1
|
Hespe S, Gray B, Puranik R, Peters S, Sweeting J, Ingles J. The role of genetic testing in management and prognosis of individuals with inherited cardiomyopathies. Trends Cardiovasc Med 2025; 35:34-44. [PMID: 39004295 DOI: 10.1016/j.tcm.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Inherited cardiomyopathies are a heterogeneous group of heart muscle conditions where disease classification has traditionally been based on clinical characteristics. However, this does not always align with genotype. While there are well described challenges of genetic testing, understanding the role of genotype in patient management is increasingly required. We take a gene-by-gene approach, reviewing current evidence for the role of genetic testing in guiding prognosis and management of individuals with inherited cardiomyopathies. In particular, focusing on causal variants in genes definitively associated with arrhythmogenic cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy. This review identifies genotype-specific disease sub-groups with strong evidence supporting the use of genetics in clinical management and highlights that at present, the spectrum of clinical utility is not reflected in current guidelines. Of 13 guideline or expert consensus statements for management of cardiomyopathies, there are seven gene-specific therapeutic recommendations that have been published from four documents. Understanding how genotype influences phenotype provides evidence for the role of genetic testing for prognostic and therapeutic purposes, moving us closer to precision-medicine based care.
Collapse
Affiliation(s)
- Sophie Hespe
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Australia
| | - Belinda Gray
- Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Rajesh Puranik
- Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Stacey Peters
- Department of Cardiology and Genomic Medicine, Royal Melbourne Hospital, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Joanna Sweeting
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
2
|
Leitner L, Schultheis M, Hofstetter F, Rudolf C, Fuchs C, Kizner V, Fiedler K, Konrad MT, Höbaus J, Genini M, Kober J, Ableitner E, Gmaschitz T, Walder D, Weitzer G. An autocrine synergistic desmin-SPARC network promotes cardiomyogenesis in cardiac stem cells. Cells Dev 2024; 181:203990. [PMID: 39734020 DOI: 10.1016/j.cdev.2024.203990] [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: 04/19/2024] [Revised: 11/15/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
The mammalian heart contains cardiac stem cells throughout life, but it has not been possible to harness or stimulate these cells to repair damaged myocardium in vivo. Assuming physiological relevance of these cells, which have evolved and have been maintained throughout mammalian evolution, we hypothesize that cardiac stem cells may contribute to cardiomyogenesis in an unorthodox manner. Since the intermediate filament protein desmin and the matricellular Secreted Protein Acidic and Rich in Cysteine (SPARC) promote cardiomyogenic differentiation during embryogenesis in a cell-autonomous and paracrine manner, respectively, we focus on their genes and employ mouse embryonic and cardiac stem cell lines as in vitro models to ask whether desmin and SPARC cooperatively influence cardiomyogenesis in cardiac stem and progenitor cells. We show that desmin also promotes cardiomyogenesis in a non-cell autonomous manner by increasing the expression and secretion of SPARC in differentiating embryonic stem cells. SPARC is also secreted by cardiac stem cells where it promotes cardiomyogenesis in an autocrine and concentration-dependent manner by upregulating the expression of myocardial transcription factors and its elicitor desmin. Desmin and SPARC interact genetically, forming a positive feedback loop and secreted autocrine and paracrine SPARC negatively affects sparc mRNA expression. Paracrine SPARC rescues cardiomyogenic desmin-haploinsufficiency in cardiac stem cells in a glycosylation-dependent manner, increases desmin expression, the phosphorylation of Smad2 and induces the expression of gata4, nkx2.5 and mef2C. Demonstration that desmin-induced autocrine secretion of SPARC in cardiac stem cells promotes cardiomyogenesis raises the possibility that a physiological function of cardiac stem cells in the adult and aging heart may be the gland-like secretion of factors such as SPARC that modulate age-related and adverse environmental influences and thereby contribute to cardiac homeostasis throughout life.
Collapse
Affiliation(s)
- Lucia Leitner
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Martina Schultheis
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Franziska Hofstetter
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Claudia Rudolf
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Christiane Fuchs
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA; Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Valeria Kizner
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Kerstin Fiedler
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Marie-Therese Konrad
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Julia Höbaus
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Marco Genini
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Julia Kober
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Elisabeth Ableitner
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Teresa Gmaschitz
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Diana Walder
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Georg Weitzer
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria.
| |
Collapse
|
3
|
Bulangalire N, Claeyssen C, Agbulut O, Cieniewski-Bernard C. Impact of MG132 induced-proteotoxic stress on αB-crystallin and desmin phosphorylation and O-GlcNAcylation and their partition towards cytoskeleton. Biochimie 2024; 226:121-135. [PMID: 38636798 DOI: 10.1016/j.biochi.2024.04.004] [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: 01/16/2024] [Revised: 03/19/2024] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
Small Heat Shock Proteins are considered as the first line of defense when proteostasis fails. Among them, αB-crystallin is expressed in striated muscles in which it interacts with desmin intermediate filaments to stabilize them, maintaining cytoskeleton's integrity and muscular functionalities. Desmin is a key actor for muscle health; its targeting by αB-crystallin is thus crucial, especially in stress conditions. αB-crystallin is phosphorylated and O-GlcNAcylated. Its phosphorylation increases consecutively to various stresses, correlated with its recruitment for cytoskeleton's safeguarding. However, phosphorylation as unique signal for cytoskeleton translocation remains controversial; indeed, O-GlcNAcylation was also proposed to be involved. Thus, there are still some gaps for a deeper comprehension of how αB-crystallin functions are finely regulated by post-translational modifications. Furthermore, desmin also bears both post-translational modifications; while desmin phosphorylation is closely linked to desmin intermediates filaments turnover, it is unclear whereas its O-GlcNAcylation could impact its proper function. In the herein paper, we aim at identifying whether phosphorylation and/or O-GlcNAcylation are involved in αB-crystallin targeting towards cytoskeleton in proteotoxic stress induced by proteasome inhibition in C2C12 myotubes. We demonstrated that proteotoxicity led to αB-crystallin's phosphorylation and O-GlcNAcylation patterns changes, both presenting a dynamic interplay depending on protein subfraction. Importantly, both post-translational modifications showed a spatio-temporal variation correlated with αB-crystallin translocation towards cytoskeleton. In contrast, we did not detect any change of desmin phosphorylation and O-GlcNAcylation. All together, these data strongly support that αB-crystallin phosphorylation/O-GlcNAcylation interplay rather than changes on desmin is a key regulator for its cytoskeleton translocation, preserving it towards stress.
Collapse
Affiliation(s)
- Nathan Bulangalire
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France; CHU Lille, Université de Lille, F-59000, Lille, France; Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Charlotte Claeyssen
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Caroline Cieniewski-Bernard
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France.
| |
Collapse
|
4
|
Findlay AR. Dominantly inherited muscle disorders: understanding their complexity and exploring therapeutic approaches. Dis Model Mech 2024; 17:dmm050720. [PMID: 39501809 PMCID: PMC11574355 DOI: 10.1242/dmm.050720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
Treatments for disabling and life-threatening hereditary muscle disorders are finally close to becoming a reality. Research has thus far focused primarily on recessive forms of muscle disease. The gene replacement strategies that are commonly employed for recessive, loss-of-function disorders are not readily translatable to most dominant myopathies owing to the presence of a normal chromosome in each nucleus, hindering the development of novel treatments for these dominant disorders. This is largely due to their complex, heterogeneous disease mechanisms that require unique therapeutic approaches. However, as viral and RNA interference-based therapies enter clinical use, key tools are now in place to develop treatments for dominantly inherited disorders of muscle. This article will review what is known about dominantly inherited disorders of muscle, specifically their genetic basis, how mutations lead to disease, and the pathomechanistic implications for therapeutic approaches.
Collapse
Affiliation(s)
- Andrew R Findlay
- Washington University Saint Louis, Neuromuscular Disease Center, 660 S. Euclid Ave., St Louis, MO 63110, USA
| |
Collapse
|
5
|
Wilson VD, Bommart S, Passerieux E, Thomas C, Pincemail J, Picot MC, Mercier J, Portet F, Arbogast S, Laoudj-Chenivesse D. Muscle strength, quantity and quality and muscle fat quantity and their association with oxidative stress in patients with facioscapulohumeral muscular dystrophy: Effect of antioxidant supplementation. Free Radic Biol Med 2024; 219:112-126. [PMID: 38574978 DOI: 10.1016/j.freeradbiomed.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
The purpose of this study was to identify causes of quadriceps muscle weakness in facioscapulohumeral muscular dystrophy (FSHD). To this aim, we evaluated quadriceps muscle and fat volumes by magnetic resonance imaging and their relationships with muscle strength and oxidative stress markers in adult patients with FSHD (n = 32) and healthy controls (n = 7), and the effect of antioxidant supplementation in 20 of the 32 patients with FSHD (n = 10 supplementation and n = 10 placebo) (NCT01596803). Compared with healthy controls, the dominant quadriceps strength and quality (muscle strength per unit of muscle volume) were decreased in patients with FSHD. In addition, fat volume was increased, without changes in total muscle volume. Moreover, in patients with FSHD, the lower strength of the non-dominant quadriceps was associated with lower muscle quality compared with the dominant muscle. Antioxidant supplementation significantly changed muscle and fat volumes in the non-dominant quadriceps, and muscle quality in the dominant quadriceps. This was associated with improved muscle strength (both quadriceps) and antioxidant response. These findings suggest that quadriceps muscle strength decline may not be simply explained by atrophy and may be influenced also by the muscle intrinsic characteristics. As FSHD is associated with increased oxidative stress, supplementation might reduce oxidative stress and increase antioxidant defenses, promoting changes in muscle function.
Collapse
Affiliation(s)
- Vinicius Dias Wilson
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Centro Universitário Estácio de Belo Horizonte, Minas Gerais, Brazil.
| | - Sébastien Bommart
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Radiology, CHU of Montpellier, Arnaud de Villeneuve Hospital, 34090, Montpellier, France.
| | - Emilie Passerieux
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| | - Claire Thomas
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; LBEPS, Univ Evry, IRBA, University Paris Saclay, 91025, Evry, France.
| | - Joël Pincemail
- Department of CREDEC, Department of Medical Chemistry, University Hospital of Liege, Sart Tilman, Liege, Belgium.
| | - Marie Christine Picot
- Department of Biostatistics and Epidemiology, University Hospital, Montpellier, France; CIC 1001-INSERM, Montpellier, France.
| | - Jacques Mercier
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Clinical Physiology, CHU of Montpellier, Montpellier, France.
| | - Florence Portet
- Department of Clinical Physiology, CHU of Montpellier, Montpellier, France; U1061 INSERM, CHU de Montpellier, Montpellier University, France.
| | - Sandrine Arbogast
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| | - Dalila Laoudj-Chenivesse
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Clinical Physiology, CHU of Montpellier, Montpellier, France.
| |
Collapse
|
6
|
Bermudez-Jimenez FJ, Protonotarios A, García-Hernández S, Pérez Asensio A, Rampazzo A, Zorio E, Brodehl A, Arias MA, Macías-Ruiz R, Fernández-Armenta J, Remior Perez P, Muñoz-Esparza C, Pilichou K, Bauce B, Merino JL, Moliner-Abós C, Ochoa JP, Barriales-Villa R, Garcia-Pavia P, Lopes LR, Syrris P, Corrado D, Elliott PM, McKenna WJ, Jimenez-Jaimez J. Phenotype and Clinical Outcomes in Desmin-Related Arrhythmogenic Cardiomyopathy. JACC Clin Electrophysiol 2024; 10:1178-1190. [PMID: 38727660 DOI: 10.1016/j.jacep.2024.02.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Desmin (DES) pathogenic variants cause a small proportion of arrhythmogenic cardiomyopathy (ACM). Outcomes data on DES-related ACM are scarce. OBJECTIVES This study sought to provide information on the clinical phenotype and outcomes of patients with ACM caused by pathogenic variants of the DES gene in a multicenter cohort. METHODS We collected phenotypic and outcomes data from 16 families with DES-related ACM from 10 European centers. We assessed in vitro DES aggregates. Major cardiac events were compared to historical controls with lamin A/C truncating variant (LMNA-tv) and filament C truncating variant (FLNC-tv) ACM. RESULTS Of 82 patients (54% males, median age: 36 years), 11 experienced sudden cardiac death (SCD) (n = 7) or heart failure death (HFd)/heart transplantation (HTx) (n = 4) before clinical evaluation. Among 68 survivors, 59 (86%) presented signs of cardiomyopathy, with left ventricular (LV) dominant (50%) or biventricular (34%) disease. Mean LV ejection fraction was 51% ± 13%; 36 of 53 had late gadolinium enhancement (ring-like pattern in 49%). During a median of 6.73 years (Q1-Q3: 3.55-9.52 years), the composite endpoint (sustained ventricular tachycardia, aborted SCD, implantable cardioverter-defibrillator therapy, SCD, HFd, and HTx) was achieved in 15 additional patients with HFd/HTx (n = 5) and SCD/aborted SCD/implantable cardioverter-defibrillator therapy/sustained ventricular tachycardia (n = 10). Male sex (P = 0.004), nonsustained ventricular tachycardia (P = 0.017) and LV ejection fraction ≤50% (P = 0.012) were associated with the composite endpoint. Males with DES variants had similar outcomes to historical FLNC-tv and LMNA-tv controls. However, females showed better outcomes than those with LMNA-tv. In vitro experiments showed the characteristic finding of DES aggregates in 7 of 12 variants. CONCLUSIONS DES ACM is associated with poor outcomes which can be predicted with potentially successful treatments, underscoring the importance of familial evaluation and genetic studies to identify at risk individuals.
Collapse
Affiliation(s)
- Francisco J Bermudez-Jimenez
- Department of Cardiology, Virgen de las Nieves University Hospital, Granada, Spain; Instituto de Investigación Biosanitaria. ibs.GRANADA, Granada, Spain
| | - Alexandros Protonotarios
- Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, London, United Kingdom; Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Soledad García-Hernández
- Instituto de Investigación Biomédica de A Coruña (INIBIC, CIBERCV), A Coruña, Spain; Health in Code SL, Cardiología, A Coruña, Spain
| | - Ana Pérez Asensio
- Department of Cardiology, Puerta del Mar University Hospital, Cádiz, Spain; Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cádiz, Spain
| | - Alessandra Rampazzo
- Departments of Biology and Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Esther Zorio
- Inherited Cardiac Diseases Unit, Cardiology Department at Hospital Universitario y Politécnico La Fe and Research Group on Inherited Heart Diseases, Sudden Death and Mechanisms of Disease (CaFaMuSMe), Barcelona, Spain; Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Miguel A Arias
- Arrhythmia Unit, Hospital Universitario de Toledo, Toledo, Spain
| | - Rosa Macías-Ruiz
- Department of Cardiology, Virgen de las Nieves University Hospital, Granada, Spain; Instituto de Investigación Biosanitaria. ibs.GRANADA, Granada, Spain
| | - Juan Fernández-Armenta
- Department of Cardiology, Puerta del Mar University Hospital, Cádiz, Spain; Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cádiz, Spain
| | - Paloma Remior Perez
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Inherited Cardiac Disease Unit, Hospital Universitario Virgen Arrixaca, Murcia, Spain
| | - Carmen Muñoz-Esparza
- Inherited Cardiac Disease Unit, Hospital Universitario Virgen Arrixaca, Murcia, Spain
| | - Kalliopi Pilichou
- Departments of Biology and Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Barbara Bauce
- Departments of Biology and Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Jose L Merino
- Viamed Santa Elena and La Paz University Hospitals, Idipaz, Madrid, Spain
| | - Carlos Moliner-Abós
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; IIB-SantPau, CIBERCV, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Juan P Ochoa
- Instituto de Investigación Biomédica de A Coruña (INIBIC, CIBERCV), A Coruña, Spain; Health in Code SL, Cardiología, A Coruña, Spain
| | - Roberto Barriales-Villa
- Instituto de Investigación Biomédica de A Coruña (INIBIC, CIBERCV), A Coruña, Spain; Department of Cardiology, Complexo Hospitalario Universitario A Coruña, Spain
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain; Inherited Cardiac Disease Unit, Hospital Universitario Virgen Arrixaca, Murcia, Spain; Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Luis R Lopes
- Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, London, United Kingdom; Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Domenico Corrado
- Departments of Biology and Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Perry M Elliott
- Inherited Cardiovascular Diseases Unit, St Bartholomew's Hospital, London, United Kingdom; Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - William J McKenna
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, United Kingdom; Instituto de Investigación Biomédica de A Coruña (INIBIC, CIBERCV), A Coruña, Spain
| | - Juan Jimenez-Jaimez
- Department of Cardiology, Virgen de las Nieves University Hospital, Granada, Spain; Instituto de Investigación Biosanitaria. ibs.GRANADA, Granada, Spain.
| |
Collapse
|
7
|
Berwanger C, Terres D, Pesta D, Eggers B, Marcus K, Wittig I, Wiesner RJ, Schröder R, Clemen CS. Immortalised murine R349P desmin knock-in myotubes exhibit a reduced proton leak and decreased ADP/ATP translocase levels in purified mitochondria. Eur J Cell Biol 2024; 103:151399. [PMID: 38412640 DOI: 10.1016/j.ejcb.2024.151399] [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: 11/10/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024] Open
Abstract
Desmin gene mutations cause myopathies and cardiomyopathies. Our previously characterised R349P desminopathy mice, which carry the ortholog of the common human desmin mutation R350P, showed marked alterations in mitochondrial morphology and function in muscle tissue. By isolating skeletal muscle myoblasts from offspring of R349P desminopathy and p53 knock-out mice, we established an immortalised cellular disease model. Heterozygous and homozygous R349P desmin knock-in and wild-type myoblasts could be well differentiated into multinucleated spontaneously contracting myotubes. The desminopathy myoblasts showed the characteristic disruption of the desmin cytoskeleton and desmin protein aggregation, and the desminopathy myotubes showed the characteristic myofibrillar irregularities. Long-term electrical pulse stimulation promoted myotube differentiation and markedly increased their spontaneous contraction rate. In both heterozygous and homozygous R349P desminopathy myotubes, this treatment restored a regular myofibrillar cross-striation pattern as seen in wild-type myotubes. High-resolution respirometry of mitochondria purified from myotubes by density gradient ultracentrifugation revealed normal oxidative phosphorylation capacity, but a significantly reduced proton leak in mitochondria from the homozygous R349P desmin knock-in cells. Consistent with a reduced proton flux across the inner mitochondrial membrane, our quantitative proteomic analysis of the purified mitochondria revealed significantly reduced levels of ADP/ATP translocases in the homozygous R349P desmin knock-in genotype. As this alteration was also detected in the soleus muscle of R349P desminopathy mice, which, in contrast to the mitochondria purified from cultured cells, showed a variety of other dysregulated mitochondrial proteins, we consider this finding to be an early step in the pathogenesis of secondary mitochondriopathy in desminopathy.
Collapse
Affiliation(s)
- Carolin Berwanger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Dominic Terres
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Dominik Pesta
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), Medical Faculty, and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Britta Eggers
- Medizinisches Proteom-Center, Medical Faculty, and Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Medical Faculty, and Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Ilka Wittig
- Functional Proteomics, Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany
| | - Rudolf J Wiesner
- Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Department of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
8
|
Hespe S, Ingles J. Expanding the Phenotypic Spectrum of Desminopathy. JACC Clin Electrophysiol 2024; 10:1191-1193. [PMID: 38925721 DOI: 10.1016/j.jacep.2024.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 03/31/2024] [Indexed: 06/28/2024]
Affiliation(s)
- Sophie Hespe
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, University of New South Wales (UNSW) Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
9
|
Polavarapu K, O'Neil D, Thompson R, Spendiff S, Nandeesh B, Vengalil S, Huddar A, Baskar D, Arunachal G, Kotambail A, Bhatia S, Tumulu SK, Matalonga L, Töpf A, Laurie S, Zeldin J, Nashi S, Unnikrishnan G, Nalini A, Lochmüller H. Partial loss of desmin expression due to a leaky splice site variant in the human DES gene is associated with neuromuscular transmission defects. Neuromuscul Disord 2024; 39:10-18. [PMID: 38669730 DOI: 10.1016/j.nmd.2024.03.011] [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: 07/31/2023] [Revised: 02/15/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024]
Abstract
Recessive desminopathies are rare and often present as severe early-onset myopathy. Here we report a milder phenotype in three unrelated patients from southern India (2 M, 1F) aged 16, 21, and 22 years, who presented with childhood-onset, gradually progressive, fatigable limb-girdle weakness, ptosis, speech and swallowing difficulties, without cardiac involvement. Serum creatine kinase was elevated, and repetitive nerve stimulation showed decrement in all. Clinical improvement was noted with pyridostigmine and salbutamol in two patients. All three patients had a homozygous substitution in intron 5: DES(NM_001927.4):c.1023+5G>A, predicted to cause a donor splice site defect. Muscle biopsy with ultrastructural analysis suggested myopathy with myofibrillar disarray, and immunohistochemistry showed partial loss of desmin with some residual staining, while western blot analysis showed reduced desmin. RT-PCR of patient muscle RNA revealed two transcripts: a reduced normal desmin transcript and a larger abnormal transcript suggesting leaky splicing at the intron 5 donor site. Sequencing of the PCR products confirmed the inclusion of intron 5 in the longer transcript, predicted to cause a premature stop codon. Thus, we provide evidence for a leaky splice site causing partial loss of desmin associated with a unique phenotypic presentation of a milder form of desmin-related recessive myopathy overlapping with congenital myasthenic syndrome.
Collapse
Affiliation(s)
- Kiran Polavarapu
- Childrens Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Daniel O'Neil
- Childrens Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Rachel Thompson
- Childrens Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Sally Spendiff
- Childrens Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Bevinahalli Nandeesh
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Akshata Huddar
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Dipti Baskar
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | | | - Saloni Bhatia
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Seetam Kumar Tumulu
- Department of Neuroradiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Leslie Matalonga
- Centro Nacional de Análisis Genómico, Baldiri Reixac 4, Barcelona 08028, Spain
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Steven Laurie
- Centro Nacional de Análisis Genómico, Baldiri Reixac 4, Barcelona 08028, Spain
| | - Joshua Zeldin
- Childrens Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | | | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| | - Hanns Lochmüller
- Childrens Hospital of Eastern Ontario Research Institute, Ottawa, Canada; Centro Nacional de Análisis Genómico, Baldiri Reixac 4, Barcelona 08028, Spain; Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada; Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.
| |
Collapse
|
10
|
Politano L. Is Cardiac Transplantation Still a Contraindication in Patients with Muscular Dystrophy-Related End-Stage Dilated Cardiomyopathy? A Systematic Review. Int J Mol Sci 2024; 25:5289. [PMID: 38791328 PMCID: PMC11121328 DOI: 10.3390/ijms25105289] [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: 03/08/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Inherited muscular diseases (MDs) are genetic degenerative disorders typically caused by mutations in a single gene that affect striated muscle and result in progressive weakness and wasting in affected individuals. Cardiac muscle can also be involved with some variability that depends on the genetic basis of the MD (Muscular Dystrophy) phenotype. Heart involvement can manifest with two main clinical pictures: left ventricular systolic dysfunction with evolution towards dilated cardiomyopathy and refractory heart failure, or the presence of conduction system defects and serious life-threatening ventricular arrhythmias. The two pictures can coexist. In these cases, heart transplantation (HTx) is considered the most appropriate option in patients who are not responders to the optimized standard therapeutic protocols. However, cardiac transplant is still considered a relative contraindication in patients with inherited muscle disorders and end-stage cardiomyopathies. High operative risk related to muscle impairment and potential graft involvement secondary to the underlying myopathy have been the two main reasons implicated in the generalized reluctance to consider cardiac transplant as a viable option. We report an overview of cardiac involvement in MDs and its possible association with the underlying molecular defect, as well as a systematic review of HTx outcomes in patients with MD-related end-stage dilated cardiomyopathy, published so far in the literature.
Collapse
Affiliation(s)
- Luisa Politano
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
11
|
Titova A, Nikolaev S, Bilyalov A, Filatov N, Brovkin S, Shestakov D, Khatkov I, Pismennaya E, Bondarev V, Antyuxina M, Shagimardanova E, Bodunova N, Gusev O. Extreme Tolerance of Extraocular Muscles to Diseases and Aging: Why and How? Int J Mol Sci 2024; 25:4985. [PMID: 38732204 PMCID: PMC11084950 DOI: 10.3390/ijms25094985] [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: 04/09/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
The extraocular muscles (EOMs) possess unique characteristics that set them apart from other skeletal muscles. These muscles, responsible for eye movements, exhibit remarkable resistance to various muscular dystrophies and aging, presenting a significant contrast to the vulnerability of skeletal muscles to these conditions. In this review, we delve into the cellular and molecular underpinnings of the distinct properties of EOMs. We explore their structural complexity, highlighting differences in fiber types, innervation patterns, and developmental origins. Notably, EOM fibers express a diverse array of myosin heavy-chain isoforms, retaining embryonic forms into adulthood. Moreover, their motor innervation is characterized by a high ratio of nerve fibers to muscle fibers and the presence of unique neuromuscular junctions. These features contribute to the specialized functions of EOMs, including rapid and precise eye movements. Understanding the mechanisms behind the resilience of EOMs to disease and aging may offer insights into potential therapeutic strategies for treating muscular dystrophies and myopathies affecting other skeletal muscles.
Collapse
Affiliation(s)
- Angelina Titova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Sergey Nikolaev
- SBHI Moscow Clinical Scientific Center Named after Loginov MHD, 111123 Moscow, Russia
| | - Airat Bilyalov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- SBHI Moscow Clinical Scientific Center Named after Loginov MHD, 111123 Moscow, Russia
| | - Nikita Filatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Sergei Brovkin
- SBHI Moscow Clinical Scientific Center Named after Loginov MHD, 111123 Moscow, Russia
| | | | - Igor Khatkov
- SBHI Moscow Clinical Scientific Center Named after Loginov MHD, 111123 Moscow, Russia
| | | | | | | | - Elena Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- SBHI Moscow Clinical Scientific Center Named after Loginov MHD, 111123 Moscow, Russia
| | - Natalia Bodunova
- SBHI Moscow Clinical Scientific Center Named after Loginov MHD, 111123 Moscow, Russia
| | - Oleg Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Life Improvement by Future Technologies (LIFT) Center, 121205 Moscow, Russia
| |
Collapse
|
12
|
Geist Hauserman J, Laverty CG, Donkervoort S, Hu Y, Silverstein S, Neuhaus SB, Saade D, Vaughn G, Malicki D, Kaur R, Li Y, Luo Y, Liu P, Burr P, Foley AR, Mohassel P, Bönnemann CG. Clinical, immunohistochemical, and genetic characterization of splice-altering biallelic DES variants: Therapeutic implications. HGG ADVANCES 2024; 5:100274. [PMID: 38358893 PMCID: PMC10876619 DOI: 10.1016/j.xhgg.2024.100274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
Pathogenic variants in the DES gene clinically manifest as progressive skeletal muscle weakness, cardiomyopathy with associated severe arrhythmias, and respiratory insufficiency, and are collectively known as desminopathies. While most DES pathogenic variants act via a dominant mechanism, recessively acting variants have also been reported. Currently, there are no effective therapeutic interventions for desminopathies of any type. Here, we report an affected individual with rapidly progressive dilated cardiomyopathy, requiring heart transplantation at age 13 years, in the setting of childhood-onset skeletal muscle weakness. We identified biallelic DES variants (c.640-13 T>A and c.1288+1 G>A) and show aberrant DES gene splicing in the affected individual's muscle. Through the generation of an inducible lentiviral system, we transdifferentiated fibroblast cultures derived from the affected individual into myoblasts and validated this system using RNA sequencing. We tested rationally designed, custom antisense oligonucleotides to screen for splice correction in these transdifferentiated cells and a functional minigene splicing assay. However, rather than correctly redirecting splicing, we found them to induce undesired exon skipping. Our results indicate that, while an individual precision-based molecular therapeutic approach to splice-altering pathogenic variants is promising, careful preclinical testing is imperative for each novel variant to test the feasibility of this type of approach for translation.
Collapse
Affiliation(s)
- Janelle Geist Hauserman
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA.
| | | | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Sarah Silverstein
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Sarah B Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Dimah Saade
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | | | | | - Rupleen Kaur
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Yuesheng Li
- DNA Sequencing and Genomics Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Yan Luo
- DNA Sequencing and Genomics Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Poching Liu
- DNA Sequencing and Genomics Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Patrick Burr
- DNA Sequencing and Genomics Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Payam Mohassel
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA.
| |
Collapse
|
13
|
Stål P, Nord H, von Hofsten J, Holmlund T, Shah F. Desmin gene expression is not ubiquitous in all upper airway myofibers and the pattern differs between healthy and sleep apnea subjects. Eur J Med Res 2024; 29:216. [PMID: 38566246 PMCID: PMC10988944 DOI: 10.1186/s40001-024-01812-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Desmin is a major cytoskeletal protein considered ubiquitous in mature muscle fibers. However, we earlier reported that a subgroup of muscle fibers in the soft palate of healthy subjects and obstructive sleep apnea patients (OSA) lacked immunoexpression for desmin. This raised the question of whether these fibers also lack messenger ribonucleic acid (mRNA) for desmin and can be considered a novel fiber phenotype. Moreover, some fibers in the OSA patients had an abnormal distribution and aggregates of desmin. Thus, the aim of the study was to investigate if these desmin protein abnormalities are also reflected in the expression of desmin mRNA in an upper airway muscle of healthy subjects and OSA patients. METHODS Muscle biopsies from the musculus uvulae in the soft palate were obtained from ten healthy male subjects and six male patients with OSA. Overnight sleep apnea registrations were done for all participants. Immunohistochemistry, in-situ hybridization, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) techniques were used to evaluate the presence of desmin protein and its mRNA. RESULTS Our findings demonstrated that a group of muscle fibers lacked expression for desmin mRNA and desmin protein in healthy individuals and OSA patients (12.0 ± 5.6% vs. 23.1 ± 10.8%, p = 0.03). A subpopulation of these fibers displayed a weak subsarcolemmal rim of desmin accompanied by a few scattered mRNA dots in the cytoplasm. The muscles of OSA patients also differed from healthy subjects by exhibiting muscle fibers with reorganized or accumulated aggregates of desmin protein (14.5 ± 6.5%). In these abnormal fibers, the density of mRNA was generally low or concentrated in specific regions. The overall quantification of desmin mRNA by RT-qPCR was significantly upregulated in OSA patients compared to healthy subjects (p = 0.01). CONCLUSIONS Our study shows evidence that muscle fibers in the human soft palate lack both mRNA and protein for desmin. This indicates a novel cytoskeletal structure and challenges the ubiquity of desmin in muscle fibers. Moreover, the observation of reorganized or accumulated aggregates of desmin mRNA and desmin protein in OSA patients suggests a disturbance in the transcription and translation process in the fibers of the patients.
Collapse
Affiliation(s)
- Per Stål
- Department of Medical and Translational Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Hanna Nord
- Department of Medical and Translational Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Medical and Translational Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Thorbjörn Holmlund
- Department of Clinical Sciences, Otorhinolaryngology, Umeå University, SE-901 87, Umeå, Sweden
| | - Farhan Shah
- Department of Medical and Translational Biology, Umeå University, SE-901 87, Umeå, Sweden.
| |
Collapse
|
14
|
Yoshioka N. Roles of dystonin isoforms in the maintenance of neural, muscle, and cutaneous tissues. Anat Sci Int 2024; 99:7-16. [PMID: 37603210 DOI: 10.1007/s12565-023-00739-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023]
Abstract
Dystonin (DST), also known as bullous pemphigoid antigen 1 (BPAG1), encodes cytoskeletal linker proteins belonging to the plakin family. The DST gene produces several isoforms, including DST-a, DST-b, and DST-e, which are expressed in neural, muscle, and cutaneous tissues, respectively. Pathogenic DST mutations cause hereditary sensory and autonomic neuropathy type 6 (HSAN-VI) and epidermolysis bullosa simplex (EBS); therefore, it is important to elucidate the roles of DST isoforms in multiple organs. Recently, we have used several Dst mutant mouse strains, in which the expression of Dst isoforms is disrupted in distinct patterns, to gain new insight into how DST functions in multiple tissues. This review provides an overview of the roles played by tissue-specific DST isoforms in neural, muscle, and cutaneous tissues.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.
| |
Collapse
|
15
|
Claeyssen C, Bulangalire N, Bastide B, Agbulut O, Cieniewski-Bernard C. Desmin and its molecular chaperone, the αB-crystallin: How post-translational modifications modulate their functions in heart and skeletal muscles? Biochimie 2024; 216:137-159. [PMID: 37827485 DOI: 10.1016/j.biochi.2023.10.002] [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: 04/28/2023] [Revised: 08/04/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
Maintenance of the highly organized striated muscle tissue requires a cell-wide dynamic network through protein-protein interactions providing an effective mechanochemical integrator of morphology and function. Through a continuous and complex trans-cytoplasmic network, desmin intermediate filaments ensure this essential role in heart and in skeletal muscle. Besides their role in the maintenance of cell shape and architecture (permitting contractile activity efficiency and conferring resistance towards mechanical stress), desmin intermediate filaments are also key actors of cell and tissue homeostasis. Desmin participates to several cellular processes such as differentiation, apoptosis, intracellular signalisation, mechanotransduction, vesicle trafficking, organelle biogenesis and/or positioning, calcium homeostasis, protein homeostasis, cell adhesion, metabolism and gene expression. Desmin intermediate filaments assembly requires αB-crystallin, a small heat shock protein. Over its chaperone activity, αB-crystallin is involved in several cellular functions such as cell integrity, cytoskeleton stabilization, apoptosis, autophagy, differentiation, mitochondria function or aggresome formation. Importantly, both proteins are known to be strongly associated to the aetiology of several cardiac and skeletal muscles pathologies related to desmin filaments disorganization and a strong disturbance of desmin interactome. Note that these key proteins of cytoskeleton architecture are extensively modified by post-translational modifications that could affect their functional properties. Therefore, we reviewed in the herein paper the impact of post-translational modifications on the modulation of cellular functions of desmin and its molecular chaperone, the αB-crystallin.
Collapse
Affiliation(s)
- Charlotte Claeyssen
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Nathan Bulangalire
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France; Université de Lille, CHU Lille, F-59000 Lille, France
| | - Bruno Bastide
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Caroline Cieniewski-Bernard
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France.
| |
Collapse
|
16
|
Palioura D, Mellidis K, Ioannidou-Kabouri K, Galatou E, Mouchtouri ET, Stamatiou R, Mavrommatis-Parasidis P, Panteris E, Varela A, Davos C, Drosatos K, Mavroidis M, Lazou A. PPARδ activation improves cardiac mitochondrial homeostasis in desmin deficient mice but does not alleviate systolic dysfunction. J Mol Cell Cardiol 2023; 183:27-41. [PMID: 37603971 DOI: 10.1016/j.yjmcc.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/22/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) δ is a major transcriptional regulator of cardiac energy metabolism with pleiotropic properties, including anti-inflammatory, anti-oxidative and cardioprotective action. In this study, we sought to investigate whether pharmacological activation of PPARδ via intraperitoneal administration of the selective ligand GW0742 could ameliorate heart failure and mitochondrial dysfunction that have been previously reported in a characterized genetic model of heart failure, the desmin null mice (Des-/-). Our studies demonstrate that treatment of Des-/- mice with the PPARδ agonist attenuated cardiac inflammation, fibrosis and cardiac remodeling. In addition, PPARδ activation alleviated oxidative stress in the failing myocardium as evidenced by decreased ROS levels. Importantly, PPARδ activation stimulated mitochondrial biogenesis, prevented mitochondrial and sarcoplasmic reticulum vacuolar degeneration and improved the mitochondrial intracellular distribution. Finally, PPARδ activation alleviated the mitochondrial respiratory dysfunction, prevented energy depletion and alleviated excessive autophagy and mitophagy in Des-/- hearts. Nevertheless, improvement of all these parameters did not suffice to overcome the significant structural deficiencies that desmin deletion incurs in cardiomyocytes and cardiac function did not improve significantly. In conclusion, pharmacological PPARδ activation in Des-/- hearts exerts protective effects during myocardial degeneration and heart failure by preserving the function and quality of the mitochondrial network. These findings implicate PPARδ agonists as a supplemental constituent of heart failure medications.
Collapse
Affiliation(s)
- Dimitra Palioura
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Kyriakos Mellidis
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Konstantina Ioannidou-Kabouri
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Eleftheria Galatou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | - Rodopi Stamatiou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | - Emmanuel Panteris
- Department of Botany, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Aimilia Varela
- Clinical, Experimental Surgery & Translational Research Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Constantinos Davos
- Clinical, Experimental Surgery & Translational Research Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece.
| |
Collapse
|
17
|
Antuofermo E, Orioles M, Murgia C, Burrai GP, Penati M, Gottardi C, Polinas M, Volpatti D, Galeotti M, Addis MF. Exploring Immunohistochemistry in Fish: Assessment of Antibody Reactivity by Western Immunoblotting. Animals (Basel) 2023; 13:2934. [PMID: 37760333 PMCID: PMC10525475 DOI: 10.3390/ani13182934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
In recent years, research on fish has seen remarkable advancements, especially in aquaculture, ornamental fish industry, and biomedical studies. Immunohistochemistry has become crucial in fish research, aiding in physiological and pathological investigations. However, the use of antibodies originally developed for mammals has raised concerns about their cross-reactivity and specificity in fish. This study systematically evaluated the reactivity of commonly used antibodies for diagnostic purposes, especially in fish pathology, including pan-cytokeratin, vimentin, S-100, glial fibrillary acidic protein, and desmin in the tissue of Sparus aurata, Dicentrarchus labrax, Oncorhynchus mykiss, and Carassius auratus. Western immunoblotting was employed to assess antibody specificity. The results revealed that the pan-cytokeratin and glial fibrillary acidic protein antibodies cross-react with all tested fish species, while S-100 demonstrated specific staining in sea bream, goldfish, and rainbow trout tissues. Conversely, vimentin and desmin antibodies displayed no reactivity. In conclusion, the anti-cytokeratin clone AE1/AE3 and the polyclonal rabbit anti-glial fibrillary acidic protein antibody, which are extensively used in mammals, were validated for fish immunohistochemical studies. Regrettably, D33 anti-desmin and V9 anti-vimentin clones are unsuitable for immunohistochemistry in the tested fish. These findings underscore the need for species-specific antibodies and proper validation for accurate immunohistochemistry analyses in fish research.
Collapse
Affiliation(s)
- Elisabetta Antuofermo
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (E.A.); (C.M.); (M.P.)
| | - Massimo Orioles
- Veterinary Pathology Unit, Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, 33100 Udine, Italy; (M.O.); (D.V.); (M.G.)
| | - Claudio Murgia
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (E.A.); (C.M.); (M.P.)
| | - Giovanni P. Burrai
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (E.A.); (C.M.); (M.P.)
| | - Martina Penati
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (M.P.); (C.G.); (M.F.A.)
| | - Chiara Gottardi
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (M.P.); (C.G.); (M.F.A.)
| | - Marta Polinas
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (E.A.); (C.M.); (M.P.)
| | - Donatella Volpatti
- Veterinary Pathology Unit, Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, 33100 Udine, Italy; (M.O.); (D.V.); (M.G.)
| | - Marco Galeotti
- Veterinary Pathology Unit, Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, 33100 Udine, Italy; (M.O.); (D.V.); (M.G.)
| | - Maria Filippa Addis
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (M.P.); (C.G.); (M.F.A.)
| |
Collapse
|
18
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
19
|
Skelton LA, Ramachandra Rao S, Allen RS, Motz CT, Pardue MT, Fliesler SJ. Retinal gliosis and phenotypic diversity of intermediate filament induction and remodeling upon acoustic blast overpressure (ABO) exposure to the rat eye. Exp Eye Res 2023; 234:109585. [PMID: 37481225 PMCID: PMC10730083 DOI: 10.1016/j.exer.2023.109585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/22/2023] [Accepted: 07/14/2023] [Indexed: 07/24/2023]
Abstract
Traumatic brain injury (TBI) caused by acoustic blast overpressure (ABO) is frequently associated with chronic visual deficits in military personnel and civilians. In this study, we characterized retinal gliotic response in adult male rats following a single ABO exposure directed to one side of the head. Expression of gliosis markers and intermediate filaments was assessed at 48 h and 1 wk post-ABO exposure, in comparison to age-matched non-exposed control retina. In response to a single ABO exposure, type III IF, glial fibrillary acidic protein (GFAP) was variably induced in a subpopulation of retinal Müller glia in ipsilateral eyes. ABO-exposed eyes exhibited radial Müller glial GFAP filament extension through the inner plexiform layer (IPL) and the inner nuclear layer (INL) through the retina in both the nasal quadrant and juxta-optic nerve head (jONH) eye regions at 1 wk post-ABO. We observed an ∼6-fold increase (p ≤ 0.05) in radial glial GFAP immunolabeling in the IPL in both eye regions, in comparison to regionally matched controls. Similarly, GFAP extension through the INL into the outer retina was elevated ∼3-fold, p ≤ 0.05 in the nasal retina, but exhibited wider variability in the jONH retina. In contrast, constitutive type III IF vimentin exhibited greater remodeling in retinal Müller glia through the jONH retina compared to the nasal retina in response to ABO. We observed areas of lateral vimentin remodeling through the Müller glial end-feet, and greater mid-outer retinal radial vimentin IF extension in a subpopulation of glia at 1 wk post-ABO. We also observed a significant increase in total retinal levels of the type III IF desmin in ABO-exposed retina vs. controls (∼1.6-fold, p ≤ 0.01). In addition, ABO-exposure elicited varied glial induction of developmentally regulated type VI family IFs (nestin and synemin) in subpopulations of Müller cells at 48 h and 1 wk post-ABO. We demonstrate that multiple glial phenotypes emerge in the rat retina following a single ABO exposure, rather than a global homogeneous retinal glial response, involving less well characterized IF protein forms which warrant further investigation in the context of ABO-induced retinal gliosis.
Collapse
Affiliation(s)
- Lara A Skelton
- Research Service, VA Western NY Healthcare System - Buffalo VAMC, Buffalo, NY, USA
| | - Sriganesh Ramachandra Rao
- Research Service, VA Western NY Healthcare System - Buffalo VAMC, Buffalo, NY, USA; Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System - Atlanta VAMC, Decatur, GA, USA
| | - Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System - Atlanta VAMC, Decatur, GA, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System - Atlanta VAMC, Decatur, GA, USA; Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Steven J Fliesler
- Research Service, VA Western NY Healthcare System - Buffalo VAMC, Buffalo, NY, USA; Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
20
|
Moneo-Corcuera D, Viedma-Poyatos Á, Stamatakis K, Pérez-Sala D. Desmin Reorganization by Stimuli Inducing Oxidative Stress and Electrophiles: Role of Its Single Cysteine Residue. Antioxidants (Basel) 2023; 12:1703. [PMID: 37760006 PMCID: PMC10525603 DOI: 10.3390/antiox12091703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/23/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
The type III intermediate filament proteins vimentin and GFAP are modulated by oxidants and electrophiles, mainly through perturbation of their single cysteine residues. Desmin, the type III intermediate filament protein specific to muscle cells, is critical for muscle homeostasis, playing a key role in sarcomere organization and mitochondrial function. Here, we have studied the impact of oxidants and cysteine-reactive agents on desmin behavior. Our results show that several reactive species and drugs induce covalent modifications of desmin in vitro, of which its single cysteine residue, C333, is an important target. Moreover, stimuli eliciting oxidative stress or lipoxidation, including H2O2, 15-deoxy-prostaglandin J2, and CoCl2-elicited chemical hypoxia, provoke desmin disorganization in H9c2 rat cardiomyoblasts transfected with wild-type desmin, which is partially attenuated in cells expressing a C333S mutant. Notably, in cells lacking other cytoplasmic intermediate filaments, network formation by desmin C333S appears less efficient than that of desmin wt, especially when these proteins are expressed as fluorescent fusion constructs. Nevertheless, in these cells, the desmin C333S organization is also protected from disruption by oxidants. Taken together, our results indicate that desmin is a target for oxidative and electrophilic stress, which elicit desmin remodeling conditioned by the presence of its single cysteine residue.
Collapse
Affiliation(s)
- Diego Moneo-Corcuera
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain; (D.M.-C.); (Á.V.-P.)
| | - Álvaro Viedma-Poyatos
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain; (D.M.-C.); (Á.V.-P.)
| | - Konstantinos Stamatakis
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain;
- Centro de Biología Molecular Severo Ochoa (UAM/CSIC), 28049 Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas (CSIC), 28040 Madrid, Spain; (D.M.-C.); (Á.V.-P.)
| |
Collapse
|
21
|
Sukhareva KS, Smolina NA, Churkina AI, Kalugina KK, Zhuk SV, Khudiakov AA, Khodot AA, Faggian G, Luciani GB, Sejersen T, Kostareva AA. Desmin mutations impact the autophagy flux in C2C12 cell in mutation-specific manner. Cell Tissue Res 2023; 393:357-375. [PMID: 37277577 PMCID: PMC10406715 DOI: 10.1007/s00441-023-03790-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 05/12/2023] [Indexed: 06/07/2023]
Abstract
Desmin is the main intermediate filament of striated and smooth muscle cells and plays a crucial role in maintaining the stability of muscle fiber during contraction and relaxation cycles. Being a component of Z-disk area, desmin integrates autophagic pathways, and the disturbance of Z-disk proteins' structure negatively affects chaperone-assisted selective autophagy (CASA). In the present study, we focused on alteration of autophagy flux in myoblasts expressing various Des mutations. We applied Western blotting, immunocytochemistry, RNA sequencing, and shRNA approach to demonstrate that DesS12F, DesA357P, DesL345P, DesL370P, and DesD399Y mutations. Mutation-specific effect on autophagy flux being most severe in aggregate-prone Des mutations such as DesL345P, DesL370P, and DesD399Y. RNA sequencing data confirmed the most prominent effect of these mutations on expression profile and, in particular, on autophagy-related genes. To verify CASA contribution to desmin aggregate formation, we suppressed CASA by knocking down Bag3 and demonstrated that it promoted aggregate formation and lead to downregulation of Vdac2 and Vps4a and upregulation of Lamp, Pink1, and Prkn. In conclusion, Des mutations showed a mutation-specific effect on autophagy flux in C2C12 cells with either a predominant impact on autophagosome maturation or on degradation and recycling processes. Aggregate-prone desmin mutations lead to the activation of basal autophagy level while suppressing the CASA pathway by knocking down Bag3 can promote desmin aggregate formation.
Collapse
Affiliation(s)
- K S Sukhareva
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia.
- Graduate School of Life and Health Science, University of Verona, Verona, Italy.
| | - N A Smolina
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - A I Churkina
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - K K Kalugina
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - S V Zhuk
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - A A Khudiakov
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - A A Khodot
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - G Faggian
- Graduate School of Life and Health Science, University of Verona, Verona, Italy
| | - G B Luciani
- Graduate School of Life and Health Science, University of Verona, Verona, Italy
| | - T Sejersen
- Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Neuropaediatrics, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - A A Kostareva
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Papadopoulos C, Malfatti E, Métay C, Keren B, Lejeune E, Buratti J, Xirou S, Chrysanthou-Piterou M, Papadimas GK. Deep Characterization of a Greek Patient with Desmin-Related Myofibrillar Myopathy and Cardiomyopathy. Int J Mol Sci 2023; 24:11181. [PMID: 37446359 DOI: 10.3390/ijms241311181] [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: 05/01/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Desmin is a class III intermediate filament protein highly expressed in cardiac, smooth and striated muscle. Autosomal dominant or recessive mutations in the desmin gene (DES) result in a variety of diseases, including cardiomyopathies and myofibrillar myopathy, collectively called desminopathies. Here we describe the clinical, histological and radiological features of a Greek patient with a myofibrillar myopathy and cardiomyopathy linked to the c.734A>G,p.(Glu245Gly) heterozygous variant in the DES gene. Moreover, through ribonucleic acid sequencing analysis in skeletal muscle we show that this variant provokes a defect in exon 3 splicing and thus should be considered clearly pathogenic.
Collapse
Affiliation(s)
- Constantinos Papadopoulos
- First Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, ERN, EURO NMD, 11528 Athens, Greece
| | - Edoardo Malfatti
- Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Université Paris Est, U955 INSERM, EnvA, EFS, IMRB, F-94010 and APHP, Henri Mondor Hospital, 94010 Créteil, France
| | - Corinne Métay
- APHP, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Centre de Génétique Moléculaire et Chromosomique, INSERM, Institut de Myologie, Groupe Hospitalier La Pitié-Salpêtrière-Charles Foix, Sorbonne Université, 75013 Paris, France
| | - Boris Keren
- APHP, Centre de Génétique Moléculaire et Chromosomique, UF Génétique du Développement, GH Pitié-Salpêtrière, 75013 Paris, France
| | - Elodie Lejeune
- APHP, Centre de Génétique Moléculaire et Chromosomique, UF Génétique du Développement, GH Pitié-Salpêtrière, 75013 Paris, France
| | - Julien Buratti
- APHP, Centre de Génétique Moléculaire et Chromosomique, UF Génétique du Développement, GH Pitié-Salpêtrière, 75013 Paris, France
| | - Sophia Xirou
- First Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, ERN, EURO NMD, 11528 Athens, Greece
| | - Margarita Chrysanthou-Piterou
- First Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, ERN, EURO NMD, 11528 Athens, Greece
| | - George K Papadimas
- First Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, ERN, EURO NMD, 11528 Athens, Greece
| |
Collapse
|
23
|
Rossi R, Torelli S, Ala P, Weston W, Morgan J, Malhotra J, Muntoni F. MyoD-induced reprogramming of human fibroblasts and urinary stem cells in vitro: protocols and their applications. Front Physiol 2023; 14:1145047. [PMID: 37265839 PMCID: PMC10229783 DOI: 10.3389/fphys.2023.1145047] [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: 01/15/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
The conversion of fibroblasts into myogenic cells is a powerful tool to both develop and test therapeutic strategies and to perform in-depth investigations of neuromuscular disorders, avoiding the need for muscle biopsies. We developed an easy, reproducible, and high-efficiency lentivirus-mediated transdifferentiation protocol, that can be used to convert healthy donor fibroblasts and a promising new cellular model, urinary stem cells (USCs), into myoblasts, that can be further differentiated into multinucleated myotubes in vitro. Transcriptome and proteome profiling of specific muscle markers (desmin, myosin, dystrophin) was performed to characterize both the myoblasts and myotubes derived from each cell type and to test the transdifferentiation-inducing capacity of MYOD1 in fibroblasts and USCs. Specifically, the Duchenne muscular dystrophy (DMD) transcripts and proteins, including both the full-length Dp427 and the short Dp71 isoform, were evaluated. The protocol was firstly developed in healthy donor fibroblasts and USCs and then used to convert DMD patients' fibroblasts, with the aim of testing the efficacy of an antisense drug in vitro. Technical issues, limitations, and problems are explained and discussed. We demonstrate that MyoD-induced-fibroblasts and USCs are a useful in vitro model of myogenic cells to investigate possible therapies for neuromuscular diseases.
Collapse
Affiliation(s)
- Rachele Rossi
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Silvia Torelli
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Pierpaolo Ala
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - William Weston
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Jennifer Morgan
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | | | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| |
Collapse
|
24
|
Zou Y, Shi H, Liu N, Wang H, Song X, Liu B. Mechanistic insights into heat shock protein 27, a potential therapeutic target for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1195464. [PMID: 37252119 PMCID: PMC10219228 DOI: 10.3389/fcvm.2023.1195464] [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: 03/28/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Heat shock protein 27 (HSP27) is a small chaperone protein that is overexpressed in a variety of cellular stress states. It is involved in regulating proteostasis and protecting cells from multiple sources of stress injury by stabilizing protein conformation and promoting the refolding of misfolded proteins. Previous studies have confirmed that HSP27 is involved in the development of cardiovascular diseases and plays an important regulatory role in this process. Herein, we comprehensively and systematically summarize the involvement of HSP27 and its phosphorylated form in pathophysiological processes, including oxidative stress, inflammatory responses, and apoptosis, and further explore the potential mechanisms and possible roles of HSP27 in the diagnosis and treatment of cardiovascular diseases. Targeting HSP27 is a promising future strategy for the treatment of cardiovascular diseases.
Collapse
|
25
|
Kakkat S, Pramanik P, Singh S, Singh AP, Sarkar C, Chakroborty D. Cardiovascular Complications in Patients with Prostate Cancer: Potential Molecular Connections. Int J Mol Sci 2023; 24:ijms24086984. [PMID: 37108147 PMCID: PMC10138415 DOI: 10.3390/ijms24086984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) and complications are often seen in patients with prostate cancer (PCa) and affect their clinical management. Despite acceptable safety profiles and patient compliance, androgen deprivation therapy (ADT), the mainstay of PCa treatment and chemotherapy, has increased cardiovascular risks and metabolic syndromes in patients. A growing body of evidence also suggests that patients with pre-existing cardiovascular conditions show an increased incidence of PCa and present with fatal forms of the disease. Therefore, it is possible that a molecular link exists between the two diseases, which has not yet been unraveled. This article provides insight into the connection between PCa and CVDs. In this context, we present our findings linking PCa progression with patients' cardiovascular health by performing a comprehensive gene expression study, gene set enrichment (GSEA) and biological pathway analysis using publicly available data extracted from patients with advanced metastatic PCa. We also discuss the common androgen deprivation strategies and CVDs most frequently reported in PCa patients and present evidence from various clinical trials that suggest that therapy induces CVD in PCa patients.
Collapse
Affiliation(s)
- Sooraj Kakkat
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Paramahansa Pramanik
- Department of Mathematics and Statistics, University of South Alabama, Mobile, AL 36688, USA
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Ajay Pratap Singh
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Chandrani Sarkar
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Debanjan Chakroborty
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
26
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Onore ME, Savarese M, Picillo E, Passamano L, Nigro V, Politano L. Bi-Allelic DES Gene Variants Causing Autosomal Recessive Myofibrillar Myopathies Affecting Both Skeletal Muscles and Cardiac Function. Int J Mol Sci 2022; 23:ijms232415906. [PMID: 36555543 PMCID: PMC9785402 DOI: 10.3390/ijms232415906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Mutations in the human desmin gene (DES) may cause both autosomal dominant and recessive cardiomyopathies leading to heart failure, arrhythmias and atrio-ventricular blocks, or progressive myopathies. Cardiac conduction disorders, arrhythmias and cardiomyopathies usually associated with progressive myopathy are the main manifestations of autosomal dominant desminopathies, due to mono-allelic pathogenic variants. The recessive forms, due to bi-allelic variants, are very rare and exhibit variable phenotypes in which premature sudden cardiac death could also occur in the first or second decade of life. We describe a further case of autosomal recessive desminopathy in an Italian boy born of consanguineous parents, who developed progressive myopathy at age 12, and dilated cardiomyopathy four years later and died of intractable heart failure at age 17. Next Generation Sequencing (NGS) analysis identified the homozygous loss-of-function variant c.634C>T; p.Arg212*, which was likely inherited from both parents. Furthermore, we performed a comparison of clinical and genetic results observed in our patient with those of cases so far reported in the literature.
Collapse
Affiliation(s)
- Maria Elena Onore
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Marco Savarese
- Folkhälsan Research Center, 00280 Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00280 Helsinki, Finland
| | - Esther Picillo
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Luigia Passamano
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Vincenzo Nigro
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
- Correspondence:
| |
Collapse
|
28
|
Desmin Knock-Out Cardiomyopathy: A Heart on the Verge of Metabolic Crisis. Int J Mol Sci 2022; 23:ijms231912020. [PMID: 36233322 PMCID: PMC9570457 DOI: 10.3390/ijms231912020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 12/05/2022] Open
Abstract
Desmin mutations cause familial and sporadic cardiomyopathies. In addition to perturbing the contractile apparatus, both desmin deficiency and mutated desmin negatively impact mitochondria. Impaired myocardial metabolism secondary to mitochondrial defects could conceivably exacerbate cardiac contractile dysfunction. We performed metabolic myocardial phenotyping in left ventricular cardiac muscle tissue in desmin knock-out mice. Our analyses revealed decreased mitochondrial number, ultrastructural mitochondrial defects, and impaired mitochondria-related metabolic pathways including fatty acid transport, activation, and catabolism. Glucose transporter 1 and hexokinase-1 expression and hexokinase activity were increased. While mitochondrial creatine kinase expression was reduced, fetal creatine kinase expression was increased. Proteomic analysis revealed reduced expression of proteins involved in electron transport mainly of complexes I and II, oxidative phosphorylation, citrate cycle, beta-oxidation including auxiliary pathways, amino acid catabolism, and redox reactions and oxidative stress. Thus, desmin deficiency elicits a secondary cardiac mitochondriopathy with severely impaired oxidative phosphorylation and fatty and amino acid metabolism. Increased glucose utilization and fetal creatine kinase upregulation likely portray attempts to maintain myocardial energy supply. It may be prudent to avoid medications worsening mitochondrial function and other metabolic stressors. Therapeutic interventions for mitochondriopathies might also improve the metabolic condition in desmin deficient hearts.
Collapse
|
29
|
Skeletal Muscle Dysfunction in Experimental Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms231810912. [PMID: 36142826 PMCID: PMC9501428 DOI: 10.3390/ijms231810912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a serious, progressive, and often fatal disease that is in urgent need of improved therapies that treat it. One of the remaining therapeutic challenges is the increasingly recognized skeletal muscle dysfunction that interferes with exercise tolerance. Here we report that in the adult rat Sugen/hypoxia (SU/Hx) model of severe pulmonary hypertension (PH), there is highly significant, almost 50%, decrease in exercise endurance, and this is associated with a 25% increase in the abundance of type II muscle fiber markers, thick sarcomeric aggregates and an increase in the levels of FoxO1 in the soleus (a predominantly type I fiber muscle), with additional alterations in the transcriptomic profiles of the diaphragm (a mixed fiber muscle) and the extensor digitorum longus (a predominantly Type II fiber muscle). In addition, soleus atrophy may contribute to impaired exercise endurance. Studies in L6 rat myoblasts have showed that myotube differentiation is associated with increased FoxO1 levels and type II fiber markers, while the inhibition of FoxO1 leads to increased type I fiber markers. We conclude that the formation of aggregates and a FoxO1-mediated shift in the skeletal muscle fiber-type specification may underlie skeletal muscle dysfunction in an experimental study of PH.
Collapse
|
30
|
Su W, van Wijk SW, Brundel BJJM. Desmin variants: Trigger for cardiac arrhythmias? Front Cell Dev Biol 2022; 10:986718. [PMID: 36158202 PMCID: PMC9500482 DOI: 10.3389/fcell.2022.986718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Desmin (DES) is a classical type III intermediate filament protein encoded by the DES gene. Desmin is abundantly expressed in cardiac, skeletal, and smooth muscle cells. In these cells, desmin interconnects several protein-protein complexes that cover cell-cell contact, intracellular organelles such as mitochondria and the nucleus, and the cytoskeletal network. The extra- and intracellular localization of the desmin network reveals its crucial role in maintaining the structural and mechanical integrity of cells. In the heart, desmin is present in specific structures of the cardiac conduction system including the sinoatrial node, atrioventricular node, and His-Purkinje system. Genetic variations and loss of desmin drive a variety of conditions, so-called desminopathies, which include desmin-related cardiomyopathy, conduction system-related atrial and ventricular arrhythmias, and sudden cardiac death. The severe cardiac disease outcomes emphasize the clinical need to understand the molecular and cellular role of desmin driving desminopathies. As the role of desmin in cardiomyopathies has been discussed thoroughly, the current review is focused on the role of desmin impairment as a trigger for cardiac arrhythmias. Here, the molecular and cellular mechanisms of desmin to underlie a healthy cardiac conduction system and how impaired desmin triggers cardiac arrhythmias, including atrial fibrillation, are discussed. Furthermore, an overview of available (genetic) desmin model systems for experimental cardiac arrhythmia studies is provided. Finally, potential implications for future clinical treatments of cardiac arrhythmias directed at desmin are highlighted.
Collapse
Affiliation(s)
- Wei Su
- Physiology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Stan W. van Wijk
- Physiology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bianca J. J. M. Brundel
- Physiology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Bianca J. J. M. Brundel,
| |
Collapse
|
31
|
Soglia F, Bordini M, Mazzoni M, Zappaterra M, Di Nunzio M, Clavenzani P, Davoli R, Meluzzi A, Sirri F, Petracci M. The evolution of vimentin and desmin in Pectoralis major muscles of broiler chickens supports their essential role in muscle regeneration. Front Physiol 2022; 13:970034. [PMID: 36134328 PMCID: PMC9483144 DOI: 10.3389/fphys.2022.970034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Vimentin (VIM) and desmin (DES) are muscle-specific proteins having crucial roles in maintaining the lateral organization and alignment of the sarcomeric structure during myofibrils’ regeneration. The present experiment was designed to ascertain the evolution of VIM and DES in Pectoralis major muscles (PM) of fast-growing (FG) and medium-growing (MG) meat-type chickens both at the protein and gene levels. MG broilers were considered as a control group whereas the evolution of VIM and DES over the growth period was evaluated in FG by collecting samples at different developmental stages (7, 14, 21, 28, 35, and 42 days). After performing a preliminary classification of the samples based on their histological features, 5 PM/sampling time/genotype were selected for western blot, immunohistochemistry (IHC), and gene expression analyses. Overall, the findings obtained at the protein level mirrored those related to their encoding genes, although a potential time lag required to observe the consequences of gene expression was evident. The two- and 3-fold higher level of the VIM-based heterodimer observed in FG at d 21 and d 28 in comparison with MG of the same age might be ascribed to the beginning and progressive development of the regenerative processes. This hypothesis is supported by IHC highlighting the presence of fibers to co-expressing VIM and DES. In addition, gene expression analyses suggested that, unlike VIM common sequence, VIM long isoform may not be directly implicated in muscle regeneration. As for DES content, the fluctuating trends observed for both the native protein and its heterodimer in FG might be ascribed to its importance for maintaining the structural organization of the regenerating fibers. Furthermore, the higher expression level of the DES gene in FG in comparison with MG further supported its potential application as a marker of muscle fibers’ regeneration. In conclusion, the findings of the present research seem to support the existence of a relationship between the occurrence of muscle regeneration and the growth rate of meat-type chickens and corroborate the potential use of VIM and DES as molecular markers of these cellular processes.
Collapse
Affiliation(s)
- Francesca Soglia
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Martina Bordini
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Maurizio Mazzoni
- Department of Veterinary Medical Sciences, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Martina Zappaterra
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
- *Correspondence: Martina Zappaterra,
| | - Mattia Di Nunzio
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Paolo Clavenzani
- Department of Veterinary Medical Sciences, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Roberta Davoli
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Adele Meluzzi
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Federico Sirri
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Massimiliano Petracci
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum–University of Bologna, Bologna, Italy
| |
Collapse
|
32
|
Yoshioka N, Kurose M, Yano M, Tran DM, Okuda S, Mori-Ochiai Y, Horie M, Nagai T, Nishino I, Shibata S, Takebayashi H. Isoform-specific mutation in Dystonin-b gene causes late-onset protein aggregate myopathy and cardiomyopathy. eLife 2022; 11:78419. [PMID: 35942699 PMCID: PMC9365387 DOI: 10.7554/elife.78419] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
Dystonin (DST), which encodes cytoskeletal linker proteins, expresses three tissue-selective isoforms: neural DST-a, muscular DST-b, and epithelial DST-e. DST mutations cause different disorders, including hereditary sensory and autonomic neuropathy 6 (HSAN-VI) and epidermolysis bullosa simplex; however, etiology of the muscle phenotype in DST-related diseases has been unclear. Because DST-b contains all of the DST-a-encoding exons, known HSAN-VI mutations could affect both DST-a and DST-b isoforms. To investigate the specific function of DST-b in striated muscles, we generated a Dst-b-specific mutant mouse model harboring a nonsense mutation. Dst-b mutant mice exhibited late-onset protein aggregate myopathy and cardiomyopathy without neuropathy. We observed desmin aggregation, focal myofibrillar dissolution, and mitochondrial accumulation in striated muscles, which are common characteristics of myofibrillar myopathy. We also found nuclear inclusions containing p62, ubiquitin, and SUMO proteins with nuclear envelope invaginations as a unique pathological hallmark in Dst-b mutation-induced cardiomyopathy. RNA-sequencing analysis revealed changes in expression of genes responsible for cardiovascular functions. In silico analysis identified DST-b alleles with nonsense mutations in populations worldwide, suggesting that some unidentified hereditary myopathy and cardiomyopathy are caused by DST-b mutations. Here, we demonstrate that the Dst-b isoform is essential for long-term maintenance of striated muscles.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Transdisciplinary Research Programs, Niigata University, Niigata, Japan
| | - Masayuki Kurose
- Department of Physiology, School of Dentistry, Iwate Medical University, Iwate, Japan
| | - Masato Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Dang Minh Tran
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shujiro Okuda
- Medical AI Center, School of Medicine, Niigata University, Niigata, Japan
| | - Yukiko Mori-Ochiai
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masao Horie
- Department of Nursing, Niigata College of Nursing, Jōetsu, Japan
| | - Toshihiro Nagai
- Electron Microscope Laboratory, Keio University, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University, Tokyo, Japan.,Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Center for Coordination of Research Facilities, Niigata University, Niigata, Japan
| |
Collapse
|
33
|
Silva AMS, Rodrigo P, Moreno CAM, Mendonça RDH, Estephan EDP, Camelo CG, Campos ED, Dias AT, Nascimento AM, Kulikowski LD, Oliveira ASB, Reed UC, Goldfarb LG, Olivé M, Zanoteli E. The Location of Disease-Causing DES Variants Determines the Severity of Phenotype and the Morphology of Sarcoplasmic Aggregates. J Neuropathol Exp Neurol 2022; 81:746-757. [PMID: 35898174 DOI: 10.1093/jnen/nlac063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Desmin (DES) is the main intermediate muscle filament that connects myofibrils individually and with the nucleus, sarcolemma, and organelles. Pathogenic variants of DES cause desminopathy, a disorder affecting the heart and skeletal muscles. We aimed to analyze the clinical features, morphology, and distribution of desmin aggregates in skeletal muscle biopsies of patients with desminopathy and to correlate these findings with the type and location of disease-causing DES variants. This retrospective study included 30 patients from 20 families with molecularly confirmed desminopathy from 2 neuromuscular referral centers. We identified 2 distinct patterns of desmin aggregates: well-demarcated subsarcolemmal aggregates and diffuse aggregates with poorly delimited borders. Pathogenic variants located in the 1B segment and the tail domain of the desmin molecule are more likely to present with early-onset cardiomyopathy compared to patients with variants in other segments. All patients with mutations in the 1B segment had well-demarcated subsarcolemmal aggregates, but none of the patients with variants in other desmin segments showed such histological features. We suggest that variants located in the 1B segment lead to well-shaped subsarcolemmal desmin aggregation and cause disease with more frequent cardiac manifestations. These findings will facilitate early identification of patients with potentially severe cardiac syndromes.
Collapse
Affiliation(s)
| | - Patricia Rodrigo
- Neuropathology Unit, Department of Pathology and Neuromuscular Unit, Department of Neurology, IDIBELL-Hospital Universitari de Bellvitge, Barcelona, Spain
| | | | | | - Eduardo de Paula Estephan
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Clara Gontijo Camelo
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Eliene Dutra Campos
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Alexandre Torchio Dias
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Amom Mendes Nascimento
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | | | - Acary Souza Bulle Oliveira
- Division of Neuromuscular Diseases, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Umbertina Conti Reed
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Lev G Goldfarb
- Department of Pathology and Molecular Medicine, Queen's University, Kingston General Hospital, Kingston, Ontario, Canada
| | - Montse Olivé
- Neuropathology Unit, Department of Pathology and Neuromuscular Unit, Department of Neurology, IDIBELL-Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
34
|
Shah F, Stål P. Myopathy of the upper airway in snoring and obstructive sleep apnea. Laryngoscope Investig Otolaryngol 2022; 7:636-645. [PMID: 35434344 PMCID: PMC9008167 DOI: 10.1002/lio2.782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 11/19/2022] Open
Abstract
Objective Previous reports of muscle changes in the upper airways of obstructive sleep apnea (OSA) patients have primarily been attributed to acquired nerve lesions due to snoring vibrations. The aim of this study was to investigate whether alterations reflecting muscle fiber injuries also occur in the upper respiratory tract of snoring and OSA patients and if these changes relate to upper airway dysfunction. Methods Muscle changes in biopsies from the soft palate of 20 patients suffering from snoring and OSA were investigated with enzyme, immunohistochemical, and morphometric techniques. Biopsies from eight healthy non‐snoring subjects were used as controls. Swallowing dysfunction was assessed with videoradiography. Results Fourteen patients had various degrees of swallowing dysfunction. The muscle samples from all the patients showed changes typical for both motor‐nerve lesions and muscle fiber injuries. The most common alterations reflecting myopathy were fibers having aggregates and disorganization of cytoskeletal proteins (15.5 ± 10.7%). Other changes were fibers with vacuole‐like structures (5.0 ± 4.4%), centrally positioned myonuclei (7.9 ± 4.8%), subsarcolemmal accumulations of nuclei, and various forms and sizes of ring fibers, that is, fibers where the myofilaments were disorganized peripherally (2.8 ± 2.8%). Conclusion The results show that muscle changes mirroring both myopathy and neuropathy co‐exist in the upper airway of snoring OSA patients. These findings suggest muscle weakness as a contributing factor to the upper airway dysfunction in OSA patients.
Collapse
Affiliation(s)
- Farhan Shah
- Laboratory of Muscle Biology, Department of Integrative Medical Biology Umeå University Umeå Sweden
| | - Per Stål
- Laboratory of Muscle Biology, Department of Integrative Medical Biology Umeå University Umeå Sweden
| |
Collapse
|
35
|
Gomes G, Seixas MR, Azevedo S, Audi K, Jurberg AD, Mermelstein C, Costa ML. What does desmin do: A bibliometric assessment of the functions of the muscle intermediate filament. Exp Biol Med (Maywood) 2022; 247:538-550. [PMID: 35130760 DOI: 10.1177/15353702221075035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Intermediate filaments were first described in muscle in 1968, and desmin was biochemically identified about 10 years afterwards. Its importance grew after the identification of desminopathies and desmin mutations that cause mostly cardiopathies. Since its characterization until recently, different functions have been attributed to desmin. Here, we use bibliometric tools to evaluate the articles published about desmin and to assess its several putative functions. We identified the most productive authors and the relationships between research groups. We studied the more frequent words among 9734 articles (September 2021) containing "desmin" on the title and abstract, to identify the major research focus. We generated an interactive spreadsheet with the 934 papers that contain "desmin" only on the title that can be used to search and quantify terms in the abstract. We further selected the articles that contained the terms "function" or "role" from the spreadsheet, which we then classified according to type of function, organelle, or tissue involved. Based on the bibliographic analysis, we assess comparatively the putative functions, and we propose an alternative explanation for the desmin function.
Collapse
Affiliation(s)
- Geyse Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
| | - Marianna R Seixas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
| | - Sarah Azevedo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
| | - Karina Audi
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
| | - Arnon D Jurberg
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil.,Faculdade de Medicina, Universidade Estácio de Sá, Rio de Janeiro 20071-001, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
| | - Manoel Luis Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21944-970, Brazil
| |
Collapse
|
36
|
|
37
|
Spörrer M, Kah D, Gerum RC, Reischl B, Huraskin D, Dessalles CA, Schneider W, Goldmann WH, Herrmann H, Thievessen I, Clemen CS, Friedrich O, Hashemolhosseini S, Schröder R, Fabry B. The desmin mutation R349P increases contractility and fragility of stem cell-generated muscle micro-tissues. Neuropathol Appl Neurobiol 2021; 48:e12784. [PMID: 34850968 DOI: 10.1111/nan.12784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/18/2021] [Accepted: 11/16/2021] [Indexed: 12/01/2022]
Abstract
AIMS Desminopathies comprise hereditary myopathies and cardiomyopathies caused by mutations in the intermediate filament protein desmin that lead to severe and often lethal degeneration of striated muscle tissue. Animal and single cell studies hinted that this degeneration process is associated with massive ultrastructural defects correlating with increased susceptibility of the muscle to acute mechanical stress. The underlying mechanism of mechanical susceptibility, and how muscle degeneration develops over time, however, has remained elusive. METHODS Here, we investigated the effect of a desmin mutation on the formation, differentiation, and contractile function of in vitro-engineered three-dimensional micro-tissues grown from muscle stem cells (satellite cells) isolated from heterozygous R349P desmin knock-in mice. RESULTS Micro-tissues grown from desmin-mutated cells exhibited spontaneous unsynchronised contractions, higher contractile forces in response to electrical stimulation, and faster force recovery compared with tissues grown from wild-type cells. Within 1 week of culture, the majority of R349P desmin-mutated tissues disintegrated, whereas wild-type tissues remained intact over at least three weeks. Moreover, under tetanic stimulation lasting less than 5 s, desmin-mutated tissues partially or completely ruptured, whereas wild-type tissues did not display signs of damage. CONCLUSIONS Our results demonstrate that the progressive degeneration of desmin-mutated micro-tissues is closely linked to extracellular matrix fibre breakage associated with increased contractile forces and unevenly distributed tensile stress. This suggests that the age-related degeneration of skeletal and cardiac muscle in patients suffering from desminopathies may be similarly exacerbated by mechanical damage from high-intensity muscle contractions. We conclude that micro-tissues may provide a valuable tool for studying the organization of myocytes and the pathogenic mechanisms of myopathies.
Collapse
Affiliation(s)
- Marina Spörrer
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Delf Kah
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Richard C Gerum
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Barbara Reischl
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, FAU, Erlangen, Germany
| | - Danyil Huraskin
- Institute of Biochemistry, Medical Faculty, FAU, Erlangen, Germany
| | - Claire A Dessalles
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany.,LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Werner Schneider
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Wolfgang H Goldmann
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany
| | - Harald Herrmann
- Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany.,Institute of Neuropathology, University Hospital Erlangen, FAU, Erlangen, Germany
| | - Ingo Thievessen
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany.,Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, FAU, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany
| | - Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, FAU, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany
| | - Rolf Schröder
- Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany.,Institute of Neuropathology, University Hospital Erlangen, FAU, Erlangen, Germany
| | - Ben Fabry
- Biophysics Group, Department of Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), FAU, Erlangen, Germany
| |
Collapse
|
38
|
Mocciaro E, Runfola V, Ghezzi P, Pannese M, Gabellini D. DUX4 Role in Normal Physiology and in FSHD Muscular Dystrophy. Cells 2021; 10:3322. [PMID: 34943834 PMCID: PMC8699294 DOI: 10.3390/cells10123322] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/10/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
In the last decade, the sequence-specific transcription factor double homeobox 4 (DUX4) has gone from being an obscure entity to being a key factor in important physiological and pathological processes. We now know that expression of DUX4 is highly regulated and restricted to the early steps of embryonic development, where DUX4 is involved in transcriptional activation of the zygotic genome. While DUX4 is epigenetically silenced in most somatic tissues of healthy humans, its aberrant reactivation is associated with several diseases, including cancer, viral infection and facioscapulohumeral muscular dystrophy (FSHD). DUX4 is also translocated, giving rise to chimeric oncogenic proteins at the basis of sarcoma and leukemia forms. Hence, understanding how DUX4 is regulated and performs its activity could provide relevant information, not only to further our knowledge of human embryonic development regulation, but also to develop therapeutic approaches for the diseases associated with DUX4. Here, we summarize current knowledge on the cellular and molecular processes regulated by DUX4 with a special emphasis on FSHD muscular dystrophy.
Collapse
Affiliation(s)
| | | | | | | | - Davide Gabellini
- Gene Expression and Muscular Dystrophy Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milano, Italy; (E.M.); (V.R.); (P.G.); (M.P.)
| |
Collapse
|
39
|
Brodehl A, Hain C, Flottmann F, Ratnavadivel S, Gaertner A, Klauke B, Kalinowski J, Körperich H, Gummert J, Paluszkiewicz L, Deutsch MA, Milting H. The Desmin Mutation DES-c.735G>C Causes Severe Restrictive Cardiomyopathy by Inducing In-Frame Skipping of Exon-3. Biomedicines 2021; 9:biomedicines9101400. [PMID: 34680517 PMCID: PMC8533191 DOI: 10.3390/biomedicines9101400] [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] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 02/02/2023] Open
Abstract
Currently, little is known about the genetic background of restrictive cardiomyopathy (RCM). Herein, we screened an index patient with RCM in combination with atrial fibrillation using a next generation sequencing (NGS) approach and identified the heterozygous mutation DES-c.735G>C. As DES-c.735G>C affects the last base pair of exon-3, it is unknown whether putative missense or splice site mutations are caused. Therefore, we applied nanopore amplicon sequencing revealing the expression of a transcript without exon-3 in the explanted myocardial tissue of the index patient. Western blot analysis verified this finding at the protein level. In addition, we performed cell culture experiments revealing an abnormal cytoplasmic aggregation of the truncated desmin form (p.D214-E245del) but not of the missense variant (p.E245D). In conclusion, we show that DES-c.735G>C causes a splicing defect leading to exon-3 skipping of the DES gene. DES-c.735G>C can be classified as a pathogenic mutation associated with RCM and atrial fibrillation. In the future, this finding might have relevance for the genetic understanding of similar cases.
Collapse
Affiliation(s)
- Andreas Brodehl
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Correspondence: (A.B.); (H.M.); Tel.: +49-(0)5731-973530 (A.B.); +49-(0)5731-973510 (H.M.)
| | - Carsten Hain
- Microbial Genomics and Biotechnology, Center for Biotechnology, Bielefeld University, D-33615 Bielefeld, Germany; (C.H.); (J.K.)
| | - Franziska Flottmann
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
| | - Sandra Ratnavadivel
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
| | - Anna Gaertner
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
| | - Bärbel Klauke
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Clinic for General and Interventional Cardiology/Angiology, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany
| | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, Center for Biotechnology, Bielefeld University, D-33615 Bielefeld, Germany; (C.H.); (J.K.)
| | - Hermann Körperich
- Heart and Diabetes Center NRW, Institute for Radiology, Nuclear Medicine and Molecular Imaging, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany;
| | - Jan Gummert
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Heart and Diabetes Center NRW, Department of Thoracic and Cardiovascular Surgery, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (L.P.); (M.-A.D.)
| | - Lech Paluszkiewicz
- Heart and Diabetes Center NRW, Department of Thoracic and Cardiovascular Surgery, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (L.P.); (M.-A.D.)
| | - Marcus-André Deutsch
- Heart and Diabetes Center NRW, Department of Thoracic and Cardiovascular Surgery, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (L.P.); (M.-A.D.)
| | - Hendrik Milting
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Correspondence: (A.B.); (H.M.); Tel.: +49-(0)5731-973530 (A.B.); +49-(0)5731-973510 (H.M.)
| |
Collapse
|
40
|
Chanpong A, Borrelli O, Thapar N. Hirschsprung disease and Paediatric Intestinal Pseudo-obstruction. Best Pract Res Clin Gastroenterol 2021; 56-57:101765. [PMID: 35331399 DOI: 10.1016/j.bpg.2021.101765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/31/2023]
Abstract
Hirschsprung disease (HSCR) and Paediatric Intestinal Pseudo-obstruction (PIPO) comprise two of the most recognized and severe disorders of gastrointestinal (GI) motility. HSCR is a developmental disorder of the enteric nervous system invariably affecting the large intestine, whereas the majority of PIPO conditions represent congenital disorders of one or more components of the neuromusculature and more diffusely affect the GI tract. Histopathology is deemed the gold standard for the diagnosis of HSCR and, arguably, of PIPO, but, other diagnostic modalities such as manometric and genetic studies have seen recent advances that may increase their utility. Especially for PIPO, management is multidisciplinary and best performed in specialist referral centres. Surgery remains the only viable treatment for HSCR and appears essential to optimize and sustain feeding and viability of intestinal function in PIPO patients. Novel therapies such as neural stem cell transplants show promise for the future.
Collapse
Affiliation(s)
- Atchariya Chanpong
- Neurogastroenterology & Motility Unit, Gastroenterology Department, Great Ormond Street Hospital for Children, London, WC1N 3JH, United Kingdom; Division of Gastroenterology and Hepatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand; Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Osvaldo Borrelli
- Neurogastroenterology & Motility Unit, Gastroenterology Department, Great Ormond Street Hospital for Children, London, WC1N 3JH, United Kingdom
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, United Kingdom; Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Queensland, 4101, Australia.
| |
Collapse
|
41
|
Ghasemizadeh A, Christin E, Guiraud A, Couturier N, Abitbol M, Risson V, Girard E, Jagla C, Soler C, Laddada L, Sanchez C, Jaque-Fernandez FI, Jacquemond V, Thomas JL, Lanfranchi M, Courchet J, Gondin J, Schaeffer L, Gache V. MACF1 controls skeletal muscle function through the microtubule-dependent localization of extra-synaptic myonuclei and mitochondria biogenesis. eLife 2021; 10:e70490. [PMID: 34448452 PMCID: PMC8500715 DOI: 10.7554/elife.70490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/10/2021] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.
Collapse
Affiliation(s)
- Alireza Ghasemizadeh
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Emilie Christin
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Alexandre Guiraud
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Nathalie Couturier
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Marie Abitbol
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
- Université Marcy l’Etoile, VetAgro SupLyonFrance
| | - Valerie Risson
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Emmanuelle Girard
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Christophe Jagla
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Cedric Soler
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Lilia Laddada
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Colline Sanchez
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Francisco-Ignacio Jaque-Fernandez
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Vincent Jacquemond
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Jean-Luc Thomas
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Marine Lanfranchi
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Julien Courchet
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Julien Gondin
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Laurent Schaeffer
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Vincent Gache
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| |
Collapse
|
42
|
Langer HT, Mossakowski AA, Avey AM, Wohlgemuth RP, Smith LR, Zbinden-Foncea H, Baar K. A mutation in desmin makes skeletal muscle less vulnerable to acute muscle damage after eccentric loading in rats. FASEB J 2021; 35:e21860. [PMID: 34411340 PMCID: PMC9292853 DOI: 10.1096/fj.202100711rr] [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: 04/27/2021] [Revised: 07/15/2021] [Accepted: 08/02/2021] [Indexed: 01/23/2023]
Abstract
Desminopathy is the most common intermediate filament disease in humans. The most frequent mutation causing desminopathy in patients is a R350P DES missense mutation. We have developed a rat model with an analogous mutation in R349P Des. To investigate the role of R349P Des in mechanical loading, we stimulated the sciatic nerve of wild‐type littermates (WT) (n = 6) and animals carrying the mutation (MUT) (n = 6) causing a lengthening contraction of the dorsi flexor muscles. MUT animals showed signs of ongoing regeneration at baseline as indicated by a higher number of central nuclei (genotype: P < .0001). While stimulation did not impact central nuclei, we found an increased number of IgG positive fibers (membrane damage indicator) after eccentric contractions with both genotypes (stimulation: P < .01). Interestingly, WT animals displayed a more pronounced increase in IgG positive fibers with stimulation compared to MUT (interaction: P < .05). In addition to altered histology, molecular signaling on the protein level differed between WT and MUT. The membrane repair protein dysferlin decreased with eccentric loading in WT but increased in MUT (interaction: P < .05). The autophagic substrate p62 was increased in both genotypes with loading (stimulation: P < .05) but tended to be more elevated in WT (interaction: P = .05). Caspase 3 levels, a central regulator of apoptotic cell death, was increased with stimulation in both genotypes (stimulation: P < .01) but more so in WT animals (interaction: P < .0001). Overall, our data indicate that R349P Des rats have a lower susceptibility to structural muscle damage of the cytoskeleton and sarcolemma with acute eccentric loading.
Collapse
Affiliation(s)
- Henning T Langer
- Functional Molecular Biology Laboratory, Department of Physiology and Membrane Biology, University of California, Davis, California, USA
| | - Agata A Mossakowski
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alec M Avey
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Ross P Wohlgemuth
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Lucas R Smith
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Herman Zbinden-Foncea
- Exercise Physiology Laboratory, School of Kinesiology, Universidad Finis Terrae, Santiago, Chile
| | - Keith Baar
- Functional Molecular Biology Laboratory, Department of Physiology and Membrane Biology, University of California, Davis, California, USA.,Neurobiology, Physiology and Behavior, University of California, Davis, California, USA.,VA Northern California Health Care System, Mather, California, USA
| |
Collapse
|
43
|
Yapa Abeywardana M, Samarasinghe KTG, Munkanatta Godage D, Ahn YH. Identification and Quantification of Glutathionylated Cysteines under Ischemic Stress. J Proteome Res 2021; 20:4529-4542. [PMID: 34382403 DOI: 10.1021/acs.jproteome.1c00473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemia reperfusion injury contributes to adverse cardiovascular diseases in part by producing a burst of reactive oxygen species that induce oxidations of many muscular proteins. Glutathionylation is one of the major protein cysteine oxidations that often serve as molecular mechanisms behind the pathophysiology associated with ischemic stress. Despite the biological significance of glutathionylation in ischemia reperfusion, identification of specific glutathionylated cysteines under ischemic stress has been limited. In this report, we have analyzed glutathionylation under oxygen-glucose deprivation (OGD) or repletion of nutrients after OGD (OGD/R) by using a clickable glutathione approach that specifically detects glutathionylated proteins. Our data find that palmitate availability induces a global level of glutathionylation and decreases cell viability during OGD/R. We have then applied a clickable glutathione-based proteomic quantification strategy, which enabled the identification and quantification of 249 glutathionylated cysteines in response to palmitate during OGD/R in the HL-1 cardiomyocyte cell line. The subsequent bioinformatic analysis found 18 glutathionylated cysteines whose genetic variants are associated with muscular disorders. Overall, our data report glutathionylated cysteines under ischemic stress that may contribute to adverse outcomes or muscular disorders.
Collapse
Affiliation(s)
| | | | | | - Young-Hoon Ahn
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
44
|
Bend, Push, Stretch: Remarkable Structure and Mechanics of Single Intermediate Filaments and Meshworks. Cells 2021; 10:cells10081960. [PMID: 34440729 PMCID: PMC8394331 DOI: 10.3390/cells10081960] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
The cytoskeleton of the eukaryotic cell provides a structural and functional scaffold enabling biochemical and cellular functions. While actin and microtubules form the main framework of the cell, intermediate filament networks provide unique mechanical properties that increase the resilience of both the cytoplasm and the nucleus, thereby maintaining cellular function while under mechanical pressure. Intermediate filaments (IFs) are imperative to a plethora of regulatory and signaling functions in mechanotransduction. Mutations in all types of IF proteins are known to affect the architectural integrity and function of cellular processes, leading to debilitating diseases. The basic building block of all IFs are elongated α-helical coiled-coils that assemble hierarchically into complex meshworks. A remarkable mechanical feature of IFs is the capability of coiled-coils to metamorphize into β-sheets under stress, making them one of the strongest and most resilient mechanical entities in nature. Here, we discuss structural and mechanical aspects of IFs with a focus on nuclear lamins and vimentin.
Collapse
|
45
|
Abstract
Assembly of intermediate filaments (IFs) is reliant upon amino-terminal head domains. These head domains are of low sequence complexity and are assumed to function in the absence of structural order. Herein, we provide evidence that the head domains of the desmin and neurofilament light (NFL) IF proteins self-associate via the formation of labile but structurally specific cross-β interaction. Disease-causing mutations in the head domains of both proteins cause enhanced cross-β interactions. By assembling desmin and NFL IFs bearing isotopically labeled head domains, we provide evidence of structural order in properly assembled biological filaments. We propose that these observations on IF head domains may be instructive to the function of low complexity domains operative in other aspects of cell biology. Low complexity (LC) head domains 92 and 108 residues in length are, respectively, required for assembly of neurofilament light (NFL) and desmin intermediate filaments (IFs). As studied in isolation, these IF head domains interconvert between states of conformational disorder and labile, β-strand–enriched polymers. Solid-state NMR (ss-NMR) spectroscopic studies of NFL and desmin head domain polymers reveal spectral patterns consistent with structural order. A combination of intein chemistry and segmental isotope labeling allowed preparation of fully assembled NFL and desmin IFs that could also be studied by ss-NMR. Assembled IFs revealed spectra overlapping with those observed for β-strand–enriched polymers formed from the isolated NFL and desmin head domains. Phosphorylation and disease-causing mutations reciprocally alter NFL and desmin head domain self-association yet commonly impede IF assembly. These observations show how facultative structural assembly of LC domains via labile, β-strand–enriched self-interactions may broadly influence cell morphology.
Collapse
|
46
|
Kim YB, Hlavaty D, Maycock J, Lechler T. Roles for Ndel1 in keratin organization and desmosome function. Mol Biol Cell 2021; 32:ar2. [PMID: 34319758 PMCID: PMC8684757 DOI: 10.1091/mbc.e21-02-0087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Keratin intermediate filaments form dynamic polymer networks that organize in specific ways dependent on the cell type, the stage of the cell cycle, and the state of the cell. In differentiated cells of the epidermis, they are organized by desmosomes, cell–cell adhesion complexes that provide essential mechanical integrity to this tissue. Despite this, we know little about how keratin organization is controlled and whether desmosomes locally regulate keratin dynamics in addition to binding preassembled filaments. Ndel1 is a desmosome-associated protein in the differentiated epidermis, though its function at these structures has not been examined. Here, we show that Ndel1 binds directly to keratin subunits through a motif conserved in all intermediate filament proteins. Further, Ndel1 was necessary for robust desmosome–keratin association and sufficient to reorganize keratins at distinct cellular sites. Lis1, a Ndel1 binding protein, was required for desmosomal localization of Ndel1, but not for its effects on keratin filaments. Finally, we use mouse genetics to demonstrate that loss of Ndel1 results in desmosome defects in the epidermis. Our data thus identify Ndel1 as a desmosome-associated protein that promotes local assembly/reorganization of keratin filaments and is essential for robust desmosome formation.
Collapse
Affiliation(s)
- Yong-Bae Kim
- Dept. of Cell Biology, Duke University Medical Center, Durham, NC 27710; USA.,Present Address - Institute of Immuno-Metabolic Disorders, ReCerise Therapeutics Inc., Seoul 07573, Republic of Korea
| | - Daniel Hlavaty
- Dept. of Cell Biology, Duke University Medical Center, Durham, NC 27710; USA.,Dept. of Dermatology, Duke University Medical Center, Durham, NC 27710; USA
| | - Jeff Maycock
- Dept. of Cell Biology, Duke University Medical Center, Durham, NC 27710; USA
| | - Terry Lechler
- Dept. of Cell Biology, Duke University Medical Center, Durham, NC 27710; USA.,Dept. of Dermatology, Duke University Medical Center, Durham, NC 27710; USA
| |
Collapse
|
47
|
Bouvet M, Dubois-Deruy E, Turkieh A, Mulder P, Peugnet V, Chwastyniak M, Beseme O, Dechaumes A, Amouyel P, Richard V, Lamblin N, Pinet F. Desmin aggrephagy in rat and human ischemic heart failure through PKCζ and GSK3β as upstream signaling pathways. Cell Death Discov 2021; 7:153. [PMID: 34226534 PMCID: PMC8257599 DOI: 10.1038/s41420-021-00549-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/29/2021] [Accepted: 06/01/2021] [Indexed: 12/21/2022] Open
Abstract
Post-translational modifications of cardiac proteins could participate to left contractile dysfunction resulting in heart failure. Using a rat model of ischemic heart failure, we showed an accumulation of phosphorylated desmin leading to toxic aggregates in cardiomyocytes, but the cellular mechanisms are unknown. The same rat model was used to decipher the kinases involved in desmin phosphorylation and the proteolytic systems present in rat and human failing hearts. We used primary cultures of neonate rat cardiomyocytes for testing specific inhibitors of kinases and for characterizing the autophagic processes able to clear desmin aggregates. We found a significant increase of active PKCζ, no modulation of ubitiquitin-proteasome system, a defect in macroautophagy, and an activation of chaperone-mediated autophagy in heart failure rats. We validated in vitro that PKCζ inhibition induced a significant decrease of GSK3β and of soluble desmin. In vitro activation of ubiquitination of proteins and of chaperone-mediated autophagy is able to decrease soluble and insoluble forms of desmin in cardiomyocytes. These data demonstrate a novel signaling pathway implicating activation of PKCζ in desmin phosphorylation associated with a defect of proteolytic systems in ischemic heart failure, leading to desmin aggrephagy. Our in vitro data demonstrated that ubiquitination of proteins and chaperone-mediated autophagy are required for eliminating desmin aggregates with the contribution of its chaperone protein, α-crystallin Β-chain. Modulation of the kinases involved under pathological conditions may help preserving desmin intermediate filaments structure and thus protect the structural integrity of contractile apparatus of cardiomyocytes by limiting desmin aggregates formation.
Collapse
Affiliation(s)
- Marion Bouvet
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Emilie Dubois-Deruy
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Annie Turkieh
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Paul Mulder
- Normandie Univ, UNIROUEN, Inserm U1096, FHU-REMOD-VHF, 76000, Rouen, France
| | - Victoriane Peugnet
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Maggy Chwastyniak
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Olivia Beseme
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Arthur Dechaumes
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Philippe Amouyel
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Vincent Richard
- Normandie Univ, UNIROUEN, Inserm U1096, FHU-REMOD-VHF, 76000, Rouen, France
| | - Nicolas Lamblin
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France
| | - Florence Pinet
- INSERM, Univ. Lille, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000, Lille, France.
| |
Collapse
|
48
|
Bertin B, Renaud Y, Jagla T, Lavergne G, Dondi C, Da Ponte JP, Junion G, Jagla K. Gelsolin and dCryAB act downstream of muscle identity genes and contribute to preventing muscle splitting and branching in Drosophila. Sci Rep 2021; 11:13197. [PMID: 34162956 PMCID: PMC8222376 DOI: 10.1038/s41598-021-92506-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/03/2021] [Indexed: 11/30/2022] Open
Abstract
A combinatorial code of identity transcription factors (iTFs) specifies the diversity of muscle types in Drosophila. We previously showed that two iTFs, Lms and Ap, play critical role in the identity of a subset of larval body wall muscles, the lateral transverse (LT) muscles. Intriguingly, a small portion of ap and lms mutants displays an increased number of LT muscles, a phenotype that recalls pathological split muscle fibers in human. However, genes acting downstream of Ap and Lms to prevent these aberrant muscle feature are not known. Here, we applied a cell type specific translational profiling (TRAP) to identify gene expression signatures underlying identity of muscle subsets including the LT muscles. We found that Gelsolin (Gel) and dCryAB, both encoding actin-interacting proteins, displayed LT muscle prevailing expression positively regulated by, the LT iTFs. Loss of dCryAB function resulted in LTs with irregular shape and occasional branched ends also observed in ap and lms mutant contexts. In contrast, enlarged and then split LTs with a greater number of myonuclei formed in Gel mutants while Gel gain of function resulted in unfused myoblasts, collectively indicating that Gel regulates LTs size and prevents splitting by limiting myoblast fusion. Thus, dCryAB and Gel act downstream of Lms and Ap and contribute to preventing LT muscle branching and splitting. Our findings offer first clues to still unknown mechanisms of pathological muscle splitting commonly detected in human dystrophic muscles and causing muscle weakness.
Collapse
Affiliation(s)
- Benjamin Bertin
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Yoan Renaud
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Teresa Jagla
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Guillaume Lavergne
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Cristiana Dondi
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Jean-Philippe Da Ponte
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Guillaume Junion
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France.
| | - Krzysztof Jagla
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France.
| |
Collapse
|
49
|
Santhoshkumar R, Preethish-Kumar V, Polavarapu K, Reghunathan D, Chaudhari S, Satyamoorthy K, Vengalil S, Nashi S, Faruq M, Joshi A, Atchayaram N, Narayanappa G. A Novel L1 Linker Mutation in DES Resulted in Total Absence of Protein. J Mol Neurosci 2021; 71:2468-2473. [PMID: 34106405 DOI: 10.1007/s12031-021-01856-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/06/2021] [Indexed: 11/29/2022]
Abstract
Desminopathies (MIM*601419) are clinically heterogeneous, manifesting with myopathy and/or cardiomyopathy and with intra-sarcoplasmic desmin-positive deposits. They have either an autosomal dominant (AD) or recessive (AR) pattern of inheritance. Desmin is a crucial intermediate filament protein regulating various cellular functions in muscle cells. Here, we report a 13-year-old girl, born of second-degree consanguineous parents, with normal developmental milestones, who presented with dilated cardiomyopathy, respiratory insufficiency and predominant distal upper limb weakness. A striking feature on muscle biopsy was the presence of a peripheral chain of nuclei in addition to myopathic features. Immunostaining showed complete lack of desmin expression, further confirmed by western blot analysis. Ultrastructurally, subsarcolemmal granular material, expanded Z-band aggregation, distortion of myofilaments, focal Z-band streaming, lobed and clustered myonuclei were observed. Next-generation sequencing revealed a novel homozygous nonsense mutation c.448C>T, p.R150X in the patient, while the parents were heterozygous carriers. Single mitochondrial DNA deletion and isolated complex IV deficiency were noted. Our findings add to the ever-expanding phenotype and molecular spectrum of desminopathies.
Collapse
Affiliation(s)
- Rashmi Santhoshkumar
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Veeramani Preethish-Kumar
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Dinesh Reghunathan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sima Chaudhari
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Muhammed Faruq
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110 007, India
| | - Aditi Joshi
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110 007, India
| | - Nalini Atchayaram
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Gayathri Narayanappa
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India.
| |
Collapse
|
50
|
Skeletal and Cardiac Muscle Disorders Caused by Mutations in Genes Encoding Intermediate Filament Proteins. Int J Mol Sci 2021; 22:ijms22084256. [PMID: 33923914 PMCID: PMC8073371 DOI: 10.3390/ijms22084256] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
Intermediate filaments are major components of the cytoskeleton. Desmin and synemin, cytoplasmic intermediate filament proteins and A-type lamins, nuclear intermediate filament proteins, play key roles in skeletal and cardiac muscle. Desmin, encoded by the DES gene (OMIM *125660) and A-type lamins by the LMNA gene (OMIM *150330), have been involved in striated muscle disorders. Diseases include desmin-related myopathy and cardiomyopathy (desminopathy), which can be manifested with dilated, restrictive, hypertrophic, arrhythmogenic, or even left ventricular non-compaction cardiomyopathy, Emery–Dreifuss Muscular Dystrophy (EDMD2 and EDMD3, due to LMNA mutations), LMNA-related congenital Muscular Dystrophy (L-CMD) and LMNA-linked dilated cardiomyopathy with conduction system defects (CMD1A). Recently, mutations in synemin (SYNM gene, OMIM *606087) have been linked to cardiomyopathy. This review will summarize clinical and molecular aspects of desmin-, lamin- and synemin-related striated muscle disorders with focus on LMNA and DES-associated clinical entities and will suggest pathogenetic hypotheses based on the interplay of desmin and lamin A/C. In healthy muscle, such interplay is responsible for the involvement of this network in mechanosignaling, nuclear positioning and mitochondrial homeostasis, while in disease it is disturbed, leading to myocyte death and activation of inflammation and the associated secretome alterations.
Collapse
|