li
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
- Laboratory of Quality and Safety of Animal Product of Liaoning Province, Jinzhou, China
| | - Fei Chen
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
- Laboratory of Quality and Safety of Animal Product of Liaoning Province, Jinzhou, China
| | - Yumin Tian
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
- Laboratory of Quality and Safety of Animal Product of Liaoning Province, Jinzhou, China
| | - Yuhong Su
- Laboratory of Quality and Safety of Animal Product of Liaoning Province, Jinzhou, China
- College of Food and Health, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
5
|
Johnson LG, Zhai C, Steadham EM, Reever LM, Prusa KJ, Nair MN, Huff-Lonergan E, Lonergan SM. Distinct myofibrillar sub-proteomic profiles are associated with the instrumental texture of aged pork loin. J Anim Sci 2023; 101:skad327. [PMID: 37751382 PMCID: PMC10629443 DOI: 10.1093/jas/skad327] [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: 08/11/2023] [Accepted: 09/22/2023] [Indexed: 09/28/2023] Open
Abstract
Fresh pork tenderness contributes to consumer satisfaction with the eating experience. Postmortem proteolysis of proteins within and between myofibrils has been closely linked with pork tenderness development. A clear understanding of the molecular features associated with pork tenderness development will provide additional targets and open the door to new solutions to improve and make pork tenderness development more consistent. Therefore, the objective was to utilize liquid chromatography and mass spectrometry with tandem mass tag (TMT) multiplexing to evaluate myofibrillar sub-proteome differences between pork chops of different instrumental star probe values. Pork loins (N = 120) were collected from a commercial harvest facility at 24 h postmortem. Quality and sensory attributes were evaluated at 24 h postmortem and after ~2 weeks of postmortem aging. Pork chops were grouped into 4 groups based on instrumental star probe value (group A,x¯ = 4.23 kg, 3.43 to 4.55 kg; group B,x¯ = 4.79 kg, 4.66 to 5.00 kg; group C,x¯ = 5.43 kg, 5.20 to 5.64 kg; group D,x¯ = 6.21 kg, 5.70 to 7.41 kg; n = 25 per group). Myofibrillar proteins from the samples aged ~2 wk were fractionated, washed, and solubilized in 8.3 M urea, 2 M thiourea, and 1% dithiothreitol. Proteins were digested with trypsin, labeled with 11-plex isobaric TMT reagents, and identified and quantified using a Q-Exactive Mass Spectrometer. Between groups A and D, 54 protein groups were differentially abundant (adjusted P < 0.05). Group A had a greater abundance of proteins related to the thick and thin filament and a lesser abundance of Z-line-associated proteins and metabolic enzymes than group D chops. These data highlight that distinct myofibrillar sub-proteomes are associated with pork chops of different tenderness values. Future research should evaluate changes immediately and earlier postmortem to further elucidate myofibrillar sub-proteome differences over the postmortem aging period.
Collapse
Affiliation(s)
- Logan G Johnson
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Chaoyu Zhai
- Department of Animal Science, University of Connecticut, Storrs, CT 06269-4040, USA
| | - Edward M Steadham
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Leah M Reever
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Kenneth J Prusa
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA
| | - Mahesh N Nair
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Steven M Lonergan
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
6
|
Bax M, Romanov V, Junday K, Giannoulatou E, Martinac B, Kovacic JC, Liu R, Iismaa SE, Graham RM. Arterial dissections: Common features and new perspectives. Front Cardiovasc Med 2022; 9:1055862. [PMID: 36561772 PMCID: PMC9763901 DOI: 10.3389/fcvm.2022.1055862] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Arterial dissections, which involve an abrupt tear in the wall of a major artery resulting in the intramural accumulation of blood, are a family of catastrophic disorders causing major, potentially fatal sequelae. Involving diverse vascular beds, including the aorta or coronary, cervical, pulmonary, and visceral arteries, each type of dissection is devastating in its own way. Traditionally they have been studied in isolation, rather than collectively, owing largely to the distinct clinical consequences of dissections in different anatomical locations - such as stroke, myocardial infarction, and renal failure. Here, we review the shared and unique features of these arteriopathies to provide a better understanding of this family of disorders. Arterial dissections occur commonly in the young to middle-aged, and often in conjunction with hypertension and/or migraine; the latter suggesting they are part of a generalized vasculopathy. Genetic studies as well as cellular and molecular investigations of arterial dissections reveal striking similarities between dissection types, particularly their pathophysiology, which includes the presence or absence of an intimal tear and vasa vasorum dysfunction as a cause of intramural hemorrhage. Pathway perturbations common to all types of dissections include disruption of TGF-β signaling, the extracellular matrix, the cytoskeleton or metabolism, as evidenced by the finding of mutations in critical genes regulating these processes, including LRP1, collagen genes, fibrillin and TGF-β receptors, or their coupled pathways. Perturbances in these connected signaling pathways contribute to phenotype switching in endothelial and vascular smooth muscle cells of the affected artery, in which their physiological quiescent state is lost and replaced by a proliferative activated phenotype. Of interest, dissections in various anatomical locations are associated with distinct sex and age predilections, suggesting involvement of gene and environment interactions in disease pathogenesis. Importantly, these cellular mechanisms are potentially therapeutically targetable. Consideration of arterial dissections as a collective pathology allows insight from the better characterized dissection types, such as that involving the thoracic aorta, to be leveraged to inform the less common forms of dissections, including the potential to apply known therapeutic interventions already clinically available for the former.
Collapse
Affiliation(s)
- Monique Bax
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Valentin Romanov
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Keerat Junday
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Jason C. Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, United States
| | - Renjing Liu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Siiri E. Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
| |
Collapse
|
7
|
Stathopoulou K, Schnittger J, Raabe J, Fleischer F, Mangels N, Piasecki A, Findlay J, Hartmann K, Krasemann S, Schlossarek S, Uebeler J, Wixler V, Blake DJ, Baillie GS, Carrier L, Ehler E, Cuello F. CMYA5 is a novel interaction partner of FHL2 in cardiac myocytes. FEBS J 2022; 289:4622-4645. [PMID: 35176204 DOI: 10.1111/febs.16402] [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: 06/23/2021] [Revised: 01/13/2022] [Accepted: 02/15/2022] [Indexed: 11/27/2022]
Abstract
Four-and-a-half LIM domains protein 2 (FHL2) is an anti-hypertrophic adaptor protein that regulates cardiac myocyte signalling and function. Herein, we identified cardiomyopathy-associated 5 (CMYA5) as a novel FHL2 interaction partner in cardiac myocytes. In vitro pull-down assays demonstrated interaction between FHL2 and the N- and C-terminal regions of CMYA5. The interaction was verified in adult cardiac myocytes by proximity ligation assays. Immunofluorescence and confocal microscopy demonstrated co-localisation in the same subcellular compartment. The binding interface between FHL2 and CMYA5 was mapped by peptide arrays. Exposure of neonatal rat ventricular myocytes to a CMYA5 peptide covering one of the FHL2 interaction sites led to an increase in cell area at baseline, but a blunted response to chronic phenylephrine treatment. In contrast to wild-type hearts, loss or reduced FHL2 expression in Fhl2-targeted knockout mouse hearts or in a humanised mouse model of hypertrophic cardiomyopathy led to redistribution of CMYA5 into the perinuclear and intercalated disc region. Taken together, our results indicate a direct interaction of the two adaptor proteins FHL2 and CMYA5 in cardiac myocytes, which might impact subcellular compartmentation of CMYA5.
Collapse
Affiliation(s)
- Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Josef Schnittger
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Frederic Fleischer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Nils Mangels
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Angelika Piasecki
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Jane Findlay
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - June Uebeler
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Viktor Wixler
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, Westfaelische Wilhelms-University, Germany
| | - Derek J Blake
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Elisabeth Ehler
- School of Cardiovascular Medicine and Sciences, BHF Research Excellence Centre, King's College London, UK.,Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences), King's College London, UK
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
8
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
9
|
van der Pijl RJ, Domenighetti AA, Sheikh F, Ehler E, Ottenheijm CAC, Lange S. The titin N2B and N2A regions: biomechanical and metabolic signaling hubs in cross-striated muscles. Biophys Rev 2021; 13:653-677. [PMID: 34745373 PMCID: PMC8553726 DOI: 10.1007/s12551-021-00836-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Muscle specific signaling has been shown to originate from myofilaments and their associated cellular structures, including the sarcomeres, costameres or the cardiac intercalated disc. Two signaling hubs that play important biomechanical roles for cardiac and/or skeletal muscle physiology are the N2B and N2A regions in the giant protein titin. Prominent proteins associated with these regions in titin are chaperones Hsp90 and αB-crystallin, members of the four-and-a-half LIM (FHL) and muscle ankyrin repeat protein (Ankrd) families, as well as thin filament-associated proteins, such as myopalladin. This review highlights biological roles and properties of the titin N2B and N2A regions in health and disease. Special emphasis is placed on functions of Ankrd and FHL proteins as mechanosensors that modulate muscle-specific signaling and muscle growth. This region of the sarcomere also emerged as a hotspot for the modulation of passive muscle mechanics through altered titin phosphorylation and splicing, as well as tethering mechanisms that link titin to the thin filament system.
Collapse
Affiliation(s)
| | - Andrea A. Domenighetti
- Shirley Ryan AbilityLab, Chicago, IL USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL USA
| | - Farah Sheikh
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Coen A. C. Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ USA
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Stephan Lange
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Mota IA, Correia CDC, Fontana PN, Carvalho AADS. Reducing body myopathy - A new pathogenic FHL1 variant and literature review. Neuromuscul Disord 2021; 31:847-853. [PMID: 34366191 DOI: 10.1016/j.nmd.2021.03.013] [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: 09/24/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 11/26/2022]
Abstract
Reducing body myopathy (RBM) is a rare disease marked by progressive muscle weakness caused by a mutation in FHL1 gene. We describe a new pathogenic variant and contrasted it with 44 other cases identified in the literature. A male child presented at age 3 suffering frequent falls and progressive muscular weakness. At age 8, he was wheelchair-bound and required ventilatory support. His mother and sister died due to the same problem. Creatine kinase was 428 IU/L (<190). Muscle biopsy showed typical reducing bodies, and genetic analysis identified a novel pathogenic hemizygous variant, c.370_375del. We identified 44 previous reported cases separated in two groups: 28 cases with mean age onset 7.6 ± 5 years and 16 with 26.7 ± 4.2 years. The time for the diagnosis was shorter to younger group. The initial symptoms, rigid spine, contractures, scoliosis and axial and neck weaknesses, dysphagia, cardiac involvement, were predominant in younger group. The variant c.369C > G predominated in younger group and c.448T > C in older one. Pathogenic variants positions seemed related to severe phenotype. Most wheelchair patients belonged to younger group. The data from this compilation and our case provided a general characterization spectrum and prognosis between two groups of age onset with RBM.
Collapse
Affiliation(s)
- Isabella Araujo Mota
- Neurorehabilitation service at Hospital Universitário Lauro Wanderley, João Pessoa, Paraíba, Brazil
| | - Carolina da Cunha Correia
- Assistant Professor of Neurology at Faculdade de Ciências Medicas - Universidade de Pernambuco (UPE), Recife, Brazil
| | - Pedro Nogueira Fontana
- Post Graduate Program in Health Sciences, Faculdade de Ciências Médicas da Universidade de Pernambuco (UPE), Recife, Brazil
| | | |
Collapse
|
11
|
Chen L, Lin HX, Yang XX, Chen DF, Dong HL, Yu H, Liu GL, Wu ZY. Clinical and genetic characteristics of Chinese patients with reducing body myopathy. Neuromuscul Disord 2021; 31:442-449. [PMID: 33846077 DOI: 10.1016/j.nmd.2021.02.009] [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: 10/27/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Abstract
Reducing body myopathy (RBM) is a rare myopathy characterized by reducing bodies (RBs) in morphological presentation. The clinical manifestations of RBM present a wide clinical spectrum, varying from infantile lethal form through childhood and adult benign forms. FHL1 gene is the causative gene of RBM. To date, only 6 Chinese RBM patients have been reported. Here, we reported the clinical presentations and genetic findings of 3 Chinese RBM patients from two families. Two novel pathogenic variants, c.395G>A and c.401_402insGAC, were identified by whole exome sequencing. Furthermore, by reviewing previous studies, we revealed that most RBM patients manifested with an early onset, symmetric, progressive limb-girdle and axial muscle weakness with joint contractures, rigid spine or scoliosis except familial female patients who exhibited asymmetric benign muscle involvements. Our results provide insightful information to help better diagnose and understand the disease.
Collapse
Affiliation(s)
- Lei Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Xia Lin
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Xia Yang
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dian-Fu Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Lin Dong
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Yu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Gong-Lu Liu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
12
|
Bobyleva LG, Shumeyko SA, Yakupova EI, Surin AK, Galzitskaya OV, Kihara H, Timchenko AA, Timchenko MA, Penkov NV, Nikulin AD, Suvorina MY, Molochkov NV, Lobanov MY, Fadeev RS, Vikhlyantsev IM, Bobylev AG. Myosin Binding Protein-C Forms Amyloid-Like Aggregates In Vitro. Int J Mol Sci 2021; 22:ijms22020731. [PMID: 33450960 PMCID: PMC7828380 DOI: 10.3390/ijms22020731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 11/17/2022] Open
Abstract
This work investigated in vitro aggregation and amyloid properties of skeletal myosin binding protein-C (sMyBP-C) interacting in vivo with proteins of thick and thin filaments in the sarcomeric A-disc. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) found a rapid (5–10 min) formation of large (>2 μm) aggregates. sMyBP-C oligomers formed both at the initial 5–10 min and after 16 h of aggregation. Small angle X-ray scattering (SAXS) and DLS revealed sMyBP-C oligomers to consist of 7–10 monomers. TEM and atomic force microscopy (AFM) showed sMyBP-C to form amorphous aggregates (and, to a lesser degree, fibrillar structures) exhibiting no toxicity on cell culture. X-ray diffraction of sMyBP-C aggregates registered reflections attributed to a cross-β quaternary structure. Circular dichroism (CD) showed the formation of the amyloid-like structure to occur without changes in the sMyBP-C secondary structure. The obtained results indicating a high in vitro aggregability of sMyBP-C are, apparently, a consequence of structural features of the domain organization of proteins of this family. Formation of pathological amyloid or amyloid-like sMyBP-C aggregates in vivo is little probable due to amino-acid sequence low identity (<26%), alternating ordered/disordered regions in the protein molecule, and S–S bonds providing for general stability.
Collapse
Affiliation(s)
- Liya G. Bobyleva
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Sergey A. Shumeyko
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Elmira I. Yakupova
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Alexey K. Surin
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
- Biological Testing Laboratory, Branch of the Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
- Laboratory of the Biochemistry of Pathogenic Microorganisms, State Research Centre for Applied Microbiology and Biotechnology, Obolensk, 142279 Serpukhov District, Russia
| | - Oxana V. Galzitskaya
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Hiroshi Kihara
- Department of Early Childhood Education, Himeji-Hinomoto College, 890 Koro, Kodera-cho, Himeji 679-2151, Japan;
| | - Alexander A. Timchenko
- Group of Experimental Research and Engineering of Oligomeric Structures, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Maria A. Timchenko
- Laboratory of Applied Enzymology, FRC PSCBR, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Nikita V. Penkov
- Laboratory of the Methods of Optical Spectral Analysis, Institute of Cell Biophysics, Russian Academy of Sciences, FRC PSCBR RAS, 142290 Pushchino, Russia;
| | - Alexey D. Nikulin
- Laboratory for Structural Studies of the Translational Apparatus, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Mariya Yu. Suvorina
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Nikolay V. Molochkov
- Laboratory of NMR Investigations of Biosystems, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Mikhail Yu. Lobanov
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Roman S. Fadeev
- Laboratory of Pharmacological Regulation of Cell Resistance, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Ivan M. Vikhlyantsev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Correspondence: (I.M.V.); (A.G.B.)
| | - Alexander G. Bobylev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Correspondence: (I.M.V.); (A.G.B.)
| |
Collapse
|
13
|
Wu J, Zhao K, Du Z, Chen Y, Zhang F, Jiang W, Zheng J, Wu X, Shen C, Xiao X. Systemic effect of FHL1 on neuromuscular junction and myotube formation via insulin-like growth factor and myostatin signaling pathways. Biochem Biophys Res Commun 2021; 537:125-131. [PMID: 33401146 DOI: 10.1016/j.bbrc.2020.12.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/16/2020] [Indexed: 11/17/2022]
Abstract
Four-and-a-half LIM domain protein 1 (FHL1) is a member of the FHL protein family that serves as a scaffold protein to maintain normal cellular structure and function. Its mutations have been implicated in multiple muscular diseases. These FHL1 related myopathies are characterized by symptoms such as progressive muscle loss, rigid or bent spine, even cardiac or respiratory failure in some patients, which implies pathological problems not only in muscles, but also in the nervous system. Moreover, decreased FHL1 protein level has been found in patients with FHL1 mutations, indicating the protein loss-of-function as a pathological cause of such diseases. These findings suggest the significance of understanding the systemic role of FHL1 in the homeostasis of nervous system and muscle. Here we reported that Fhl1 loss in C2C12 myotubes obscured acetylcholine receptor (AChR) clustering in addition to myotube fusion, which was associated with impaired MuSK phosphorylation. Mechanistically, myostatin-SMAD2/3 signaling was enhanced, whereas IGF-PI3K-AKT signaling was suppressed in Fhl1-/- C2C12 myotubes. Reversion of these molecular alterations rescued AChR clustering and differentiation deficits. These data outline a systemic regulation of AChR clustering and myotube fusion by FHL1, which may offer clues for mechanism study and development of therapeutic strategies to treat FHL1 related myopathies.
Collapse
Affiliation(s)
- Jiamei Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Kai Zhao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zengmin Du
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Ying Chen
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Feixu Zhang
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wei Jiang
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jing Zheng
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| | - Xia Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| | - Chengyong Shen
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| | - Xiao Xiao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
14
|
Huang C, Hou C, Ijaz M, Yan T, Li X, Li Y, Zhang D. Proteomics discovery of protein biomarkers linked to meat quality traits in post-mortem muscles: Current trends and future prospects: A review. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.09.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Diurnal Differences in Human Muscle Isometric Force In Vivo Are Associated with Differential Phosphorylation of Sarcomeric M-Band Proteins. Proteomes 2020; 8:proteomes8030022. [PMID: 32859009 PMCID: PMC7565642 DOI: 10.3390/proteomes8030022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/06/2020] [Accepted: 08/25/2020] [Indexed: 12/25/2022] Open
Abstract
We investigated whether diurnal differences in muscle force output are associated with the post-translational state of muscle proteins. Ten physically active men (mean ± SD; age 26.7 ± 3.7 y) performed experimental sessions in the morning (08:00 h) and evening (17:00 h), which were counterbalanced in order of administration and separated by at least 72 h. Knee extensor maximal voluntary isometric contraction (MVIC) force and peak rate of force development (RFD) were measured, and samples of vastus lateralis were collected immediately after exercise. MVIC force was greater in the evening (mean difference of 67 N, 10.2%; p < 0.05). Two-dimensional (2D) gel analysis encompassed 122 proteoforms and discovered 6 significant (p < 0.05; false discovery rate [FDR] = 10%) diurnal differences. Phosphopeptide analysis identified 1693 phosphopeptides and detected 140 phosphopeptides from 104 proteins that were more (p < 0.05, FDR = 22%) phosphorylated in the morning. Myomesin 2, muscle creatine kinase, and the C-terminus of titin exhibited the most robust (FDR < 10%) diurnal differences. Exercise in the morning, compared to the evening, coincided with a greater phosphorylation of M-band-associated proteins in human muscle. These protein modifications may alter the M-band structure and disrupt force transmission, thus potentially explaining the lower force output in the morning.
Collapse
|
16
|
Formation of Aberrant Myotubes by Myoblasts Lacking Myosin VI Is Associated with Alterations in the Cytoskeleton Organization, Myoblast Adhesion and Fusion. Cells 2020; 9:cells9071673. [PMID: 32664530 PMCID: PMC7408620 DOI: 10.3390/cells9071673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
We have previously postulated that unconventional myosin VI (MVI) could be involved in myoblast differentiation. Here, we addressed the mechanism(s) of its involvement using primary myoblast culture derived from the hindlimb muscles of Snell’s waltzer mice, the natural MVI knockouts (MVI-KO). We observed that MVI-KO myotubes were formed faster than control heterozygous myoblasts (MVI-WT), with a three-fold increase in the number of myosac-like myotubes with centrally positioned nuclei. There were also changes in the levels of the myogenic transcription factors Pax7, MyoD and myogenin. This was accompanied by changes in the actin cytoskeleton and adhesive structure organization. We observed significant decreases in the levels of proteins involved in focal contact formation, such as talin and focal adhesion kinase (FAK). Interestingly, the levels of proteins involved in intercellular communication, M-cadherin and drebrin, were also affected. Furthermore, time-dependent alterations in the levels of the key proteins for myoblast membrane fusion, myomaker and myomerger, without effect on their cellular localization, were observed. Our data indicate that in the absence of MVI, the mechanisms controlling cytoskeleton organization, as well as myoblast adhesion and fusion, are dysregulated, leading to the formation of aberrant myotubes.
Collapse
|
17
|
Zhang X, Antonelo D, Hendrix J, To V, Campbell Y, Von Staden M, Li S, Suman SP, Zhai W, Chen J, Zhu H, Schilling W. Proteomic Characterization of Normal and Woody Breast Meat from Broilers of Five Genetic Strains. MEAT AND MUSCLE BIOLOGY 2020. [DOI: 10.22175/mmb.8759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Woody breast (WB) is an emergent broiler myopathy that is macroscopically characterized by hardened areas of the Pectoralis major muscle. Five genetic strains (strains 1–5) of mixed-sex broilers were fed either a control or an amino acid (AA)-reduced diet (20% reduction of digestible lysine, total sulfur AAs, and threonine) for 8 wk. Differences between whole-muscle proteome profiles of normal breast (NB; n = 6 gels) and WB tissue (n = 6 gels) were characterized for (1) broiler strains 1–5 that were fed with a control diet and collected at 0 min; (2) strain 5 (control diet) that were collected at 15 min, 4 h, and 24 h; (3) strain 5 (0 min) that were fed with a control and an AA-reduced diet. Birds that yielded WB were heavier and had a greater pH at death (pH0min) than normal birds. Results indicated that 21 proteins were more abundant (P < 0.05) and 3 proteins were less abundant (P < 0.05) in WB compared with NB. The differentially abundant proteins in each comparison were consistently upregulated or downregulated in WB tissue although the different protein profiles were noticed for each comparison. Strains 2 and 5 had more protein profile differences between WB and NB meat than strains 1, 3, and 4, which potentially indicates a stronger genetic component for strains 2 and 5 with respect to WB formation. The proteins that were more abundant in WB compared to NB are involved in carbohydrate metabolism, oxidative stress, cytoskeleton structure, and transport and signaling. Ingenuity Pathway Analysis indicated that regulated pathways in WB were mainly related to carbohydrate metabolism, cellular repair, cellular organization and maintenance, and cell death and survival. The results support the potential causes of WB myopathy, including the presence of hypoxia, oxidative stress, increased apoptosis, misfolded proteins, and inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Wes Schilling
- Mississippi State University Department of Food Science, Nutrition and Health Promotion
| |
Collapse
|
18
|
Heling LWHJ, Geeves MA, Kad NM. MyBP-C: one protein to govern them all. J Muscle Res Cell Motil 2020; 41:91-101. [PMID: 31960266 PMCID: PMC7109175 DOI: 10.1007/s10974-019-09567-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/29/2019] [Indexed: 12/19/2022]
Abstract
The heart is an extraordinarily versatile pump, finely tuned to respond to a multitude of demands. Given the heart pumps without rest for decades its efficiency is particularly relevant. Although many proteins in the heart are essential for viability, the non-essential components can attract numerous mutations which can cause disease, possibly through alterations in pumping efficiency. Of these, myosin binding protein C is strongly over-represented with ~ 40% of all known mutations in hypertrophic cardiomyopathy. Therefore, a complete understanding of its molecular function in the cardiac sarcomere is warranted. In this review, we revisit contemporary and classical literature to clarify both the current standing of this fast-moving field and frame future unresolved questions. To date, much effort has been directed at understanding MyBP-C function on either thick or thin filaments. Here we aim to focus questions on how MyBP-C functions at a molecular level in the context of both the thick and thin filaments together. A concept that emerges is MyBP-C acts to govern interactions on two levels; controlling myosin access to the thin filament by sequestration on the thick filament, and controlling the activation state and access of myosin to its binding sites on the thin filament. Such affects are achieved through directed interactions mediated by phosphorylation (of MyBP-C and other sarcomeric components) and calcium.
Collapse
Affiliation(s)
- L W H J Heling
- School of Biosciences, University of Kent, Canterbury, CT2 7NH, UK
| | - M A Geeves
- School of Biosciences, University of Kent, Canterbury, CT2 7NH, UK
| | - N M Kad
- School of Biosciences, University of Kent, Canterbury, CT2 7NH, UK.
| |
Collapse
|
19
|
FHL-1 is not involved in pressure overload-induced maladaptive right ventricular remodeling and dysfunction. Basic Res Cardiol 2020; 115:17. [PMID: 31980934 PMCID: PMC6981327 DOI: 10.1007/s00395-019-0767-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/06/2019] [Indexed: 12/31/2022]
Abstract
AIMS The cytoskeletal signaling protein four and-a-half LIM domains 1 (FHL-1) has recently been identified as a novel key player in pulmonary hypertension as well as in left heart diseases. In this regard, FHL-1 has been implicated in dysregulated hypertrophic signaling in pulmonary arterial smooth muscle cells leading to pulmonary hypertension. In mice, FHL-1-deficiency (FHL-1-/-) led to an attenuated hypertrophic signaling associated with a blunted hypertrophic response of the pressure-overloaded left ventricle (LV). However, the role of FHL-1 in right heart hypertrophy has not yet been addressed. METHODS AND RESULTS We investigated FHL-1 expression in C57Bl/6 mice subjected to chronic biomechanical stress and found it to be enhanced in the right ventricle (RV). Next, we subjected FHL-1-/- and corresponding wild-type mice to pressure overload of the RV by pulmonary arterial banding for various time points. However, in contrast to the previously published study in LV-pressure overload, which was confirmed here, RV hypertrophy and hypertrophic signaling was not diminished in FHL-1-/- mice. In detail, right ventricular pressure overload led to hypertrophy, dilatation and fibrosis of the RV from both FHL-1-/- and wild-type mice. RV remodeling was associated with impaired RV function as evidenced by reduced tricuspid annular plane systolic excursion. Additionally, PAB induced upregulation of natriuretic peptides and slight downregulation of phospholamban and ryanodine receptor 2 in the RV. However, there was no difference between genotypes in the degree of expression change. CONCLUSION FHL-1 pathway is not involved in the control of adverse remodeling in the pressure overloaded RV.
Collapse
|
20
|
Heller SA, Shih R, Kalra R, Kang PB. Emery-Dreifuss muscular dystrophy. Muscle Nerve 2019; 61:436-448. [PMID: 31840275 PMCID: PMC7154529 DOI: 10.1002/mus.26782] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 12/04/2019] [Accepted: 12/07/2019] [Indexed: 12/19/2022]
Abstract
Emery-Dreifuss muscular dystrophy (EDMD) is a rare muscular dystrophy, but is particularly important to diagnose due to frequent life-threatening cardiac complications. EDMD classically presents with muscle weakness, early contractures, cardiac conduction abnormalities and cardiomyopathy, although the presence and severity of these manifestations vary by subtype and individual. Associated genes include EMD, LMNA, SYNE1, SYNE2, FHL1, TMEM43, SUN1, SUN2, and TTN, encoding emerin, lamin A/C, nesprin-1, nesprin-2, FHL1, LUMA, SUN1, SUN2, and titin, respectively. The Online Mendelian Inheritance in Man database recognizes subtypes 1 through 7, which captures most but not all of the associated genes. Genetic diagnosis is essential whenever available, but traditional diagnostic tools can help steer the evaluation toward EDMD and assist with interpretation of equivocal genetic test results. Management is primarily supportive, but it is important to monitor patients closely, especially for potential cardiac complications. There is a high potential for progress in the treatment of EDMD in the coming years.
Collapse
Affiliation(s)
- Scott A Heller
- Department of Neurology, University of Florida College of Medicine, Gainesville, Florida
| | - Renata Shih
- Congenital Heart Center, University of Florida College of Medicine, Gainesville, Florida
| | - Raghav Kalra
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida
| | - Peter B Kang
- Department of Neurology, University of Florida College of Medicine, Gainesville, Florida.,Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida.,Genetics Institute and Myology Institute, University of Florida, Gainesville, Florida
| |
Collapse
|
21
|
Coats CJ, Heywood WE, Virasami A, Ashrafi N, Syrris P, Dos Remedios C, Treibel TA, Moon JC, Lopes LR, McGregor CGA, Ashworth M, Sebire NJ, McKenna WJ, Mills K, Elliott PM. Proteomic Analysis of the Myocardium in Hypertrophic Obstructive Cardiomyopathy. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e001974. [PMID: 30562113 DOI: 10.1161/circgen.117.001974] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is characterized by a complex phenotype that is only partly explained by the biological effects of individual genetic variants. The aim of this study was to use proteomic analysis of myocardial tissue to explore the postgenomic phenotype. METHODS Label-free proteomic analysis was used initially to compare protein profiles in myocardial samples from 11 patients with HCM undergoing surgical myectomy with control samples from 6 healthy unused donor hearts. Differentially expressed proteins of interest were validated in myocardial samples from 65 unrelated individuals (HCM [n=51], controls [n=7], and aortic stenosis [n=7]) by the development and use of targeted multiple reaction monitoring-based triple quadrupole mass spectrometry. RESULTS In this exploratory study, 1586 proteins were identified with 151 proteins differentially expressed in HCM samples compared with controls ( P<0.05). Protein expression profiling showed that many proteins identified in the initial discovery study were associated with metabolism, muscle contraction, calcium regulation, and oxidative stress. Proteins downregulated in HCM versus controls included creatine kinase M-type, fructose-bisphosphate aldolase A, and phosphoglycerate mutase ( P<0.001). Proteins upregulated in HCM included lumican, carbonic anhydrase 3, desmin, α-actin skeletal, and FHL1 (four and a half LIM domain protein 1; P<0.01). Myocardial lumican concentration correlated with the left atrial area (ρ=0.34, P=0.015), late gadolinium enhancement on cardiac magnetic resonance imaging ( P=0.03) and the presence of a pathogenic sarcomere mutation ( P=0.04). CONCLUSIONS The myocardial proteome of HCM provides supporting evidence for dysregulation of metabolic and structural proteins. The finding that lumican is raised in HCM hearts provides insight into the myocardial fibrosis that characterizes this disease.
Collapse
Affiliation(s)
- Caroline J Coats
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.).,University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Wendy E Heywood
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Alex Virasami
- Histopathology Unit, Great Ormond Street Hospital for Children, London, United Kingdom (A.V., M.A., N.J.S.)
| | - Nadia Ashrafi
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Petros Syrris
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
| | - Cris Dos Remedios
- Department of Anatomy and Histology, Bosch Institute, The University of Sydney, New South Wales, Australia (C.d.R.)
| | - Thomas A Treibel
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.).,Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| | - James C Moon
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.).,Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| | - Luis R Lopes
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.).,Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| | - Christopher G A McGregor
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
| | - Michael Ashworth
- Histopathology Unit, Great Ormond Street Hospital for Children, London, United Kingdom (A.V., M.A., N.J.S.)
| | - Neil J Sebire
- Histopathology Unit, Great Ormond Street Hospital for Children, London, United Kingdom (A.V., M.A., N.J.S.)
| | - William J McKenna
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
| | - Kevin Mills
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Perry M Elliott
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.).,Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| |
Collapse
|
22
|
Gaertner-Rommel A, Tiesmeier J, Jakob T, Strickmann B, Veit G, Bachmann-Mennenga B, Paluszkiewicz L, Klingel K, Schulz U, Laser KT, Karger B, Pfeiffer H, Milting H. Molecular autopsy and family screening in a young case of sudden cardiac death reveals an unusually severe case of FHL1 related hypertrophic cardiomyopathy. Mol Genet Genomic Med 2019; 7:e841. [PMID: 31293105 PMCID: PMC6687666 DOI: 10.1002/mgg3.841] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a genetic cardiomyopathy with a prevalence of about 1:200. It is characterized by left ventricular hypertrophy, diastolic dysfunction and interstitial fibrosis; HCM might lead to sudden cardiac death (SCD) especially in the young. Due to low autopsy frequencies of sudden unexplained deaths (SUD) the true prevalence of SCD and especially of HCM among SUD remains unclear. Even in cases of proven SCD genetic testing is not a routine procedure precluding appropriate risk stratification and counseling of relatives. METHODS Here we report a case of SCD in a 19-year-old investigated by combined forensic and molecular autopsy. RESULTS During autopsy of the index-patient HCM was detected. As no other possible cause of death could be uncovered by forensic autopsy the event was classified as SCD. Molecular autopsy identified two (probably) pathogenic genetic variants in FHL1 and MYBPC3. The MYBPC3 variant had an incomplete penetrance. The FHL1 variant was a de novo mutation. We detected reduced FHL1 mRNA levels and no FHL1 protein in muscle samples suggesting nonsense-mediated mRNA decay and/or degradation of the truncated protein in the SCD victim revealing a plausible disease mechanism. CONCLUSION The identification of the genetic cause of the SCD contributed to the rational counseling of the relatives and risk assessment within the family. Furthermore our study revealed evidences for the pathomechanism of FHL1 mutations.
Collapse
Affiliation(s)
- Anna Gaertner-Rommel
- Klinikum der Ruhr-Universität Bochum, Klinik für Thorax- und Kardiovaskularchirurgie und Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Herz- und Diabeteszentrum NRW, Bad Oeynhausen, Germany
| | - Jens Tiesmeier
- Mühlenkreiskliniken, Krankenhaus Lübbecke-Rahden, Institut für Anästhesiologie, Intensiv- und Notfallmedizin, Medizin Campus OWL, Ruhr-Universität Bochum, Bochum, Germany
| | - Thomas Jakob
- Klinikum Herford, Universitätsklinik für Anästhesiologie, Medizin Campus OWL, Ruhr-Universität Bochum, Herford, Germany
| | | | - Gunter Veit
- Mühlenkreiskliniken, Krankenhaus Lübbecke-Rahden, Institut für Anästhesiologie, Intensiv- und Notfallmedizin, Medizin Campus OWL, Ruhr-Universität Bochum, Bochum, Germany
| | - Bernd Bachmann-Mennenga
- Mühlenkreiskliniken, Johannes Wesling Klinikum, Universitätsinstitut für Anästhesiologie, Intensiv- und Notfallmedizin, Medizin Campus OWL, Ruhr-Universität Bochum, Minden, Germany
| | - Lech Paluszkiewicz
- Klinikum der Ruhr-Universität Bochum, Klinik für Thorax- und Kardiovaskularchirurgie und Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Herz- und Diabeteszentrum NRW, Bad Oeynhausen, Germany
| | - Karin Klingel
- Kardiopathologie, Universitätsklinikum Tübingen, Institut für Pathologie und Neuropathologie, Tubingen, Germany
| | - Uwe Schulz
- Klinikum der Ruhr-Universität Bochum, Klinik für Thorax- und Kardiovaskularchirurgie und Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Herz- und Diabeteszentrum NRW, Bad Oeynhausen, Germany
| | - Kai T Laser
- Zentrum für angeborene Herzfehler, Herz- und Diabeteszentrum NRW, Klinikum der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Bernd Karger
- Universitätsklinikum Münster, Institut für Rechtsmedizin, Münster, Germany
| | - Heidi Pfeiffer
- Universitätsklinikum Münster, Institut für Rechtsmedizin, Münster, Germany
| | - Hendrik Milting
- Klinikum der Ruhr-Universität Bochum, Klinik für Thorax- und Kardiovaskularchirurgie und Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Herz- und Diabeteszentrum NRW, Bad Oeynhausen, Germany
| |
Collapse
|
23
|
Han S, Cui C, Wang Y, He H, Shen X, Chen Y, Liu Z, Zhu Q, Li D, Yin H. FHL3 negatively regulates the differentiation of skeletal muscle satellite cells in chicken. 3 Biotech 2019; 9:206. [PMID: 31139537 DOI: 10.1007/s13205-019-1735-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022] Open
Abstract
As a member of four and a half LIM domain proteins, FHL3 gene-encoded protein (FHL3) plays an important role in skeletal muscle development and contraction. In this study, we determined the potential role of FHL3 in the proliferation and differentiation of primary satellite cells in chicken. RT-qPCR results showed that FHL3 mRNA was highly expressed in skeletal muscle in 12 chicken tissues. Four cell proliferation assays (CCK8 assay; EDU staining assay; flow cytometric detection of cell cycle assay; and detection of cell proliferation marker genes Ki67 and N-Ras assay) revealed that FHL3 knockdown had no effect on the proliferation rate of chicken satellite cells. FHL3 knockdown promoted the differentiation of satellite cells into myotubes, as evidenced by increased fusion index, number of nuclei per myotube, Myog, Myh7, Myf5, and Mrf4 mRNA expressions, and myog and myosin heavy chain protein expressions of myogenic markers (P < 0.05). These results showed that the FHL3 was a negative regulator of the differentiation and fusion of chicken satellite cells into myotubes. However, FHL3 expression was increased during the differentiation of chicken satellite cells into myotubes. The study suggested that FHL3 might have different functions in chicken myotubes compared with that in chicken satellite cells.
Collapse
|
24
|
Stavusis J, Lace B, Schäfer J, Geist J, Inashkina I, Kidere D, Pajusalu S, Wright NT, Saak A, Weinhold M, Haubenberger D, Jackson S, Kontrogianni-Konstantopoulos A, Bönnemann CG. Novel mutations in MYBPC1 are associated with myogenic tremor and mild myopathy. Ann Neurol 2019; 86:129-142. [PMID: 31025394 DOI: 10.1002/ana.25494] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To define a distinct, dominantly inherited, mild skeletal myopathy associated with prominent and consistent tremor in two unrelated, three-generation families. METHODS Clinical evaluations as well as exome and panel sequencing analyses were performed in affected and nonaffected members of two families to identify genetic variants segregating with the phenotype. Histological assessment of a muscle biopsy specimen was performed in 1 patient, and quantitative tremor analysis was carried out in 2 patients. Molecular modeling studies and biochemical assays were performed for both mutations. RESULTS Two novel missense mutations in MYBPC1 (p.E248K in family 1 and p.Y247H in family 2) were identified and shown to segregate perfectly with the myopathy/tremor phenotype in the respective families. MYBPC1 encodes slow myosin binding protein-C (sMyBP-C), a modular sarcomeric protein playing structural and regulatory roles through its dynamic interaction with actin and myosin filaments. The Y247H and E248K mutations are located in the NH2 -terminal M-motif of sMyBP-C. Both mutations result in markedly increased binding of the NH2 terminus to myosin, possibly interfering with normal cross-bridge cycling as the first muscle-based step in tremor genesis. The clinical tremor features observed in all mutation carriers, together with the tremor physiology studies performed in family 2, suggest amplification by an additional central loop modulating the clinical tremor phenomenology. INTERPRETATION Here, we link two novel missense mutations in MYBPC1 with a dominant, mild skeletal myopathy invariably associated with a distinctive tremor. The molecular, genetic, and clinical studies are consistent with a unique sarcomeric origin of the tremor, which we classify as "myogenic tremor." ANN NEUROL 2019.
Collapse
Affiliation(s)
- Janis Stavusis
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Baiba Lace
- Latvian Biomedical Research and Study Centre, Riga, Latvia.,Centre Hospitalier Universitaire de Québec, Ville de Québec, QC, Canada
| | - Jochen Schäfer
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Inna Inashkina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Dita Kidere
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia.,Department of Clinical Genetics, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA
| | - Annika Saak
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | - Manja Weinhold
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | - Dietrich Haubenberger
- Clinical Trials Unit, Office of the Clinical Director, NINDS Intramural Research Program, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| | - Sandra Jackson
- Department of Neurology-Uniklinikum CG Carus, Dresden, Germany
| | | | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| |
Collapse
|
25
|
Biendarra-Tiegs SM, Li X, Ye D, Brandt EB, Ackerman MJ, Nelson TJ. Single-Cell RNA-Sequencing and Optical Electrophysiology of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Reveal Discordance Between Cardiac Subtype-Associated Gene Expression Patterns and Electrophysiological Phenotypes. Stem Cells Dev 2019; 28:659-673. [PMID: 30892143 PMCID: PMC6534093 DOI: 10.1089/scd.2019.0030] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The ability to accurately phenotype cells differentiated from human induced pluripotent stem cells (hiPSCs) is essential for their application in modeling developmental and disease processes, yet also poses a particular challenge without the context of anatomical location. Our specific objective was to determine if single-cell gene expression was sufficient to predict the electrophysiology of iPSC-derived cardiac lineages, to evaluate the concordance between molecular and functional surrogate markers. To this end, we used the genetically encoded voltage indicator ArcLight to profile hundreds of hiPSC-derived cardiomyocytes (hiPSC-CMs), thus identifying patterns of electrophysiological maturation and increased prevalence of cells with atrial-like action potentials (APs) between days 11 and 42 of differentiation. To profile expression patterns of cardiomyocyte subtype-associated genes, single-cell RNA-seq was performed at days 12 and 40 after the populations were fully characterized with the high-throughput ArcLight platform. Although we could detect global gene expression changes supporting progressive differentiation, individual cellular expression patterns alone were not able to delineate the individual cardiomyocytes into atrial, ventricular, or nodal subtypes as functionally documented by electrophysiology measurements. Furthermore, our efforts to understand the distinct electrophysiological properties associated with day 12 versus day 40 hiPSC-CMs revealed that ion channel regulators SLMAP, FGF12, and FHL1 were the most significantly increased genes at day 40, categorized by electrophysiology-related gene functions. Notably, FHL1 knockdown during differentiation was sufficient to significantly modulate APs toward ventricular-like electrophysiology. Thus, our results establish the inability of subtype-associated gene expression patterns to specifically categorize hiPSC-derived cells according to their functional electrophysiology, and yet, altered FHL1 expression is able to redirect electrophysiological maturation of these developing cells. Therefore, noncanonical gene expression patterns of cardiac maturation may be sufficient to direct functional maturation of cardiomyocytes, with canonical gene expression patterns being insufficient to temporally define cardiac subtypes of in vitro differentiation.
Collapse
Affiliation(s)
- Sherri M Biendarra-Tiegs
- 1 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,2 Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Xing Li
- 2 Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,3 Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Dan Ye
- 4 Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Emma B Brandt
- 1 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,2 Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael J Ackerman
- 4 Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,5 Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,6 Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Timothy J Nelson
- 1 Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.,2 Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota.,5 Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.,6 Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.,7 Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
26
|
Abstract
The 4-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein that can interact with cell surface receptors, cytosolic adaptor and structural proteins, kinases, and nuclear transcription factors. It is involved in numerous functional activities, including the epithelial-mesenchymal transition, cell proliferation, apoptosis, adhesion, migration, structural stability, and gene expression. Despite this, FHL2-knockout (KO) mice are viable and fertile with no obvious abnormalities, rather suggesting a high capacity for fine-tuning adjustment and functional redundancy of FHL2. Indeed, challenging FHL2-KO cells or mice provided numerous evidences for the great functional significance of FHL2. In recent years, several reviews have been published describing the high capacity of FHL2 to bind diverse proteins as well as the versatile functions of FHL2, emphasizing in particular its role in cardiovascular diseases and carcinogenesis. Here, we view the function of FHL2 from a different perspective. We summarize the published data demonstrating the impact of FHL2 on wound healing and inflammation. FHL2 seems to be involved in numerous steps of these extremely complex and multidirectional but tightly regulated tissue remodeling processes, supporting tissue repair and coordinating inflammation. Deficiency of FHL2 not only slows down ongoing wound healing but also often turns it into a chronic condition.-Wixler, V. The role of FHL2 in wound healing and inflammation.
Collapse
Affiliation(s)
- Viktor Wixler
- Centre for Molecular Biology of Inflammation, Institute of Molecular Virology, Westfaelische Wilhelms University Muenster, Muenster, Germany
| |
Collapse
|
27
|
Hashimoto S, Nobuta R, Izawa T, Inada T. Translation arrest as a protein quality control system for aberrant translation of the 3'-UTR in mammalian cells. FEBS Lett 2019; 593:777-787. [PMID: 30883710 DOI: 10.1002/1873-3468.13362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/23/2019] [Accepted: 03/11/2019] [Indexed: 01/05/2023]
Abstract
Read-through or mutations of a stop codon resulting in translation of the 3'-UTR produce potentially toxic C-terminally extended proteins. However, quality control mechanisms for such proteins are poorly understood in mammalian cells. Here, a comprehensive analysis of the 3'-UTRs of genes associated with hereditary diseases identified novel arrest-inducing sequences in the 3'-UTRs of 23 genes that can repress the levels of their protein products. In silico analysis revealed that the hydrophobicity of the polypeptides encoded in the 3'-UTRs is correlated with arrest efficiency. These results provide new insight into quality control mechanisms mediated by 3'-UTRs to prevent the production of C-terminally extended cytotoxic proteins.
Collapse
Affiliation(s)
- Satoshi Hashimoto
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan
| | - Risa Nobuta
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan
| | - Toshiaki Izawa
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan
| | - Toshifumi Inada
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Japan
| |
Collapse
|
28
|
Coats CJ, Heywood WE, Virasami A, Ashrafi N, Syrris P, dos Remedios C, Treibel TA, Moon JC, Lopes LR, McGregor CG, Ashworth M, Sebire NJ, McKenna WJ, Mills K, Elliott PM. Proteomic Analysis of the Myocardium in Hypertrophic Obstructive Cardiomyopathy. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2018. [DOI: 10.1161/circgenetics.117.001974] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Caroline J. Coats
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Wendy E. Heywood
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Alex Virasami
- Histopathology Unit, Great Ormond Street Hospital for Children, London, United Kingdom (A.V., M.A., N.J.S.)
| | - Nadia Ashrafi
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Petros Syrris
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
| | - Cris dos Remedios
- Department of Anatomy and Histology, Bosch Institute, The University of Sydney, New South Wales, Australia (C.d.R.)
| | - Thomas A. Treibel
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| | - James C. Moon
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| | - Luis R. Lopes
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| | - Christopher G.A. McGregor
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
| | - Michael Ashworth
- Histopathology Unit, Great Ormond Street Hospital for Children, London, United Kingdom (A.V., M.A., N.J.S.)
| | - Neil J. Sebire
- Histopathology Unit, Great Ormond Street Hospital for Children, London, United Kingdom (A.V., M.A., N.J.S.)
| | - William J. McKenna
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
| | - Kevin Mills
- University College London Great Ormond Street Institute of Child Health, London, United Kingdom (C.J.C., W.E.H., N.A., K.M.)
| | - Perry M. Elliott
- University College London Institute of Cardiovascular Science, London, United Kingdom (C.J.C., P.S., T.A.T., J.C.M., L.R.L., C.G.A.M., W.J.M., P.M.E.)
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom (T.A.T., J.C.M., L.R.L., P.M.E.)
| |
Collapse
|
29
|
Brull A, Morales Rodriguez B, Bonne G, Muchir A, Bertrand AT. The Pathogenesis and Therapies of Striated Muscle Laminopathies. Front Physiol 2018; 9:1533. [PMID: 30425656 PMCID: PMC6218675 DOI: 10.3389/fphys.2018.01533] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023] Open
Abstract
Emery-Dreifuss muscular dystrophy (EDMD) is a genetic condition characterized by early contractures, skeletal muscle weakness, and cardiomyopathy. During the last 20 years, various genetic approaches led to the identification of causal genes of EDMD and related disorders, all encoding nuclear envelope proteins. By their respective localization either at the inner nuclear membrane or the outer nuclear membrane, these proteins interact with each other and establish a connection between the nucleus and the cytoskeleton. Beside this physical link, these proteins are also involved in mechanotransduction, responding to environmental cues, such as increased tension of the cytoskeleton, by the activation or repression of specific sets of genes. This ability of cells to adapt to environmental conditions is altered in EDMD. Increased knowledge on the pathophysiology of EDMD has led to the development of drug or gene therapies that have been tested on mouse models. This review proposed an overview of the functions played by the different proteins involved in EDMD and related disorders and the current therapeutic approaches tested so far.
Collapse
Affiliation(s)
- Astrid Brull
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Blanca Morales Rodriguez
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France.,Sanofi R&D, Chilly Mazarin, France
| | - Gisèle Bonne
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Antoine Muchir
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Anne T Bertrand
- Sorbonne Université, INSERM, Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| |
Collapse
|
30
|
McNamara JW, Sadayappan S. Skeletal myosin binding protein-C: An increasingly important regulator of striated muscle physiology. Arch Biochem Biophys 2018; 660:121-128. [PMID: 30339776 DOI: 10.1016/j.abb.2018.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/07/2018] [Accepted: 10/11/2018] [Indexed: 12/22/2022]
Abstract
The Myosin Binding Protein-C (MyBP-C) family is a group of sarcomeric proteins important for striated muscle structure and function. Comprising approximately 2% of the myofilament mass, MyBP-C has important roles in both contraction and relaxation. Three paralogs of MyBP-C are encoded by separate genes with distinct expression profiles in striated muscle. In mammals, cardiac MyBP-C is limited to the heart, and it is the most extensively studied owing to its involvement in cardiomyopathies. However, the roles of two skeletal paralogs, slow and fast, in muscle biology remain poorly characterized. Nonetheless, both have been recently implicated in the development of skeletal myopathies. This calls for a better understanding of their function in the pathophysiology of distal arthrogryposis. This review characterizes MyBP-C as a whole and points out knowledge gaps that still remain with respect to skeletal MyBP-C.
Collapse
Affiliation(s)
- James W McNamara
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45236, USA
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45236, USA.
| |
Collapse
|
31
|
Keßler M, Kieltsch A, Kayvanpour E, Katus H, Schoser B, Schessl J, Just S, Rottbauer W. A zebrafish model for FHL1-opathy reveals loss-of-function effects of human FHL1 mutations. Neuromuscul Disord 2018; 28:521-531. [DOI: 10.1016/j.nmd.2018.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/27/2017] [Accepted: 03/01/2018] [Indexed: 11/16/2022]
|
32
|
Ding J, Peng Z, Wu D, Miao J, Liu B, Wang L. A transcriptomics study of differentiated C2C12 myoblasts identified novel functional responses to 17β-estradiol. Cell Biol Int 2018; 42:965-974. [PMID: 29570902 DOI: 10.1002/cbin.10962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/17/2018] [Indexed: 11/11/2022]
Affiliation(s)
- JingJing Ding
- Medical Research Center of Shengjing Hospital; China Medical University; Shenyang 110004 China
| | - ZhaoHong Peng
- Medical Research Center of Shengjing Hospital; China Medical University; Shenyang 110004 China
| | - Di Wu
- Medical Research Center of Shengjing Hospital; China Medical University; Shenyang 110004 China
| | - JiaNing Miao
- Medical Research Center of Shengjing Hospital; China Medical University; Shenyang 110004 China
| | - Bo Liu
- Medical Research Center of Shengjing Hospital; China Medical University; Shenyang 110004 China
| | - LiLi Wang
- Medical Research Center of Shengjing Hospital; China Medical University; Shenyang 110004 China
| |
Collapse
|
33
|
Ziat E, Mamchaoui K, Beuvin M, Nelson I, Azibani F, Spuler S, Bonne G, Bertrand AT. FHL1B Interacts with Lamin A/C and Emerin at the Nuclear Lamina and is Misregulated in Emery-Dreifuss Muscular Dystrophy. J Neuromuscul Dis 2018; 3:497-510. [PMID: 27911330 DOI: 10.3233/jnd-160169] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Emery-Dreifuss muscular dystrophy (EDMD) is associated with mutations in EMD and LMNA genes, encoding for the nuclear envelope proteins emerin and lamin A/C, indicating that EDMD is a nuclear envelope disease. We recently reported mutations in FHL1 gene in X-linked EDMD. FHL1 encodes FHL1A, and the two minor isoforms FHL1B and FHL1C. So far, none have been described at the nuclear envelope. OBJECTIVE To gain insight into the pathophysiology of EDMD, we focused our attention on the poorly characterized FHL1B isoform. METHODS The amount and the localisation of FHL1B were evaluated in control and diseased human primary myoblasts using immunofluorescence and western blotting. RESULTS We found that in addition to a cytoplasmic localization, this isoform strongly accumulated at the nuclear envelope of primary human myoblasts, like but independently of lamin A/C and emerin. During myoblast differentiation, we observed a major reduction of FHL1B protein expression, especially in the nucleus. Interestingly, we found elevated FHL1B expression level in myoblasts from an FHL1-related EDMD patient where the FHL1 mutation only affects FHL1A, as well as in myoblasts from an LMNA-related EDMD patient. CONCLUSIONS Altogether, the specific localization of FHL1B and its modulation in disease-patient's myoblasts confirmed FHL1-related EDMD as a nuclear envelope disease.
Collapse
Affiliation(s)
- Esma Ziat
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France.,Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation between Max-Delbrück-Center for Molecular Medicine and Charite Medical Faculty, Berlin, Germany
| | - Kamel Mamchaoui
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France
| | - Maud Beuvin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France
| | - Isabelle Nelson
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France
| | - Feriel Azibani
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation between Max-Delbrück-Center for Molecular Medicine and Charite Medical Faculty, Berlin, Germany
| | - Gisèle Bonne
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France
| | - Anne T Bertrand
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center of Research in Myology, F-75013 Paris, France
| |
Collapse
|
34
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
35
|
Berkholz J, Eberle R, Boller K, Munz B. siRNA-mediated inhibition of skNAC and Smyd1 expression disrupts myofibril organization: Immunofluorescence and electron microscopy study in C2C12 cells. Micron 2018; 108:6-10. [PMID: 29499397 DOI: 10.1016/j.micron.2018.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 11/30/2022]
Abstract
skNAC (skeletal and heart muscle-specific variant of nascent polypeptide-associated complex) and Smyd1 (SET and MYND domain-containing 1) form a protein dimer which is specific for striated muscle cells. Its function is largely unknown. On the one hand, skNAC-Smyd1 appears to control transcriptional processes in the nucleus, on the other hand, specifically at later stages of myogenic differentiation, both proteins translocate to the sarcoplasm and at least Smyd1 specifically associates with sarcomeric structures and might control myofibrillogenesis and/or sarcomere architecture. Here, using immunofluorescence and electron microscopy, we analyzed sarcomere formation and myofibril organization after siRNA-mediated knockdown of skNAC or Smyd1 expression in murine C2C12 skeletal muscle cells. We found that inhibition of skNAC or Smyd1 expression indeed prevents myofibrillogenesis and sarcomere formation, leading to a disorganized array of myofilaments predominantly within the region immediately beneath the plasma membrane.
Collapse
Affiliation(s)
- Janine Berkholz
- Charité - University Medicine Berlin, Institute of Physiology, Charitéplatz 1, D-10117 Berlin, Germany
| | - Regina Eberle
- Paul-Ehrlich-Institute, Paul-Ehrlich-Str. 51-59, D-63225 Langen, Germany
| | - Klaus Boller
- Paul-Ehrlich-Institute, Paul-Ehrlich-Str. 51-59, D-63225 Langen, Germany
| | - Barbara Munz
- University Hospital Tubingen - Medical Clinic - Department of Sports Medicine, Hoppe-Seyler-Str. 6, D-72076 Tubingen, Germany.
| |
Collapse
|
36
|
Charó NL, Galigniana NM, Piwien-Pilipuk G. Heterochromatin protein (HP)1γ is not only in the nucleus but also in the cytoplasm interacting with actin in both cell compartments. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:432-443. [PMID: 29208528 DOI: 10.1016/j.bbamcr.2017.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/07/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022]
Abstract
Confocal and electron microscopy images, and WB analysis of cellular fractions revealed that HP1γ is in the nucleus but also in the cytoplasm of C2C12 myoblasts, myotubes, skeletal and cardiac muscles, N2a, HeLa and HEK293T cells. Signal specificity was tested with different antibodies and by HP1γ knockdown. Leptomycin B treatment of myoblasts increased nuclear HP1γ, suggesting that its nuclear export is Crm-1-dependent. HP1γ exhibited a filamentous pattern of staining partially co-localizing with actin in the cytoplasm of myotubes and myofibrils. Immunoelectron microscopic analysis showed high-density immunogold particles that correspond to HP1γ localized to the Z-disk and A-band of the sarcomere of skeletal muscle. HP1γ partially co-localized with actin in C2C12 myotubes and murine myofibrils. Importantly, actin co-immunoprecipitated with HP1γ in the nuclear and cytosolic fractions of myoblasts. Actin co-immunoprecipitated with HP1γ in myoblasts incubated in the absence or presence of the actin depolymerizing agent cytochalasin D, suggesting that HP1γ may interact with G-and F-actin. In the cytoplasm, HP1γ was associated to the perinuclear actin cap that controls nuclear shape and position. In the nucleus, re-ChIP assays showed that HP1γ-actin associates to the promoter and transcribed regions of the house keeping gene GAPDH, suggesting that HP1γ may function as a scaffold protein for the recruitment of actin to control gene expression. When HP1γ was knocked-down, myoblasts were unable to differentiate or originated thin myotubes. In summary, HP1γ is present in the nucleus and the cytoplasm interacting with actin, a protein complex that may exert different functions depending on its subcellular localization.
Collapse
Affiliation(s)
- Nancy L Charó
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IByME) - CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia M Galigniana
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IByME) - CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Graciela Piwien-Pilipuk
- Laboratory of Nuclear Architecture, Instituto de Biología y Medicina Experimental (IByME) - CONICET, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
37
|
Exome sequencing identifies variants in two genes encoding the LIM-proteins NRAP and FHL1 in an Italian patient with BAG3 myofibrillar myopathy. J Muscle Res Cell Motil 2016; 37:101-15. [PMID: 27443559 PMCID: PMC5010835 DOI: 10.1007/s10974-016-9451-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/09/2016] [Indexed: 11/06/2022]
Abstract
Myofibrillar myopathies (MFMs) are genetically heterogeneous dystrophies characterized by the disintegration of Z-disks and myofibrils and are associated with mutations in genes encoding Z-disk or Z-disk-related proteins. The c.626 C > T (p.P209L) mutation in the BAG3 gene has been described as causative of a subtype of MFM. We report a sporadic case of a 26-year-old Italian woman, affected by MFM with axonal neuropathy, cardiomyopathy, rigid spine, who carries the c.626 C > T mutation in the BAG3 gene. The patient and her non-consanguineous healthy parents and brother were studied with whole exome sequencing (WES) to further investigate the genetic basis of this complex phenotype. In the patient, we found that the BAG3 mutation is associated with variants in the NRAP and FHL1 genes that encode muscle-specific, LIM domain containing proteins. Quantitative real time PCR, immunohistochemistry and Western blot analysis of the patient’s muscular biopsy showed the absence of NRAP expression and FHL1 accumulation in aggregates in the affected skeletal muscle tissue. Molecular dynamic analysis of the mutated FHL1 domain showed a modification in its surface charge, which could affect its capability to bind its target proteins. To our knowledge this is the first study reporting, in a BAG3 MFM, the simultaneous presence of genetic variants in the BAG3 and FHL1 genes (previously described as independently associated with MFMs) and linking the NRAP gene to MFM for the first time.
Collapse
|
38
|
Wu SM, Shih LH, Lee JY, Shen YJ, Lee HH. Estrogen enhances activity of Wnt signaling during osteogenesis by inducing Fhl1 expression. J Cell Biochem 2016; 116:1419-30. [PMID: 25676585 DOI: 10.1002/jcb.25102] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 01/23/2015] [Indexed: 01/13/2023]
Abstract
Estrogen is a crucial hormone for osteoclast inhibition and for preventing osteoporosis. However, the hormone's role in osteoblast growth and differentiation remains unclear. The complexity of estrogen's role in guiding osteoblast behavior arises partly from crosstalk with other signaling pathways, including Wnt signaling. In this study, we show that the Wnt agonist, LiCl, induced Fhl1 gene expression, which substantially enhanced osteoblast differentiation. Staining with alizarin red revealed that MC3T3-E1 mineralization was enhanced by overexpression of Fhl1. In addition, Fhl1 promoted the expression of the osteogenic markers, Runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and osteopontin (OPN), whereas MC3T3-E1 cells with gene knockdown of Fhl1 exhibited limited mineralization and expression of Runx2, OCN, and OPN. We further demonstrate evidences from quantitative reverse transcription real-time polymerase chain reaction and reporter assay that Fhl1 expression was synergistically stimulated by estrogen (E2) and LiCl, but reduced by the estrogen-receptor inhibitor fulvestrant (ICI 182,780). However, estrogen could not enhance osteogenesis while Fhl1 expression was knocked down. Because estrogen and Wnt signaling frequently interact in developmental processes, we propose that Fhl1 can be an acting molecule mediating both signaling pathways during osteogenesis.
Collapse
Affiliation(s)
- Shao-Min Wu
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Lan-Hsin Shih
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Jing-Yu Lee
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Yi-Jun Shen
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Hu-Hui Lee
- Department of Bio-Agricultural Sciences, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| |
Collapse
|
39
|
Willis T, Wood C, Hudson J, Polvikoski T, Barresi R, Lochmüller H, Bushby K, Straub V. Muscle hypertrophy as the presenting sign in a patient with a completeFHL1deletion. Clin Genet 2016; 90:166-70. [DOI: 10.1111/cge.12695] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/02/2015] [Accepted: 11/12/2015] [Indexed: 12/19/2022]
Affiliation(s)
- T.A. Willis
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
| | - C.L. Wood
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
| | - J. Hudson
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
| | - T. Polvikoski
- Institute of Neuroscience; Newcastle University; Newcastle upon Tyne UK
| | - R. Barresi
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
- Rare Diseases Advisory Group Service for Neuromuscular Diseases; Muscle Immunoanalysis Unit; Newcastle upon Tyne UK
| | - H. Lochmüller
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
| | - K. Bushby
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
| | - V. Straub
- John Walton Muscular Dystrophy Research Centre; Institute of Genetic Medicine; Newcastle upon Tyne UK
| |
Collapse
|
40
|
Albrecht I, Wick C, Hallgren Å, Tjärnlund A, Nagaraju K, Andrade F, Thompson K, Coley W, Phadke A, Diaz-Gallo LM, Bottai M, Nennesmo I, Chemin K, Herrath J, Johansson K, Wikberg A, Ytterberg AJ, Zubarev RA, Danielsson O, Krystufkova O, Vencovsky J, Landegren N, Wahren-Herlenius M, Padyukov L, Kämpe O, Lundberg IE. Development of autoantibodies against muscle-specific FHL1 in severe inflammatory myopathies. J Clin Invest 2015; 125:4612-24. [PMID: 26551678 DOI: 10.1172/jci81031] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 09/25/2015] [Indexed: 11/17/2022] Open
Abstract
Mutations of the gene encoding four-and-a-half LIM domain 1 (FHL1) are the causative factor of several X-linked hereditary myopathies that are collectively termed FHL1-related myopathies. These disorders are characterized by severe muscle dysfunction and damage. Here, we have shown that patients with idiopathic inflammatory myopathies (IIMs) develop autoimmunity to FHL1, which is a muscle-specific protein. Anti-FHL1 autoantibodies were detected in 25% of IIM patients, while patients with other autoimmune diseases or muscular dystrophies were largely anti-FHL1 negative. Anti-FHL1 reactivity was predictive for muscle atrophy, dysphagia, pronounced muscle fiber damage, and vasculitis. FHL1 showed an altered expression pattern, with focal accumulation in the muscle fibers of autoantibody-positive patients compared with a homogeneous expression in anti-FHL1-negative patients and healthy controls. We determined that FHL1 is a target of the cytotoxic protease granzyme B, indicating that the generation of FHL1 fragments may initiate FHL1 autoimmunity. Moreover, immunization of myositis-prone mice with FHL1 aggravated muscle weakness and increased mortality, suggesting a direct link between anti-FHL1 responses and muscle damage. Together, our findings provide evidence that FHL1 may be involved in the pathogenesis not only of genetic FHL1-related myopathies but also of autoimmune IIM. Importantly, these results indicate that anti-FHL1 autoantibodies in peripheral blood have promising potential as a biomarker to identify a subset of severe IIM.
Collapse
|
41
|
Tran MK, Kurakula K, Koenis DS, de Vries CJM. Protein-protein interactions of the LIM-only protein FHL2 and functional implication of the interactions relevant in cardiovascular disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:219-28. [PMID: 26548523 DOI: 10.1016/j.bbamcr.2015.11.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/02/2015] [Accepted: 11/03/2015] [Indexed: 11/26/2022]
Abstract
FHL2 belongs to the LIM-domain only proteins and contains four and a half LIM domains, each of which are composed of two zinc finger structures. FHL2 exhibits specific interaction with proteins exhibiting diverse functions, including transmembrane receptors, transcription factors and transcription co-regulators, enzymes, and structural proteins. The function of these proteins is regulated by FHL2, which modulates intracellular signal transduction pathways involved in a plethora of cellular tasks. The present review summarizes the current knowledge on the protein interactome of FHL2 and provides an overview of the functional implication of these interactions in apoptosis, migration, and regulation of nuclear receptor function. FHL2 was originally identified in the heart and there is extensive literature available on the role of FHL2 in the cardiovascular system, which is also summarized in this review.
Collapse
Affiliation(s)
- M Khang Tran
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Kondababu Kurakula
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Duco S Koenis
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
42
|
RNA sequencing reveals a slow to fast muscle fiber type transition after olanzapine infusion in rats. PLoS One 2015; 10:e0123966. [PMID: 25893406 PMCID: PMC4404103 DOI: 10.1371/journal.pone.0123966] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/02/2015] [Indexed: 11/19/2022] Open
Abstract
Second generation antipsychotics (SGAs), like olanzapine, exhibit acute metabolic side effects leading to metabolic inflexibility, hyperglycemia, adiposity and diabetes. Understanding how SGAs affect the skeletal muscle transcriptome could elucidate approaches for mitigating these side effects. Male Sprague-Dawley rats were infused intravenously with vehicle or olanzapine for 24h using a dose leading to a mild hyperglycemia. RNA-Seq was performed on gastrocnemius muscle, followed by alignment of the data with the Rat Genome Assembly 5.0. Olanzapine altered expression of 1347 out of 26407 genes. Genes encoding skeletal muscle fiber-type specific sarcomeric, ion channel, glycolytic, O2- and Ca2+-handling, TCA cycle, vascularization and lipid oxidation proteins and pathways, along with NADH shuttles and LDH isoforms were affected. Bioinformatics analyses indicate that olanzapine decreased the expression of slower and more oxidative fiber type genes (e.g., type 1), while up regulating those for the most glycolytic and least metabolically flexible, fast twitch fiber type, IIb. Protein turnover genes, necessary to bring about transition, were also up regulated. Potential upstream regulators were also identified. Olanzapine appears to be rapidly affecting the muscle transcriptome to bring about a change to a fast-glycolytic fiber type. Such fiber types are more susceptible than slow muscle to atrophy, and such transitions are observed in chronic metabolic diseases. Thus these effects could contribute to the altered body composition and metabolic disease olanzapine causes. A potential interventional strategy is implicated because aerobic exercise, in contrast to resistance exercise, can oppose such slow to fast fiber transitions.
Collapse
|
43
|
The sarcomeric M-region: a molecular command center for diverse cellular processes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:714197. [PMID: 25961035 PMCID: PMC4413555 DOI: 10.1155/2015/714197] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/08/2015] [Indexed: 02/07/2023]
Abstract
The sarcomeric M-region anchors thick filaments and withstands the mechanical stress of contractions by deformation, thus enabling distribution of physiological forces along the length of thick filaments. While the role of the M-region in supporting myofibrillar structure and contractility is well established, its role in mediating additional cellular processes has only recently started to emerge. As such, M-region is the hub of key protein players contributing to cytoskeletal remodeling, signal transduction, mechanosensing, metabolism, and proteasomal degradation. Mutations in genes encoding M-region related proteins lead to development of severe and lethal cardiac and skeletal myopathies affecting mankind. Herein, we describe the main cellular processes taking place at the M-region, other than thick filament assembly, and discuss human myopathies associated with mutant or truncated M-region proteins.
Collapse
|
44
|
Emmanuele V, Kubota A, Garcia-Diaz B, Garone C, Akman HO, Sánchez-Gutiérrez D, Escudero LM, Kariya S, Homma S, Tanji K, Quinzii CM, Hirano M. Fhl1 W122S causes loss of protein function and late-onset mild myopathy. Hum Mol Genet 2014; 24:714-26. [PMID: 25274776 DOI: 10.1093/hmg/ddu490] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A member of the four-and-a-half-LIM (FHL) domain protein family, FHL1, is highly expressed in human adult skeletal and cardiac muscle. Mutations in FHL1 have been associated with diverse X-linked muscle diseases: scapuloperoneal (SP) myopathy, reducing body myopathy, X-linked myopathy with postural muscle atrophy, rigid spine syndrome (RSS) and Emery-Dreifuss muscular dystrophy. In 2008, we identified a missense mutation in the second LIM domain of FHL1 (c.365 G>C, p.W122S) in a family with SP myopathy. We generated a knock-in mouse model harboring the c.365 G>C Fhl1 mutation and investigated the effects of this mutation at three time points (3-5 months, 7-10 months and 18-20 months) in hemizygous male and heterozygous female mice. Survival was comparable in mutant and wild-type animals. We observed decreased forelimb strength and exercise capacity in adult hemizygous male mice starting from 7 to 10 months of age. Western blot analysis showed absence of Fhl1 in muscle at later stages. Thus, adult hemizygous male, but not heterozygous female, mice showed a slowly progressive phenotype similar to human patients with late-onset muscle weakness. In contrast to SP myopathy patients with the FHL1 W122S mutation, mutant mice did not manifest cytoplasmic inclusions (reducing bodies) in muscle. Because muscle weakness was evident prior to loss of Fhl1 protein and without reducing bodies, our findings indicate that loss of function is responsible for the myopathy in the Fhl1 W122S knock-in mice.
Collapse
Affiliation(s)
- Valentina Emmanuele
- Department of Neurology Pediatric Clinic, Istituto di Ricovero e Cura a Carattere Scientifico G. Gaslini, University of Genoa, Genoa 16100, Italy and
| | | | | | | | | | - Daniel Sánchez-Gutiérrez
- Departamento de Biología Celular, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universdad de Sevilla, 41013 Seville, Spain
| | - Luis M Escudero
- Departamento de Biología Celular, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universdad de Sevilla, 41013 Seville, Spain
| | | | - Shunichi Homma
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Kurenai Tanji
- Department of Neurology Department of Pathology and Cell Biology
| | | | | |
Collapse
|
45
|
Sabatelli P, Castagnaro S, Tagliavini F, Chrisam M, Sardone F, Demay L, Richard P, Santi S, Maraldi NM, Merlini L, Sandri M, Bonaldo P. Aggresome-Autophagy Involvement in a Sarcopenic Patient with Rigid Spine Syndrome and a p.C150R Mutation in FHL1 Gene. Front Aging Neurosci 2014; 6:215. [PMID: 25191266 PMCID: PMC4137286 DOI: 10.3389/fnagi.2014.00215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/04/2014] [Indexed: 11/13/2022] Open
Abstract
The four-and-half LIM domain protein 1 (FHL1) is highly expressed in skeletal and cardiac muscle. Mutations of the FHL1 gene have been associated with diverse chronic myopathies including reducing body myopathy, rigid spine syndrome (RSS), and Emery–Dreifuss muscular dystrophy. We investigated a family with a mutation (p.C150R) in the second LIM domain of FHL1. In this family, a brother and a sister were affected by RSS, and their mother had mild lower limbs weakness. The 34-year-old female had an early and progressive rigidity of the cervical spine and severe respiratory insufficiency. Muscle mass evaluated by DXA was markedly reduced, while fat mass was increased to 40%. CT scan showed an almost complete substitution of muscle by fibro-adipose tissue. Muscle biopsy showed accumulation of FHL1 throughout the cytoplasm and around myonuclei into multiprotein aggregates with aggresome/autophagy features as indicated by ubiquitin, p62, and LC3 labeling. DNA deposits, not associated with nuclear lamina components and histones, were also detected in the aggregates, suggesting nuclear degradation. Ultrastructural analysis showed the presence of dysmorphic nuclei, accumulation of tubulofilamentous and granular material, and perinuclear accumulation of autophagic vacuoles. These data point to involvement of the aggresome–autophagy pathway in the pathophysiological mechanism underlying the muscle pathology of FHL1 C150R mutation.
Collapse
Affiliation(s)
- Patrizia Sabatelli
- Institute of Molecular Genetics, CNR-National Research Council of Italy , Bologna , Italy ; SC Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Silvia Castagnaro
- Department of Molecular Medicine, University of Padova , Padova , Italy
| | - Francesca Tagliavini
- Institute of Molecular Genetics, CNR-National Research Council of Italy , Bologna , Italy ; SC Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Martina Chrisam
- Department of Molecular Medicine, University of Padova , Padova , Italy
| | - Francesca Sardone
- Institute of Molecular Genetics, CNR-National Research Council of Italy , Bologna , Italy ; SC Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Laurence Demay
- UF Cardiogénétique et Myogénétique, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière , Paris , France
| | - Pascale Richard
- UF Cardiogénétique et Myogénétique, Centre de Génétique, Hôpitaux Universitaires de la Pitié Salpêtrière , Paris , France
| | - Spartaco Santi
- Institute of Molecular Genetics, CNR-National Research Council of Italy , Bologna , Italy ; SC Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Nadir M Maraldi
- Institute of Molecular Genetics, CNR-National Research Council of Italy , Bologna , Italy
| | - Luciano Merlini
- SC Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Marco Sandri
- Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine , Padova , Italy ; Department of Biomedical Science, University of Padova , Padova , Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
46
|
Semsarian C, Ingles J. Expanding the genetic spectrum of hypertrophic cardiomyopathy: X marks the spot. ACTA ACUST UNITED AC 2014; 6:528-30. [PMID: 24347617 DOI: 10.1161/circgenetics.113.000377] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Newtown, Australia
| | | |
Collapse
|
47
|
Elf K, Shevchenko G, Nygren I, Larsson L, Bergquist J, Askmark H, Artemenko K. Alterations in muscle proteome of patients diagnosed with amyotrophic lateral sclerosis. J Proteomics 2014; 108:55-64. [PMID: 24846852 DOI: 10.1016/j.jprot.2014.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/24/2014] [Accepted: 05/11/2014] [Indexed: 12/13/2022]
Abstract
UNLABELLED Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by progressive muscle paralysis. Currently clinical tools for ALS diagnostics do not perform well enough and their improvement is needed. The objective of this study was to identify specific protein alterations related to the development of ALS using tiny muscle biopsies. We applied a shotgun proteomics and quantitative dimethyl labeling in order to analyze the global changes in human skeletal muscle proteome of ALS versus healthy subjects for the first time. 235 proteins were quantified and 11 proteins were found significantly regulated in ALS muscles. These proteins are involved in muscle development and contraction, metabolic processes, enzyme activity, regulation of apoptosis and transport activity. In order to eliminate a risk to confuse ALS with other denervations, muscle biopsies of patients with postpolio syndrome and Charcot-Marie-Tooth disease (negative controls) were compared to those of ALS and controls. Only few proteins significantly regulated in ALS patients compared to controls were affected differently in negative controls. These proteins (BTB and kelch domain-containing protein 10, myosin light chain 3, glycogen debranching enzyme, transitional endoplasmic reticulum ATPase), individually or as a panel, could be selected for estimation of ALS diagnosis and development. BIOLOGICAL SIGNIFICANCE ALS is a devastating neurodegenerative disease, and luckily, very rare: only one to two people out of 100,000 develop ALS yearly. This fact, however, makes studies of ALS very challenging since it is very difficult to collect the representative set of clinical samples and this may take up to several years. In this study we collected the muscle biopsies from 12 ALS patients and compared the ALS muscle proteome against the one from control subjects. We suggested the efficient method for such comprehensive quantitative analysis by LC-MS and performed it for the first time using human ALS material. This gel- and antibody-free method can be widely applied for muscle proteome studies and has been used by us for revealing of the specific protein alterations associated with ALS.
Collapse
Affiliation(s)
- Kristin Elf
- Department of Neuroscience, Unit of Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Ganna Shevchenko
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden
| | - Ingela Nygren
- Department of Neuroscience, Unit of Neurology, Uppsala University, Uppsala, Sweden
| | - Lars Larsson
- Department of Neuroscience, Unit of Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden
| | - Håkan Askmark
- Department of Neuroscience, Unit of Neurology, Uppsala University, Uppsala, Sweden
| | - Konstantin Artemenko
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
48
|
Bertrand AT, Bönnemann CG, Bonne G. 199th ENMC international workshop: FHL1 related myopathies, June 7-9, 2013, Naarden, The Netherlands. Neuromuscul Disord 2014; 24:453-62. [PMID: 24613424 DOI: 10.1016/j.nmd.2014.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/04/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Anne T Bertrand
- Inserm, U974, Paris F-75013, France; Sorbonne Universités, UPMC Univ Paris 06, Myology Center of Research, UM76; CNRS FRE 3617, Institut de Myologie, Paris F-75013, France
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Gisèle Bonne
- Inserm, U974, Paris F-75013, France; Sorbonne Universités, UPMC Univ Paris 06, Myology Center of Research, UM76; CNRS FRE 3617, Institut de Myologie, Paris F-75013, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, U.F. Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, Paris F-75013, France.
| | | |
Collapse
|
49
|
Wang LL, Peng ZH, Fan Y, Li LY, Wu D, Zhang Y, Miao JN, Bai YZ, Yuan ZW, Wang WL, Sun KL. Dynamic expression of molecules that control limb muscle development including Fhl1 in hind limbs of different gestational age. APMIS 2014; 122:766-71. [PMID: 24475781 DOI: 10.1111/apm.12217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/21/2013] [Indexed: 01/07/2023]
Abstract
Muscle abnormality could be a key reason for congenital clubfoot (CCF) deformity, which manifests itself during fetal development. FHL1 down-regulated expression is involved in the formation of skeletal muscle abnormalities in CCF and FHL1 gene mutations contribute to the development of some kinds of myopathies. Therefore, detecting dynamic expression of Fhl1 and other molecules (Hgf, MyoD1, Myogenin, and Myh4) that control limb muscle development in hind limbs of different gestational age will provide a foundation for further research on the molecular mechanism involves in the myopathies or CCF. The dynamic gene expression levels of Fhl1, Hgf, MyoD1, Myogenin, and Myh4 in the lower limbs of E16, E17, E19, and E20 rat embryos were examined by real-time RT-PCR. Immunofluorescence was used to detect formation of specific muscle fibers (fast or slow fibers) in distal E17 hind limbs. The expression levels of Fhl1, Hgf, MyoD1, Myogenin, and Myh4 were varying in hind limbs of different gestational age. Real-time PCR results showed that all the genes that control skeletal muscle development except for Fhl1 exhibited a peak in E17 lower limbs. Immunofluorescence results showed obviously positive fast-myosin in the distal E17 lower limbs and meanwhile slow-myosin had no apparently signals. E17 was a critical time point for terminal skeletal muscle differentiation in the lower limbs of rat embryos.
Collapse
Affiliation(s)
- Li-Li Wang
- Key laboratory of health ministry for congenital malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wilding BR, McGrath MJ, Bonne G, Mitchell CA. FHL1 mutations that cause clinically distinct human myopathies form protein aggregates and impair myoblast differentiation. J Cell Sci 2014; 127:2269-81. [DOI: 10.1242/jcs.140905] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
FHL1 mutations cause several clinically heterogeneous myopathies including Reducing Body Myopathy (RBM), Scapuloperoneal Myopathy (SPM) and X-Linked Myopathy with Postural Muscle Atrophy (XMPMA). The molecular mechanisms underlying the pathogenesis of FHL1 myopathies are unknown. Protein aggregates designated “Reducing Bodies” (RBs) containing mutant FHL1 are detected in RBM muscle but not several other FHL1 myopathies. Here RBM, SPM and XMPMA FHL1 mutants were expressed in C2C12 cells and showed equivalent protein expression to wild-type FHL1 and formed aggregates positive for the RB stain Menadione-NBT, analogous to RBM muscle aggregates. However HCM and EDMD FHL1 mutants generally exhibited reduced expression. Wild-type FHL1 promotes myoblast differentiation however RBM, SPM and XMPMA mutations impaired differentiation, consistent with loss-of-normal function. Furthermore, SPM and XMPMA mutants retarded myotube formation relative to vector control consistent with a dominant-negative/toxic function. Mutant FHL1 myotube formation was partially rescued by expression of the FHL1-binding partner constitutively-active NFATc1. This is the first study to show FHL1 mutations identified in several clinically distinct myopathies lead to similar protein aggregation and impaired myotube formation suggesting a common pathogenic mechanism despite heterogenous clinical features.
Collapse
|