1
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
2
|
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
|
3
|
Geryk M, Charpentier F. Pathophysiological mechanisms of cardiomyopathies induced by desmin gene variants located in the C-Terminus of segment 2B. J Cell Physiol 2024; 239:e31254. [PMID: 38501553 DOI: 10.1002/jcp.31254] [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: 12/12/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024]
Abstract
Desmin, the most abundant intermediate filament in cardiomyocytes, plays a key role in maintaining cardiomyocyte structure by interconnecting intracellular organelles, and facilitating cardiomyocyte interactions with the extracellular matrix and neighboring cardiomyocytes. As a consequence, mutations in the desmin gene (DES) can lead to desminopathies, a group of diseases characterized by variable and often severe cardiomyopathies along with skeletal muscle disorders. The basic desmin intermediate filament structure is composed of four segments separated by linkers that further assemble into dimers, tetramers and eventually unit-length filaments that compact radially to give the final form of the filament. Each step in this process is critical for proper filament formation and allow specific interactions within the cell. Mutations within the desmin gene can disrupt filament formation, as seen by aggregate formation, and thus have severe cardiac and skeletal outcomes, depending on the locus of the mutation. The focus of this review is to outline the cardiac molecular consequences of mutations located in the C-terminal part of segment 2B. This region is crucial for ensuring proper desmin filament formation and is a known hotspot for mutations that significantly impact cardiac function.
Collapse
Affiliation(s)
- Michelle Geryk
- Nantes Université, CNRS, INSERM, L'institut du thorax, Nantes, F-44000, France
| | - Flavien Charpentier
- Nantes Université, CNRS, INSERM, L'institut du thorax, Nantes, F-44000, France
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Blackwell DJ, Schmeckpeper J, Knollmann BC. Animal Models to Study Cardiac Arrhythmias. Circ Res 2022; 130:1926-1964. [PMID: 35679367 DOI: 10.1161/circresaha.122.320258] [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] [Indexed: 11/16/2022]
Abstract
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide, accounting for 10% to 15% of all deaths. Although most arrhythmias are due to acquired heart disease, inherited channelopathies and cardiomyopathies disproportionately affect children and young adults. Arrhythmogenesis is complex, involving anatomic structure, ion channels and regulatory proteins, and the interplay between cells in the conduction system, cardiomyocytes, fibroblasts, and the immune system. Animal models of arrhythmia are powerful tools for studying not only molecular and cellular mechanism of arrhythmogenesis but also more complex mechanisms at the whole heart level, and for testing therapeutic interventions. This review summarizes basic and clinical arrhythmia mechanisms followed by an in-depth review of published animal models of genetic and acquired arrhythmia disorders.
Collapse
Affiliation(s)
- Daniel J Blackwell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Jeffrey Schmeckpeper
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
7
|
Hakibilen C, Delort F, Daher MT, Joanne P, Cabet E, Cardoso O, Bourgois-Rocha F, Tian C, Rivas E, Madruga M, Ferreiro A, Lilienbaum A, Vicart P, Agbulut O, Hénon S, Batonnet-Pichon S. Desmin Modulates Muscle Cell Adhesion and Migration. Front Cell Dev Biol 2022; 10:783724. [PMID: 35350386 PMCID: PMC8957967 DOI: 10.3389/fcell.2022.783724] [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: 09/26/2021] [Accepted: 02/09/2022] [Indexed: 12/04/2022] Open
Abstract
Cellular adhesion and migration are key functions that are disrupted in numerous diseases. We report that desmin, a type-III muscle-specific intermediate filament, is a novel cell adhesion regulator. Expression of p.R406W mutant desmin, identified in patients with desmin-related myopathy, modified focal adhesion area and expression of adhesion-signaling genes in myogenic C2C12 cells. Satellite cells extracted from desmin-knock-out (DesKO) and desmin-knock-in-p.R405W (DesKI-R405W) mice were less adhesive and migrated faster than those from wild-type mice. Moreover, we observed mislocalized and aggregated vinculin, a key component of cell adhesion, in DesKO and DesKI-R405W muscles. Vinculin expression was also increased in desmin-related myopathy patient muscles. Together, our results establish a novel role for desmin in cell-matrix adhesion, an essential process for strength transmission, satellite cell migration and muscle regeneration. Our study links the patho-physiological mechanisms of desminopathies to adhesion/migration defects, and may lead to new cellular targets for novel therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Eva Cabet
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | | | | | - Cuixia Tian
- Department of Neurology, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Eloy Rivas
- Servicio de Anatomia Patologica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain
| | - Marcos Madruga
- Unidad de Neurologia Pediatrica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain
| | - Ana Ferreiro
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France.,APHP, Centre de Référence Maladies Neuromusculaires Nord/Est/Ile-de-France, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | - Patrick Vicart
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France.,Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France.,Université de Paris, MSC, UMR 7067, CNRS, Paris, France.,Department of Neurology, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, United States.,Servicio de Anatomia Patologica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain.,Unidad de Neurologia Pediatrica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain.,APHP, Centre de Référence Maladies Neuromusculaires Nord/Est/Ile-de-France, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Sylvie Hénon
- Université de Paris, MSC, UMR 7067, CNRS, Paris, France
| | | |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Skeletal Muscle Mitochondria Dysfunction in Genetic Neuromuscular Disorders with Cardiac Phenotype. Int J Mol Sci 2021; 22:ijms22147349. [PMID: 34298968 PMCID: PMC8307986 DOI: 10.3390/ijms22147349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction is considered the major contributor to skeletal muscle wasting in different conditions. Genetically determined neuromuscular disorders occur as a result of mutations in the structural proteins of striated muscle cells and therefore are often combined with cardiac phenotype, which most often manifests as a cardiomyopathy. The specific roles played by mitochondria and mitochondrial energetic metabolism in skeletal muscle under muscle-wasting conditions in cardiomyopathies have not yet been investigated in detail, and this aspect of genetic muscle diseases remains poorly characterized. This review will highlight dysregulation of mitochondrial representation and bioenergetics in specific skeletal muscle disorders caused by mutations that disrupt the structural and functional integrity of muscle cells.
Collapse
|
12
|
Fischer B, Dittmann S, Brodehl A, Unger A, Stallmeyer B, Paul M, Seebohm G, Kayser A, Peischard S, Linke WA, Milting H, Schulze-Bahr E. Functional characterization of novel alpha-helical rod domain desmin (DES) pathogenic variants associated with dilated cardiomyopathy, atrioventricular block and a risk for sudden cardiac death. Int J Cardiol 2020; 329:167-174. [PMID: 33373648 DOI: 10.1016/j.ijcard.2020.12.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Desmin is the major intermediate filament (IF) protein in human heart and skeletal muscle. So-called 'desminopathies' are disorders due to pathogenic variants in the DES gene and are associated with skeletal myopathies and/or various types of cardiomyopathies. So far, only a limited number of DES pathogenic variants have been identified and functionally characterized. METHODS AND RESULTS Using a Sanger- and next generation sequencing (NGS) approach in patients with various types of cardiomyopathies, we identified two novel, non-synonymous missense DES variants: p.(Ile402Thr) and p.(Glu410Lys). Mutation carriers developed dilated (DCM) or arrhythmogenic cardiomyopathy (ACM), and cardiac conduction disease, leading to spare out the exercise-induced polymorphic ventricular tachycardia; we moved this variant to data in brief. To investigate the functional impact of these four DES variants, transfection experiments using SW-13 and H9c2 cells with native and mutant desmin were performed and filament assembly was analyzed by confocal microscopy. The DES_p.(Ile402Thr) and DES_p.(Glu410Lys) cells showed filament assembly defects forming cytoplasmic desmin aggregates. Furthermore, immunohistochemical and ultrastructural analysis of myocardial tissue from mutation carriers with the DES_p.(Glu410Lys) pathogenic variant supported the in vitro results. CONCLUSIONS Our in vitro results supported the classification of DES_p.(Ile402Thr) and DES_p.(Glu410Lys) as novel pathogenic variants and demonstrated that the cardiac phenotypes associated with DES variants are diverse and cell culture experiments improve in silico analysis and genetic counseling because the pathogenicity of a variant can be clarified.
Collapse
Affiliation(s)
- Björn Fischer
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Sven Dittmann
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany.
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Muenster, Germany
| | - Birgit Stallmeyer
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Matthias Paul
- Department of Cardiology I, University Hospital Muenster, Muenster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Anne Kayser
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Stefan Peischard
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | | | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Disease (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
13
|
246th ENMC International Workshop: Protein aggregate myopathies 24-26 May 2019, Hoofddorp, The Netherlands. Neuromuscul Disord 2020; 31:158-166. [PMID: 33303357 DOI: 10.1016/j.nmd.2020.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022]
|
14
|
Eiber N, Fröb F, Schowalter M, Thiel C, Clemen CS, Schröder R, Hashemolhosseini S. Lack of Desmin in Mice Causes Structural and Functional Disorders of Neuromuscular Junctions. Front Mol Neurosci 2020; 13:567084. [PMID: 33192292 PMCID: PMC7649329 DOI: 10.3389/fnmol.2020.567084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022] Open
Abstract
Desmin, the major intermediate filament (IF) protein in muscle cells, interlinks neighboring myofibrils and connects the whole myofibrillar apparatus to myonuclei, mitochondria, and the sarcolemma. However, desmin is also known to be enriched at postsynaptic membranes of neuromuscular junctions (NMJs). The pivotal role of the desmin IF cytoskeletal network is underscored by the fact that over 120 mutations of the human DES gene cause hereditary and sporadic myopathies and cardiomyopathies. A subgroup of human desminopathies comprises autosomal recessive cases resulting in the complete abolition of desmin protein. In these patients, who display a more severe phenotype than the autosomal dominant cases, it has been reported that some individuals also suffer from a myasthenic syndrome in addition to the classical occurrence of myopathy and cardiomyopathy. Since further studies on the NMJ pathology are hampered by the lack of available human striated muscle biopsy specimens, we exploited homozygous desmin knock-out mice which closely mirror the striated muscle pathology of human patients lacking desmin protein. Here, we report on the impact of the lack of desmin on the structure and function of NMJs and the transcription of genes coding for postsynaptic proteins. Desmin knock-out mice display a fragmentation of NMJs in soleus, but not in the extensor digitorum longus muscle. Moreover, soleus muscle fibers show larger NMJs. Further, transcription levels of acetylcholine receptor (AChR) genes are increased in muscles from desmin knock-out mice, especially of the AChRγ subunit, which is known as a marker of muscle fiber regeneration. Electrophysiological recordings depicted a pathological decrement of nerve-dependent endplate potentials and an increased rise time of the nerve-independent miniature endplate potentials. The latter appears related to the fragmentation of NMJs in desmin knockout mice. Our study highlights the essential role of desmin for the structural and functional integrity of mammalian NMJs.
Collapse
Affiliation(s)
- Nane Eiber
- Institute of Biochemistry, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Fröb
- Institute of Biochemistry, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Mirjam Schowalter
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Thiel
- Medical Faculty, Institute of Human Genetics, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph S Clemen
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany.,Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany.,Medical Faculty, Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Said Hashemolhosseini
- Institute of Biochemistry, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany.,Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
15
|
Herrmann H, Cabet E, Chevalier NR, Moosmann J, Schultheis D, Haas J, Schowalter M, Berwanger C, Weyerer V, Agaimy A, Meder B, Müller OJ, Katus HA, Schlötzer-Schrehardt U, Vicart P, Ferreiro A, Dittrich S, Clemen CS, Lilienbaum A, Schröder R. Dual Functional States of R406W-Desmin Assembly Complexes Cause Cardiomyopathy With Severe Intercalated Disc Derangement in Humans and in Knock-In Mice. Circulation 2020; 142:2155-2171. [PMID: 33023321 DOI: 10.1161/circulationaha.120.050218] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mutations in the human desmin gene cause myopathies and cardiomyopathies. This study aimed to elucidate molecular mechanisms initiated by the heterozygous R406W-desmin mutation in the development of a severe and early-onset cardiac phenotype. METHODS We report an adolescent patient who underwent cardiac transplantation as a result of restrictive cardiomyopathy caused by a heterozygous R406W-desmin mutation. Sections of the explanted heart were analyzed with antibodies specific to 406W-desmin and to intercalated disc proteins. Effects of the R406W mutation on the molecular properties of desmin were addressed by cell transfection and in vitro assembly experiments. To prove the genuine deleterious effect of the mutation on heart tissue, we further generated and analyzed R405W-desmin knock-in mice harboring the orthologous form of the human R406W-desmin. RESULTS Microscopic analysis of the explanted heart revealed desmin aggregates and the absence of desmin filaments at intercalated discs. Structural changes within intercalated discs were revealed by the abnormal organization of desmoplakin, plectin, N-cadherin, and connexin-43. Next-generation sequencing confirmed the DES variant c.1216C>T (p.R406W) as the sole disease-causing mutation. Cell transfection studies disclosed a dual behavior of R406W-desmin with both its integration into the endogenous intermediate filament system and segregation into protein aggregates. In vitro, R406W-desmin formed unusually thick filaments that organized into complex filament aggregates and fibrillar sheets. In contrast, assembly of equimolar mixtures of mutant and wild-type desmin generated chimeric filaments of seemingly normal morphology but with occasional prominent irregularities. Heterozygous and homozygous R405W-desmin knock-in mice develop both a myopathy and a cardiomyopathy. In particular, the main histopathologic results from the patient are recapitulated in the hearts from R405W-desmin knock-in mice of both genotypes. Moreover, whereas heterozygous knock-in mice have a normal life span, homozygous animals die at 3 months of age because of a smooth muscle-related gastrointestinal phenotype. CONCLUSIONS We demonstrate that R406W-desmin provokes its severe cardiotoxic potential by a novel pathomechanism, where the concurrent dual functional states of mutant desmin assembly complexes underlie the uncoupling of desmin filaments from intercalated discs and their structural disorganization.
Collapse
Affiliation(s)
- Harald Herrmann
- Institute of Neuropathology (H.H., D.S., M.S., R.S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany.,Molecular Genetics, German Cancer Research Center, Heidelberg, Germany (H.H.)
| | - Eva Cabet
- Basic and Translational Myology, Unit of Functional and Adaptive Biology (E.C., P.V., A.F., A.L.), University of Paris, France
| | - Nicolas R Chevalier
- Laboratoire Matière et Systèmes Complexes (N.R.C.), University of Paris, France
| | - Julia Moosmann
- Department of Pediatric Cardiology (J.M., S.D.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Dorothea Schultheis
- Institute of Neuropathology (H.H., D.S., M.S., R.S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Jan Haas
- Institute for Cardiomyopathies Heidelberg, Heart Center Heidelberg, University of Heidelberg, Germany (J.H., B.M.)
| | - Mirjam Schowalter
- Institute of Neuropathology (H.H., D.S., M.S., R.S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Carolin Berwanger
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany (C.B., C.S.C.)
| | - Veronika Weyerer
- Institute of Pathology (V.W., A.A.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Abbas Agaimy
- Institute of Pathology (V.W., A.A.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies Heidelberg, Heart Center Heidelberg, University of Heidelberg, Germany (J.H., B.M.).,Department of Genetics, Stanford University School of Medicine, CA (B.M.)
| | - Oliver J Müller
- Internal Medicine III, University Hospital Schleswig-Holstein and University of Kiel, and German Center for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Kiel, Germany (O.J.M.)
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, and German Center for Cardiovascular Research, partner site Heidelberg/Mannheim, Heidelberg, Germany (H.A.K.)
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology (U.S.-S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Patrick Vicart
- Basic and Translational Myology, Unit of Functional and Adaptive Biology (E.C., P.V., A.F., A.L.), University of Paris, France
| | - Ana Ferreiro
- Basic and Translational Myology, Unit of Functional and Adaptive Biology (E.C., P.V., A.F., A.L.), University of Paris, France.,Reference Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, Assistance publique-Hôpitaux de Paris, France (A.F.)
| | - Sven Dittrich
- Department of Pediatric Cardiology (J.M., S.D.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany (C.B., C.S.C.).,Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Germany(C.S.C.)
| | - Alain Lilienbaum
- Basic and Translational Myology, Unit of Functional and Adaptive Biology (E.C., P.V., A.F., A.L.), University of Paris, France
| | - Rolf Schröder
- Institute of Neuropathology (H.H., D.S., M.S., R.S.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| |
Collapse
|
16
|
Langer HT, Mossakowski AA, Willis BJ, Grimsrud KN, Wood JA, Lloyd KCK, Zbinden-Foncea H, Baar K. Generation of desminopathy in rats using CRISPR-Cas9. J Cachexia Sarcopenia Muscle 2020; 11:1364-1376. [PMID: 32893996 PMCID: PMC7567154 DOI: 10.1002/jcsm.12619] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/04/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Desminopathy is a clinically heterogeneous muscle disease caused by over 60 different mutations in desmin. The most common mutation with a clinical phenotype in humans is an exchange of arginine to proline at position 350 of desmin leading to p.R350P. We created the first CRISPR-Cas9 engineered rat model for a muscle disease by mirroring the R350P mutation in humans. METHODS Using CRISPR-Cas9 technology, Des c.1045-1046 (AGG > CCG) was introduced into exon 6 of the rat genome causing p.R349P. The genotype of each animal was confirmed via quantitative PCR. Six male rats with a mutation in desmin (n = 6) between the age of 120-150 days and an equal number of wild type littermates (n = 6) were used for experiments. Maximal plantar flexion force was measured in vivo and combined with the collection of muscle weights, immunoblotting, and histological analysis. In addition to the baseline phenotyping, we performed a synergist ablation study in the same animals. RESULTS We found a difference in the number of central nuclei between desmin mutants (1 ± 0.4%) and wild type littermates (0.2 ± 0.1%; P < 0.05). While muscle weights did not differ, we found the levels of many structural proteins to be altered in mutant animals. Dystrophin and syntrophin were increased 54% and 45% in desmin mutants, respectively (P < 0.05). Dysferlin and Annexin A2, proteins associated with membrane repair, were increased two-fold and 32%, respectively, in mutants (P < 0.05). Synergist ablation caused similar increases in muscle weight between mutant and wild type animals, but changes in fibre diameter revealed that fibre hypertrophy in desmin mutants was hampered compared with wild type animals (P < 0.05). CONCLUSIONS We created a novel animal model for desminopathy that will be a useful tool in furthering our understanding of the disease. While mutant animals at an age corresponding to a preclinical age in humans show no macroscopic differences, microscopic and molecular changes are already present. Future studies should aim to further decipher those biological changes that precede the clinical progression of disease and test therapeutic approaches to delay disease progression.
Collapse
Affiliation(s)
- Henning T Langer
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| | - Agata A Mossakowski
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.,Department of Neurology with Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Universität zu Berlin, and Berlin Institute of Health, Humboldt, CA, USA
| | | | - Kristin N Grimsrud
- Mouse Biology Program, University of California, Davis, CA, USA.,Dept. of Pathology, School of Medicine, University of California, Davis, CA, USA
| | - Joshua A Wood
- Mouse Biology Program, University of California, Davis, CA, USA
| | - Kevin C K Lloyd
- Mouse Biology Program, University of California, Davis, CA, USA.,Dept. of Surgery, School of Medicine, University of California, Davis, CA, USA
| | | | - Keith Baar
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.,Neurobiology, Physiology and Behavior, University of California, Davis, CA, USA
| |
Collapse
|
17
|
Dennhag N, Liu JX, Nord H, von Hofsten J, Pedrosa Domellöf F. Absence of Desmin in Myofibers of the Zebrafish Extraocular Muscles. Transl Vis Sci Technol 2020; 9:1. [PMID: 32953241 PMCID: PMC7476663 DOI: 10.1167/tvst.9.10.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/14/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose To study the medial rectus (MR) muscle of zebrafish (Daniorerio) with respect to the pattern of distribution of desmin and its correlation to distinct types of myofibers and motor endplates. Methods The MRs of zebrafish were examined using confocal microscopy in whole-mount longitudinal specimens and in cross sections processed for immunohistochemistry with antibodies against desmin, myosin heavy chain isoforms, and innervation markers. Desmin patterns were correlated to major myofiber type and type of innervation. A total of 1382 myofibers in nine MR muscles were analyzed. Results Four distinct desmin immunolabeling patterns were found in the zebrafish MRs. Approximately a third of all slow myofibers lacked desmin, representing 8.5% of the total myofiber population. The adult zebrafish MR muscle displayed en grappe, en plaque, and multiterminal en plaque neuromuscular junctions (NMJs) with intricate patterns of desmin immunolabeling. Conclusions The MRs of zebrafish showed important similarities with the human extraocular muscles with regard to the pattern of desmin distribution and presence of the major types of NMJs and can be regarded as an adequate model to further study the role of desmin and the implications of heterogeneity in cytoskeletal protein composition. Translational Relevance The establishment of a zebrafish model to study the cytoskeleton in muscles that are particularly resistant to muscle disease opens new avenues to understand human myopathies and muscle dystrophies and may provide clues to new therapies.
Collapse
Affiliation(s)
- Nils Dennhag
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jing-Xia Liu
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Hanna Nord
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jonas von Hofsten
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Fatima Pedrosa Domellöf
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Department of Clinical Science, Ophthalmology, Umeå University, Umeå, Sweden
| |
Collapse
|
18
|
Li L, Zhang Q, Lei X, Huang Y, Hu J. MAP4 as a New Candidate in Cardiovascular Disease. Front Physiol 2020; 11:1044. [PMID: 32982783 PMCID: PMC7479186 DOI: 10.3389/fphys.2020.01044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022] Open
Abstract
Microtubule and mitochondrial dysfunction have been implicated in the pathogenesis of cardiovascular diseases (CVDs), including cardiac hypertrophy, fibrosis, heart failure, and hypoxic/ischemic related heart dysfunction. Microtubule dynamics instability leads to disrupted cell homeostasis and cell shape, decreased cell survival, and aberrant cell division and cell cycle, while mitochondrial dysfunction contributes to abnormal metabolism and calcium flux, increased cell death, oxidative stress, and inflammation, both of which causing cell and tissue dysfunction followed by CVDs. A cytosolic skeleton protein, microtubule-associated protein 4 (MAP4), belonging to the family of microtubule-associated proteins (MAPs), is widely expressed in non-neural cells and possesses an important role in microtubule dynamics. Increased MAP4 phosphorylation results in microtubule instability. In addition, MAP4 also expresses in mitochondria and reveals a crucial role in maintaining mitochondrial homeostasis. Phosphorylated MAP4 promotes mitochondrial apoptosis, followed by cardiac injury. The aim of the present review is to highlight the novel role of MAP4 as a potential candidate in multiple cardiovascular pathologies.
Collapse
Affiliation(s)
- Lingfei Li
- Department of Dermatology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xia Lei
- Department of Dermatology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiongyu Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
19
|
Pollmann C, Haug M, Reischl B, Prölß G, Pöschel T, Rupitsch SJ, Clemen CS, Schröder R, Friedrich O. Growing Old Too Early: Skeletal Muscle Single Fiber Biomechanics in Ageing R349P Desmin Knock-in Mice Using the MyoRobot Technology. Int J Mol Sci 2020; 21:ijms21155501. [PMID: 32752098 PMCID: PMC7432536 DOI: 10.3390/ijms21155501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 11/16/2022] Open
Abstract
Muscle biomechanics relies on active motor protein assembly and passive strain transmission through cytoskeletal structures. The desmin filament network aligns myofibrils at the z-discs, provides nuclear–sarcolemmal anchorage and may also serve as memory for muscle repositioning following large strains. Our previous analyses of R349P desmin knock-in mice, an animal model for the human R350P desminopathy, already depicted pre-clinical changes in myofibrillar arrangement and increased fiber bundle stiffness. As the effect of R349P desmin on axial biomechanics in fully differentiated single muscle fibers is unknown, we used our MyoRobot to compare passive visco-elasticity and active contractile biomechanics in single fibers from fast- and slow-twitch muscles from adult to senile mice, hetero- or homozygous for the R349P desmin mutation with wild type littermates. We demonstrate that R349P desmin presence predominantly increased axial stiffness in both muscle types with a pre-aged phenotype over wild type fibers. Axial viscosity and Ca2+-mediated force were largely unaffected. Mutant single fibers showed tendencies towards faster unloaded shortening over wild type fibers. Effects of aging seen in the wild type appeared earlier in the mutant desmin fibers. Our single-fiber experiments, free of extracellular matrix, suggest that compromised muscle biomechanics is not exclusively attributed to fibrosis but also originates from an impaired intermediate filament network.
Collapse
Affiliation(s)
- Charlotte Pollmann
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Bavaria, Germany; (C.P.); (B.R.); (G.P.); (O.F.)
| | - Michael Haug
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Bavaria, Germany; (C.P.); (B.R.); (G.P.); (O.F.)
- Graduate School in Advanced Optical Technologies, Paul-Gordan-Str. 6, 91052 Erlangen, Bavaria, Germany
- School of Medical Sciences, University of New South Wales, Wallace Wurth Building, 18 High St, Sydney, NSW 2052, Australia
- Correspondence:
| | - Barbara Reischl
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Bavaria, Germany; (C.P.); (B.R.); (G.P.); (O.F.)
| | - Gerhard Prölß
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Bavaria, Germany; (C.P.); (B.R.); (G.P.); (O.F.)
| | - Thorsten Pöschel
- Institute of Multi Scale Simulation of Particulate Systems, Friedrich-Alexander-University Erlangen-Nürnberg, Nägelbachstr. 49b, 91052 Erlangen, Bavaria, Germany;
| | - Stefan J Rupitsch
- Institute of Sensor Technology, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3/5, 91052 Erlangen, Bavaria, Germany;
| | - Christoph S Clemen
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Linder Höhe, 51147 Cologne, North Rhine-Westphalia, Germany;
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Bavaria, Germany;
- Insitute of Vegetative Physiology, Medical Faculty, University of Cologne, Center of Physiology and Pathophysiology, Robert-Koch-Street 39, 50931 Cologne, North Rhine-Westphalia, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Bavaria, Germany;
- Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Bavaria, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Bavaria, Germany; (C.P.); (B.R.); (G.P.); (O.F.)
- Graduate School in Advanced Optical Technologies, Paul-Gordan-Str. 6, 91052 Erlangen, Bavaria, Germany
- School of Medical Sciences, University of New South Wales, Wallace Wurth Building, 18 High St, Sydney, NSW 2052, Australia
- Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Bavaria, Germany
- Victor Chang Cardiac Research Institute, Lowy Packer Building, 405 Liverpool St, Sydney, NSW 2010, Australia
- Optical Imaging Centre Erlangen OICE, Cauerstr. 3, 91058 Erlangen, Bavaria, Germany
| |
Collapse
|
20
|
Gerull B, Brodehl A. Genetic Animal Models for Arrhythmogenic Cardiomyopathy. Front Physiol 2020; 11:624. [PMID: 32670084 PMCID: PMC7327121 DOI: 10.3389/fphys.2020.00624] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Arrhythmogenic cardiomyopathy has been clinically defined since the 1980s and causes right or biventricular cardiomyopathy associated with ventricular arrhythmia. Although it is a rare cardiac disease, it is responsible for a significant proportion of sudden cardiac deaths, especially in athletes. The majority of patients with arrhythmogenic cardiomyopathy carry one or more genetic variants in desmosomal genes. In the 1990s, several knockout mouse models of genes encoding for desmosomal proteins involved in cell-cell adhesion revealed for the first time embryonic lethality due to cardiac defects. Influenced by these initial discoveries in mice, arrhythmogenic cardiomyopathy received an increasing interest in human cardiovascular genetics, leading to the discovery of mutations initially in desmosomal genes and later on in more than 25 different genes. Of note, even in the clinic, routine genetic diagnostics are important for risk prediction of patients and their relatives with arrhythmogenic cardiomyopathy. Based on improvements in genetic animal engineering, different transgenic, knock-in, or cardiac-specific knockout animal models for desmosomal and nondesmosomal proteins have been generated, leading to important discoveries in this field. Here, we present an overview about the existing animal models of arrhythmogenic cardiomyopathy with a focus on the underlying pathomechanism and its importance for understanding of this disease. Prospectively, novel mechanistic insights gained from the whole animal, organ, tissue, cellular, and molecular levels will lead to the development of efficient personalized therapies for treatment of arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Brenda Gerull
- Comprehensive Heart Failure Center Wuerzburg, Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany.,Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospitals of the Ruhr-University of Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
21
|
AAV-mediated cardiac gene transfer of wild-type desmin in mouse models for recessive desminopathies. Gene Ther 2020; 27:516-524. [PMID: 32322014 PMCID: PMC7674151 DOI: 10.1038/s41434-020-0147-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 11/23/2022]
Abstract
Mutations in the human desmin gene cause autosomal-dominant and recessive cardiomyopathies and myopathies with marked phenotypic variability. Here, we investigated the effects of adeno-associated virus (AAV)-mediated cardiac wild-type desmin expression in homozygous desmin knockout (DKO) and homozygous R349P desmin knockin (DKI) mice. These mice serve as disease models for two subforms of autosomal-recessive desminopathies, the former for the one with a complete lack of desmin protein and the latter for the one with solely mutant desmin protein expression in conjunction with protein aggregation pathology in striated muscle. Two-month-old mice were injected with either a single dose of 5 × 1012 AAV9-hTNT2-mDes (AAV-Des) vector genomes or NaCl as control. One week after injection, mice were subjected to a forced swimming exercise protocol for 4 weeks. Cardiac function was monitored over a period of 15 month after injection and before the mice were sacrificed for biochemical and morphological analysis. AAV-mediated cardiac expression of wild-type desmin in both the homozygous DKO and DKI backgrounds reached levels seen in wild-type mice. Notably, AAV-Des treated DKO mice showed a regular subcellular distribution of desmin as well as a normalization of functional and morphological cardiac parameters. Treated DKI mice, however, showed an aberrant subcellular localization of desmin, unchanged functional cardiac parameters, and a trend toward an increased cardiac fibrosis. In conclusion, the effect of a high-dose AAV9-based desmin gene therapy is highly beneficial for the heart in DKO animals, but not in DKI mice.
Collapse
|
22
|
Stöckigt F, Eichhorn L, Beiert T, Knappe V, Radecke T, Steinmetz M, Nickenig G, Peeva V, Kudin AP, Kunz WS, Berwanger C, Kamm L, Schultheis D, Schlötzer-Schrehardt U, Clemen CS, Schröder R, Schrickel JW. Heart failure after pressure overload in autosomal-dominant desminopathies: Lessons from heterozygous DES-p.R349P knock-in mice. PLoS One 2020; 15:e0228913. [PMID: 32126091 PMCID: PMC7053759 DOI: 10.1371/journal.pone.0228913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/27/2020] [Indexed: 11/21/2022] Open
Abstract
Background Mutations in the human desmin gene (DES) cause autosomal-dominant and -recessive cardiomyopathies, leading to heart failure, arrhythmias, and AV blocks. We analyzed the effects of vascular pressure overload in a patient-mimicking p.R349P desmin knock-in mouse model that harbors the orthologue of the frequent human DES missense mutation p.R350P. Methods and results Transverse aortic constriction (TAC) was performed on heterozygous (HET) DES-p.R349P mice and wild-type (WT) littermates. Echocardiography demonstrated reduced left ventricular ejection fraction in HET-TAC (WT-sham: 69.5 ± 2.9%, HET-sham: 64.5 ± 4.7%, WT-TAC: 63.5 ± 4.9%, HET-TAC: 55.7 ± 5.4%; p<0.01). Cardiac output was significantly reduced in HET-TAC (WT sham: 13088 ± 2385 μl/min, HET sham: 10391 ± 1349μl/min, WT-TAC: 8097 ± 1903μl/min, HET-TAC: 5793 ± 2517μl/min; p<0.01). Incidence and duration of AV blocks as well as the probability to induce ventricular tachycardias was highest in HET-TAC. We observed reduced mtDNA copy numbers in HET-TAC (WT-sham: 12546 ± 406, HET-sham: 13526 ± 781, WT-TAC: 11155 ± 3315, HET-TAC: 8649 ± 1582; p = 0.025), but no mtDNA deletions. The activity of respiratory chain complexes I and IV showed the greatest reductions in HET-TAC. Conclusion Pressure overload in HET mice aggravated the clinical phenotype of cardiomyopathy and resulted in mitochondrial dysfunction. Preventive avoidance of pressure overload/arterial hypertension in desminopathy patients might represent a crucial therapeutic measure.
Collapse
Affiliation(s)
- Florian Stöckigt
- Department of Cardiology, University Hospital Bonn, Bonn, Germany
- Department of Cardiology, Krankenhaus Porz, Urbacher Weg, Cologne, Germany
- * E-mail:
| | - Lars Eichhorn
- Department of Anesthesiology, University Hospital Bonn, Bonn, Germany
| | - Thomas Beiert
- Department of Cardiology, University Hospital Bonn, Bonn, Germany
| | - Vincent Knappe
- Department of Cardiology, University Hospital Bonn, Bonn, Germany
| | - Tobias Radecke
- Department of Cardiology, University Hospital Essen, Hufelandstraße, Essen, Germany
| | - Martin Steinmetz
- Department of Cardiology, University Hospital Essen, Hufelandstraße, Essen, Germany
| | - Georg Nickenig
- Department of Cardiology, University Hospital Bonn, Bonn, Germany
| | - Viktoriya Peeva
- Institute of Experimental Epileptology and Cognition Research, Bonn, Germany
- Department of Epileptology, University Hospital of Bonn, Bonn, Germany
| | - Alexei P. Kudin
- Institute of Experimental Epileptology and Cognition Research, Bonn, Germany
- Department of Epileptology, University Hospital of Bonn, Bonn, Germany
| | - Wolfram S. Kunz
- Institute of Experimental Epileptology and Cognition Research, Bonn, Germany
- Department of Epileptology, University Hospital of Bonn, Bonn, Germany
| | - Carolin Berwanger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Linder Höhe, Cologne, Germany
| | - Lisa Kamm
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage, Erlangen, Germany
| | - Dorothea Schultheis
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage, Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Opthalmology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage, Erlangen, Germany
| | - Christoph S. Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Linder Höhe, Cologne, Germany
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage, Erlangen, Germany
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage, Erlangen, Germany
| | - Jan W. Schrickel
- Department of Cardiology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
23
|
Ward M, Iskratsch T. Mix and (mis-)match - The mechanosensing machinery in the changing environment of the developing, healthy adult and diseased heart. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118436. [PMID: 30742931 PMCID: PMC7042712 DOI: 10.1016/j.bbamcr.2019.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
The composition and the stiffness of cardiac microenvironment change during development and/or in heart disease. Cardiomyocytes (CMs) and their progenitors sense these changes, which decides over the cell fate and can trigger CM (progenitor) proliferation, differentiation, de-differentiation or death. The field of mechanobiology has seen a constant increase in output that also includes a wealth of new studies specific to cardiac or cardiomyocyte mechanosensing. As a result, mechanosensing and transduction in the heart is increasingly being recognised as a main driver of regulating the heart formation and function. Recent work has for instance focused on measuring the molecular, physical and mechanical changes of the cellular environment - as well as intracellular contributors to the passive stiffness of the heart. On the other hand, a variety of new studies shed light into the molecular machinery that allow the cardiomyocytes to sense these properties. Here we want to discuss the recent work on this topic, but also specifically focus on how the different components are regulated at various stages during development, in health or disease in order to highlight changes that might contribute to disease progression and heart failure.
Collapse
Key Words
- cm, cardiomyocytes
- hcm, hypertrophic cardiomyopathy
- dcm, dilated cardiomyopathy
- icm, idiopathic cardiomyopathy
- myh, myosin heavy chain
- tnnt, troponin t
- tnni, troponin i
- afm, atomic force microscope
- mre, magnetic resonance elastography
- swe, ultrasound cardiac shear-wave elastography
- lv, left ventricle
- lox, lysyl oxidase
- loxl, lysyl oxidase like protein
- lh, lysyl hydroxylase
- lys, lysin
- lccs, lysald-derived collagen crosslinks
- hlccs, hylald-derived collagen crosslinks
- pka, protein kinase a
- pkc, protein kinase c
- vash1, vasohibin-1
- svbp, small vasohibin binding protein
- tcp, tubulin carboxypeptidase
- ttl, tubulin tyrosine ligase
- mrtf, myocardin-related transcription factor
- gap, gtpase activating protein
- gef, guanine nucleotide exchange factor
Collapse
Affiliation(s)
- Matthew Ward
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom.
| |
Collapse
|
24
|
Paulin D, Hovhannisyan Y, Kasakyan S, Agbulut O, Li Z, Xue Z. Synemin-related skeletal and cardiac myopathies: an overview of pathogenic variants. Am J Physiol Cell Physiol 2020; 318:C709-C718. [PMID: 32023076 DOI: 10.1152/ajpcell.00485.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review analyzes data concerning patients with cardiomyopathies or skeletal myopathies associated with a variation in the intermediate filament (IF) synemin gene (SYNM), also referred to as desmuslin (DMN). Molecular studies demonstrate that synemin copolymerizes with desmin and vimentin IF and interacts with vinculin, α-actinin, α-dystrobrevin, dystrophin, talin, and zyxin. It has been found that synemin is an A-kinase-anchoring protein (AKAP) that anchors protein kinase A (PKA) and modulates the PKA-dependent phosphorylation of several cytoskeletal substrates such as desmin. Because several IF proteins, including desmin, have been implicated in human genetic disorders such as dominant or recessive congenital and adult-onset myopathy, synemin becomes a significant candidate for cardiac and skeletal myopathies of unknown etiology. Because SYNM is a new candidate gene that displays numerous sequence polymorphisms, in this review, we summarize the genetic and clinical literature about SYNM mutations. Protein-changing variants (missense, frameshifts, nonsense) were further evaluated based on structural modifications and amino acid interactions. We present in silico modeling of helical salt-bridges between residues to evaluate the impact of the synemin networks crucial to interactions with cytoskeletal proteins. Finally, a discussion is featured regarding certain variants that may contribute to the disease state.
Collapse
Affiliation(s)
- Denise Paulin
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Yeranuhi Hovhannisyan
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Serdar Kasakyan
- Duzen Laboratories Group, Center of Genetic Diagnosis, Istanbul, Turkey
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Zhenlin Li
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Zhigang Xue
- Sorbonne Université, Institut de Biologie Paris-Seine, CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| |
Collapse
|
25
|
Robertson R, Conte TC, Dicaire MJ, Rymar VV, Sadikot AF, Bryson-Richardson RJ, Lavoie JN, O'Ferrall E, Young JC, Brais B. BAG3 P215L/KO Mice as a Model of BAG3 P209L Myofibrillar Myopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:554-562. [PMID: 31953038 DOI: 10.1016/j.ajpath.2019.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 10/25/2022]
Abstract
BCL-2-associated athanogene 3 (BAG3) is a co-chaperone to heat shock proteins important in degrading misfolded proteins through chaperone-assisted selective autophagy. The recurrent dominant BAG3-P209L mutation results in a severe childhood-onset myofibrillar myopathy (MFM) associated with progressive muscle weakness, cardiomyopathy, and respiratory failure. Because a homozygous knock-in (KI) strain for the mP215L mutation homologous to the human P209L mutation did not have a gross phenotype, compound heterozygote knockout (KO) and KI mP215L mice were generated to establish whether further reduction in BAG3 expression would lead to a phenotype. The KI/KO mice have a significant decrease in voluntary movement compared with wild-type and KI/KI mice in the open field starting at 7 months. The KI/KI and KI/KO mice both have significantly smaller muscle fiber cross-sectional area. However, only the KI/KO mice have clear skeletal muscle histologic changes in MFM. As in patient muscle, there are increased levels of BAG3-interacting proteins, such as p62, heat shock protein B8, and αB-crystallin. The KI/KO mP215L strain is the first murine model of BAG3 myopathy that resembles the human skeletal muscle pathologic features. The results support the hypothesis that the pathologic development of MFM requires a significant decrease in BAG3 protein level and not only a gain of function caused by the dominant missense mutation.
Collapse
Affiliation(s)
- Rebecca Robertson
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Talita C Conte
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Marie-Josée Dicaire
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Cone Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Abbas F Sadikot
- Cone Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | - Josée N Lavoie
- Centre de Recherche sur le Cancer, l'Université Laval, Québec, Quebec, Canada; Oncology Axis, Centre de Recherche du Centre Hospitalier Universitaire (CHU), Québec-Université Laval, Québec, Quebec, Canada; Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, l'Université Laval, Québec, Quebec, Canada
| | - Erin O'Ferrall
- Rare Neurological Diseases Group, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jason C Young
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Bernard Brais
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
26
|
A Novel DES L115F Mutation Identified by Whole Exome Sequencing is Associated with Inherited Cardiac Conduction Disease. Int J Mol Sci 2019; 20:ijms20246227. [PMID: 31835587 PMCID: PMC6940838 DOI: 10.3390/ijms20246227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/03/2019] [Accepted: 12/08/2019] [Indexed: 12/15/2022] Open
Abstract
Inherited cardiac conduction disease (CCD) is rare; it is caused by a large number of mutations in genes encoding cardiac ion channels and cytoskeletal proteins. Recently, whole-exome sequencing has been successfully used to identify causal mutations for rare monogenic Mendelian diseases. We used trio-based whole-exome sequencing to study a Chinese family with multiple family members affected by CCD, and identified a heterozygous missense mutation (c.343C>T, p.Leu115Phe) in the desmin (DES) gene as the most likely candidate causal mutation for the development of CCD in this family. The mutation is novel and is predicted to affect the conformation of the coiled-coil rod domain of DES according to structural model prediction. Its pathogenicity in desmin protein aggregation was further confirmed by expressing the mutation, both in a cellular model and a CRISPR/CAS9 knock-in mouse model. In conclusion, our results suggest that whole-exome sequencing is a feasible approach to identify candidate genes underlying inherited conduction diseases.
Collapse
|
27
|
Ciryam P, Antalek M, Cid F, Tartaglia GG, Dobson CM, Guettsches AK, Eggers B, Vorgerd M, Marcus K, Kley RA, Morimoto RI, Vendruscolo M, Weihl CC. A metastable subproteome underlies inclusion formation in muscle proteinopathies. Acta Neuropathol Commun 2019; 7:197. [PMID: 31796104 PMCID: PMC6891963 DOI: 10.1186/s40478-019-0853-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 11/21/2019] [Indexed: 01/20/2023] Open
Abstract
Protein aggregation is a pathological feature of neurodegenerative disorders. We previously demonstrated that protein inclusions in the brain are composed of supersaturated proteins, which are abundant and aggregation-prone, and form a metastable subproteome. It is not yet clear, however, whether this phenomenon is also associated with non-neuronal protein conformational disorders. To respond to this question, we analyzed proteomic datasets from biopsies of patients with genetic and acquired protein aggregate myopathy (PAM) by quantifying the changes in composition, concentration and aggregation propensity of proteins in the fibers containing inclusions and those surrounding them. We found that a metastable subproteome is present in skeletal muscle from healthy patients. The expression of this subproteome escalate as proteomic samples are taken more proximal to the pathologic inclusion, eventually exceeding its solubility limits and aggregating. While most supersaturated proteins decrease or maintain steady abundance across healthy fibers and inclusion-containing fibers, proteins within the metastable subproteome rise in abundance, suggesting that they escape regulation. Taken together, our results show in the context of a human conformational disorder that the supersaturation of a metastable subproteome underlies widespread aggregation and correlates with the histopathological state of the tissue.
Collapse
Affiliation(s)
- Prajwal Ciryam
- Department of Neurology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Matthew Antalek
- Rice Institute for Biomedical Research, Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Fernando Cid
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Gian Gaetano Tartaglia
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Anne-Katrin Guettsches
- Department of Neurology, Heimer Institute of Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Britta Eggers
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute of Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Rudolf A Kley
- Department of Neurology, St. Marien Hospital Borken, University of Witten/Herdecke, Borken, Germany
| | - Richard I Morimoto
- Rice Institute for Biomedical Research, Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Conrad C Weihl
- Department of Neurology and Hope Center for Neurological Disease, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
28
|
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, Milting H. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. Int J Mol Sci 2019; 20:ijms20184381. [PMID: 31489928 PMCID: PMC6770343 DOI: 10.3390/ijms20184381] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, many pathogenic or likely pathogenic genetic mutations in over hundred different genes have been described for non-ischemic, genetic cardiomyopathies. However, the functional knowledge about most of these mutations is still limited because the generation of adequate animal models is time-consuming and challenging. Therefore, human induced pluripotent stem cells (iPSCs) carrying specific cardiomyopathy-associated mutations are a promising alternative. Since the original discovery that pluripotency can be artificially induced by the expression of different transcription factors, various patient-specific-induced pluripotent stem cell lines have been generated to model non-ischemic, genetic cardiomyopathies in vitro. In this review, we describe the genetic landscape of non-ischemic, genetic cardiomyopathies and give an overview about different human iPSC lines, which have been developed for the disease modeling of inherited cardiomyopathies. We summarize different methods and protocols for the general differentiation of human iPSCs into cardiomyocytes. In addition, we describe methods and technologies to investigate functionally human iPSC-derived cardiomyocytes. Furthermore, we summarize novel genome editing approaches for the genetic manipulation of human iPSCs. This review provides an overview about the genetic landscape of inherited cardiomyopathies with a focus on iPSC technology, which might be of interest for clinicians and basic scientists interested in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hans Ebbinghaus
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Jan Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Sandra Ratnavadivel
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| |
Collapse
|
29
|
Desmin forms toxic, seeding-competent amyloid aggregates that persist in muscle fibers. Proc Natl Acad Sci U S A 2019; 116:16835-16840. [PMID: 31371504 PMCID: PMC6708308 DOI: 10.1073/pnas.1908263116] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Protein aggregation and the deposition of amyloid is a common feature in neurodegeneration, but can also be seen in degenerative muscle diseases known as myofibrillar myopathies (MFMs). Hallmark pathology in MFM patient muscle is myofibrillar disarray, aggregation of the muscle-specific intermediate filament, desmin, and amyloid. In some cases, a missense mutation in desmin leads to its destabilization and aggregation. The present study demonstrates that similar to neurodegenerative proteins, desmin can form amyloid and template the amyloidogenic conversion of unaggregated desmin protein. This desmin-derived amyloid is toxic to myocytes and persists when introduced into skeletal muscle, in contrast to unaggregated desmin. These data demonstrate that desmin itself can form amyloid and expand the mechanism of proteinopathies to skeletal muscle. Desmin-associated myofibrillar myopathy (MFM) has pathologic similarities to neurodegeneration-associated protein aggregate diseases. Desmin is an abundant muscle-specific intermediate filament, and disease mutations lead to its aggregation in cells, animals, and patients. We reasoned that similar to neurodegeneration-associated proteins, desmin itself may form amyloid. Desmin peptides corresponding to putative amyloidogenic regions formed seeding-competent amyloid fibrils. Amyloid formation was increased when disease-associated mutations were made within the peptide, and this conversion was inhibited by the anti-amyloid compound epigallocatechin-gallate. Moreover, a purified desmin fragment (aa 117 to 348) containing both amyloidogenic regions formed amyloid fibrils under physiologic conditions. Desmin fragment-derived amyloid coaggregated with full-length desmin and was able to template its conversion into fibrils in vitro. Desmin amyloids were cytotoxic to myotubes and disrupted their myofibril organization compared with desmin monomer or other nondesmin amyloids. Finally, desmin fragment amyloid persisted when introduced into mouse skeletal muscle. These data suggest that desmin forms seeding-competent amyloid that is toxic to myofibers. Moreover, small molecules known to interfere with amyloid formation and propagation may have therapeutic potential in MFM.
Collapse
|
30
|
The MyoRobot technology discloses a premature biomechanical decay of skeletal muscle fiber bundles derived from R349P desminopathy mice. Sci Rep 2019; 9:10769. [PMID: 31341183 PMCID: PMC6656739 DOI: 10.1038/s41598-019-46723-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/03/2019] [Indexed: 01/05/2023] Open
Abstract
Mutations in the Des gene coding for the muscle-specific intermediate filament protein desmin lead to myopathies and cardiomyopathies. We previously generated a R349P desmin knock-in mouse strain as a patient-mimicking model for the corresponding most frequent human desmin mutation R350P. Since nothing is known about the age-dependent changes in the biomechanics of affected muscles, we investigated the passive and active biomechanics of small fiber bundles from young (17–23 wks), adult (25–45 wks) and aged (>60 wks) heterozygous and homozygous R349P desmin knock-in mice in comparison to wild-type littermates. We used a novel automated biomechatronics platform, the MyoRobot, to perform coherent quantitative recordings of passive (resting length-tension curves, visco-elasticity) and active (caffeine-induced force transients, pCa-force, ‘slack-tests’) parameters to determine age-dependent effects of the R349P desmin mutation in slow-twitch soleus and fast-twitch extensor digitorum longus small fiber bundles. We demonstrate that active force properties are not affected by this mutation while passive steady-state elasticity is vastly altered in R349P desmin fiber bundles compatible with a pre-aged phenotype exhibiting stiffer muscle preparations. Visco-elasticity on the other hand, was not altered. Our study represents the first systematic age-related characterization of small muscle fiber bundle preparation biomechanics in conjunction with inherited desminopathy.
Collapse
|
31
|
Sit B, Gutmann D, Iskratsch T. Costameres, dense plaques and podosomes: the cell matrix adhesions in cardiovascular mechanosensing. J Muscle Res Cell Motil 2019; 40:197-209. [PMID: 31214894 PMCID: PMC6726830 DOI: 10.1007/s10974-019-09529-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/15/2019] [Indexed: 12/12/2022]
Abstract
The stiffness of the cardiovascular environment changes during ageing and in disease and contributes to disease incidence and progression. For instance, increased arterial stiffness can lead to atherosclerosis, while stiffening of the heart due to fibrosis can increase the chances of heart failure. Cells can sense the stiffness of the extracellular matrix through integrin adhesions and other mechanosensitive structures and in response to this initiate mechanosignalling pathways that ultimately change the cellular behaviour. Over the past decades, interest in mechanobiology has steadily increased and with this also our understanding of the molecular basis of mechanosensing and transduction. However, much of our knowledge about the mechanisms is derived from studies investigating focal adhesions in non-muscle cells, which are distinct in several regards from the cell-matrix adhesions in cardiomyocytes (costameres) or vascular smooth muscle cells (dense plaques or podosomes). Therefore, we will look here first at the evidence for mechanical sensing in the cardiovascular system, before comparing the different cytoskeletal arrangements and adhesion sites in cardiomyocytes and vascular smooth muscle cells and what is known about mechanical sensing through the various structures.
Collapse
Affiliation(s)
- Brian Sit
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Daniel Gutmann
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK.
| |
Collapse
|
32
|
Rodríguez MA, Liu JX, Parkkonen K, Li Z, Pedrosa Domellöf F. The Cytoskeleton in the Extraocular Muscles of Desmin Knockout Mice. Invest Ophthalmol Vis Sci 2019; 59:4847-4855. [PMID: 30347079 DOI: 10.1167/iovs.18-24508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effect of absence of desmin on the extraocular muscles (EOMs) with focus on the structure and composition of the cytoskeleton. Methods The distribution of synemin, syncoilin, plectin, nestin, and dystrophin was evaluated on cross and longitudinal sections of EOMs and limb muscles from 1-year-old desmin knockout mice (desmin-/-) by immunofluorescence. General morphology was evaluated with hematoxylin and eosin while mitochondrial content and distribution were evaluated by succinate dehydrogenase (SDH) and modified Gomori trichrome stainings. Results The muscle fibers of the EOMs in desmin-/- mice were remarkably well preserved in contrast to those in the severely affected soleus and the slightly affected gastrocnemius muscles. There were no signs of muscular pathology in the EOMs and all cytoskeletal proteins studied showed a correct location at sarcolemma and Z-discs. However, an increase of SDH staining and mitochondrial aggregates under the sarcolemma was detected. Conclusions The structure of the EOMs was well preserved in the absence of desmin. We suggest that desmin is not necessary for correct synemin, syncoilin, plectin, and dystrophin location on the cytoskeleton of EOMs. However, it is needed to maintain an appropriate mitochondrial distribution in both EOMs and limb muscles.
Collapse
Affiliation(s)
| | - Jing-Xia Liu
- Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | - Kimmo Parkkonen
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
33
|
Asatryan B, Medeiros-Domingo A. Molecular and genetic insights into progressive cardiac conduction disease. Europace 2019; 21:1145-1158. [DOI: 10.1093/europace/euz109] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
Abstract
Progressive cardiac conduction disease (PCCD) is often a primarily genetic disorder, with clinical and genetic overlaps with other inherited cardiac and metabolic diseases. A number of genes have been implicated in PCCD pathogenesis with or without structural heart disease or systemic manifestations. Precise genetic diagnosis contributes to risk stratification, better selection of specific therapy and allows familiar cascade screening. Cardiologists should be aware of the different phenotypes emerging from different gene-mutations and the potential risk of sudden cardiac death. Genetic forms of PCCD often overlap or coexist with other inherited heart diseases or manifest in the context of multisystem syndromes. Despite the significant advances in the knowledge of the genetic architecture of PCCD and overlapping diseases, in a measurable fraction of PCCD cases, including in familial clustering of disease, investigations of known cardiac disease-associated genes fail to reveal the underlying substrate, suggesting that new causal genes are yet to be discovered. Here, we provide insight into genetics and molecular mechanisms of PCCD and related diseases. We also highlight the phenotypic overlaps of PCCD with other inherited cardiac and metabolic diseases, present unmet challenges in clinical practice, and summarize the available therapeutic options for affected patients.
Collapse
Affiliation(s)
- Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, Switzerland
| | | |
Collapse
|
34
|
Franck A, Lainé J, Moulay G, Lemerle E, Trichet M, Gentil C, Benkhelifa-Ziyyat S, Lacène E, Bui MT, Brochier G, Guicheney P, Romero N, Bitoun M, Vassilopoulos S. Clathrin plaques and associated actin anchor intermediate filaments in skeletal muscle. Mol Biol Cell 2019; 30:579-590. [PMID: 30601711 PMCID: PMC6589689 DOI: 10.1091/mbc.e18-11-0718] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clathrin plaques are stable features of the plasma membrane observed in several cell types. They are abundant in muscle, where they localize at costameres that link the contractile apparatus to the sarcolemma and connect the sarcolemma to the basal lamina. Here, we show that clathrin plaques and surrounding branched actin filaments form microdomains that anchor a three-dimensional desmin intermediate filament (IF) web. Depletion of clathrin plaque and branched actin components causes accumulation of desmin tangles in the cytoplasm. We show that dynamin 2, whose mutations cause centronuclear myopathy (CNM), regulates both clathrin plaques and surrounding branched actin filaments, while CNM-causing mutations lead to desmin disorganization in a CNM mouse model and patient biopsies. Our results suggest a novel paradigm in cell biology, wherein clathrin plaques act as platforms capable of recruiting branched cortical actin, which in turn anchors IFs, both essential for striated muscle formation and function.
Collapse
Affiliation(s)
- Agathe Franck
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Jeanne Lainé
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France.,Department of Physiology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Gilles Moulay
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Eline Lemerle
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Michaël Trichet
- Institut de Biologie Paris-Seine, Sorbonne Université, CNRS, FR3631 Paris, France
| | - Christel Gentil
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Sofia Benkhelifa-Ziyyat
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Emmanuelle Lacène
- Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Mai Thao Bui
- Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Guy Brochier
- Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Pascale Guicheney
- Institute of Cardiometabolism and Nutrition, UMRS 1166, INSERM , Sorbonne Université, F-75013 Paris, France
| | - Norma Romero
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France.,Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Marc Bitoun
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Stéphane Vassilopoulos
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| |
Collapse
|
35
|
Li L, Zhang Q, Zhang X, Zhang J, Wang X, Ren J, Jia J, Zhang D, Jiang X, Zhang J, Mei H, Chen B, Hu J, Huang Y. Microtubule associated protein 4 phosphorylation leads to pathological cardiac remodeling in mice. EBioMedicine 2018; 37:221-235. [PMID: 30327268 PMCID: PMC6286641 DOI: 10.1016/j.ebiom.2018.10.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/23/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022] Open
Abstract
Background Cardiac remodeling is a pathophysiological process that involves various changes in heart, including cardiac hypertrophy and fibrosis. Cardiac remodeling following pathological stimuli is common trigger leading to cardiac maladaptation and onset of heart failure, and their pathogenesis remains unclear. Methods Heart specimens of tetralogy of Fallot (TOF) patients, myocardial infarction (MI) and transverse aortic constriction (TAC) mouse models were collected to determine changes of microtubule associated protein 4 (MAP4) phosphorylation. MAP4 (S667A, S737E and S760E) knock in (MAP4 KI) mouse and cultured neonatal mouse cardiomyocytes or fibroblasts were used to investigate changes of cardiac phenotypes and possible mechanisms with a variety of approaches, including functional, histocytological and pathological observations. Findings Elevated cardiac phosphorylation of MAP4 (S737 and S760) was observed in TOF patients, MI and TAC mouse models. In MAP4 KI mice, age-dependent cardiac phenotypes, including cardiac hypertrophy, fibrosis, diastolic and systolic dysfunction were observed. In addition, increased cardiomyocyte apoptosis together with microtubule disassembly and mitochondrial translocation of phosphorylated MAP4 was detected prior to the onset of cardiac remodeling, and p38/MAPK was demonstrated to be the possible signaling pathway that mediated MAP4 (S737 and S760) phosphorylation. Interpretation Our data reveal for the first time that MAP4 drives pathological cardiac remodeling through its phosphorylation. These findings bear the therapeutic potential to ameliorate pathological cardiac remodeling by attenuating MAP4 phosphorylation. Fund This work was supported by the Key Program of National Natural Science Foundation of China (No.81430042) and National Natural Science Foundation of China (No.81671913).
Collapse
Affiliation(s)
- Lingfei Li
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xingyue Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Junhui Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuefeng Wang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA
| | - Jiezhi Jia
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xupin Jiang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiaping Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Department of Plastic Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Mei
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, CT, USA
| | - Bing Chen
- Endocrinology Department, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiongyu Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Endocrinology Department, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Yuesheng Huang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
36
|
Winter L, Unger A, Berwanger C, Spörrer M, Türk M, Chevessier F, Strucksberg KH, Schlötzer-Schrehardt U, Wittig I, Goldmann WH, Marcus K, Linke WA, Clemen CS, Schröder R. Imbalances in protein homeostasis caused by mutant desmin. Neuropathol Appl Neurobiol 2018; 45:476-494. [PMID: 30179276 DOI: 10.1111/nan.12516] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/17/2018] [Indexed: 12/19/2022]
Abstract
AIMS We investigated newly generated immortalized heterozygous and homozygous R349P desmin knock-in myoblasts in conjunction with the corresponding desminopathy mice as models for desminopathies to analyse major protein quality control processes in response to the presence of R349P mutant desmin. METHODS We used hetero- and homozygous R349P desmin knock-in mice for analyses and for crossbreeding with p53 knock-out mice to generate immortalized R349P desmin knock-in skeletal muscle myoblasts and myotubes. Skeletal muscle sections and cultured muscle cells were investigated by indirect immunofluorescence microscopy, proteasomal activity measurements and immunoblotting addressing autophagy rate, chaperone-assisted selective autophagy and heat shock protein levels. Muscle sections were further analysed by transmission and immunogold electron microscopy. RESULTS We demonstrate that mutant desmin (i) increases proteasomal activity, (ii) stimulates macroautophagy, (iii) dysregulates the chaperone assisted selective autophagy and (iv) elevates the protein levels of αB-crystallin and Hsp27. Both αB-crystallin and Hsp27 as well as Hsp90 displayed translocation patterns from Z-discs as well as Z-I junctions, respectively, to the level of sarcomeric I-bands in dominant and recessive desminopathies. CONCLUSIONS Our findings demonstrate that the presence of R349P mutant desmin causes a general imbalance in skeletal muscle protein homeostasis via aberrant activity of all major protein quality control systems. The augmented activity of these systems and the subcellular shift of essential heat shock proteins may deleteriously contribute to the previously observed increased turnover of desmin itself and desmin-binding partners, which triggers progressive dysfunction of the extrasarcomeric cytoskeleton and the myofibrillar apparatus in the course of the development of desminopathies.
Collapse
Affiliation(s)
- L Winter
- Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany.,Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - A Unger
- Department of Cardiovascular Physiology, Ruhr-University Bochum, Bochum, Germany.,Institute for Genetics of Heart Diseases, University Hospital Münster, Münster, Germany
| | - C Berwanger
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany.,Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - M Spörrer
- Center for Medical Physics and Technology, Biophysics Group, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - M Türk
- Department of Neurology, University Hospital Erlangen, Erlangen, Germany
| | - F Chevessier
- Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - K-H Strucksberg
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | | | - I Wittig
- Functional Proteomics, SFB815 Core Unit, Medical School, Goethe University, Frankfurt, Germany
| | - W H Goldmann
- Center for Medical Physics and Technology, Biophysics Group, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - K Marcus
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - W A Linke
- Department of Cardiovascular Physiology, Ruhr-University Bochum, Bochum, Germany.,Institute of Physiology II, University of Münster, Münster, Germany
| | - C S Clemen
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany.,Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - R Schröder
- Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
37
|
Clemen CS, Winter L, Strucksberg KH, Berwanger C, Türk M, Kornblum C, Florin A, Aguilar-Pimentel JA, Amarie OV, Becker L, Garrett L, Hans W, Moreth K, Neff F, Pingen L, Rathkolb B, Rácz I, Rozman J, Treise I, Fuchs H, Gailus-Durner V, de Angelis MH, Vorgerd M, Eichinger L, Schröder R. The heterozygous R155C VCP mutation: Toxic in humans! Harmless in mice? Biochem Biophys Res Commun 2018; 503:2770-2777. [PMID: 30100055 DOI: 10.1016/j.bbrc.2018.08.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/04/2018] [Indexed: 12/12/2022]
Abstract
Heterozygous missense mutations in the human VCP gene cause inclusion body myopathy associated with Paget disease of bone and fronto-temporal dementia (IBMPFD) and amyotrophic lateral sclerosis (ALS). The exact molecular mechanisms by which VCP mutations cause disease manifestation in different tissues are incompletely understood. In the present study, we report the comprehensive analysis of a newly generated R155C VCP knock-in mouse model, which expresses the ortholog of the second most frequently occurring human pathogenic VCP mutation. Heterozygous R155C VCP knock-in mice showed decreased plasma lactate, serum albumin and total protein concentrations, platelet numbers, and liver to body weight ratios, and increased oxygen consumption and CD8+/Ly6C + T-cell fractions, but none of the typical human IBMPFD or ALS pathologies. Breeding of heterozygous mice did not yield in the generation of homozygous R155C VCP knock-in animals. Immunoblotting showed identical total VCP protein levels in human IBMPFD and murine R155C VCP knock-in tissues as compared to wild-type controls. However, while in human IBMPFD skeletal muscle tissue 70% of the total VCP mRNA was derived from the mutant allele, in R155C VCP knock-in mice only 5% and 7% mutant mRNA were detected in skeletal muscle and brain tissue, respectively. The lack of any obvious IBMPFD or ALS pathology could thus be a consequence of the very low expression of mutant VCP. We conclude that the increased and decreased fractions of the R155C mutant VCP mRNA in man and mice, respectively, are due to missense mutation-induced, divergent alterations in the biological half-life of the human and murine mutant mRNAs. Furthermore, our work suggests that therapy approaches lowering the expression of the mutant VCP mRNA below a critical threshold may ameliorate the intrinsic disease pathology.
Collapse
Affiliation(s)
- Christoph S Clemen
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789, Bochum, Germany; Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany.
| | - Lilli Winter
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany; Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, 1090, Vienna, Austria
| | - Karl-Heinz Strucksberg
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Carolin Berwanger
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789, Bochum, Germany; Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Matthias Türk
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Cornelia Kornblum
- Department of Neurology, University Hospital Bonn, 53125, Bonn, Germany; Center for Rare Diseases Bonn, University Hospital Bonn, 53127, Bonn, Germany
| | - Alexandra Florin
- Institute for Pathology, University Hospital Cologne, 50937, Cologne, Germany
| | - Juan Antonio Aguilar-Pimentel
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Oana Veronica Amarie
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Kristin Moreth
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Frauke Neff
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Laura Pingen
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, 81377, Munich, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Ildikó Rácz
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127, Bonn, Germany; Clinic of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, 53127, Bonn, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Irina Treise
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, 85354, Freising, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789, Bochum, Germany
| | - Ludwig Eichinger
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany.
| |
Collapse
|
38
|
Abstract
Intermediate filaments (IFs) are one of the three major elements of the cytoskeleton. Their stability, intrinsic mechanical properties, and cell type-specific expression patterns distinguish them from actin and microtubules. By providing mechanical support, IFs protect cells from external forces and participate in cell adhesion and tissue integrity. IFs form an extensive and elaborate network that connects the cell cortex to intracellular organelles. They act as a molecular scaffold that controls intracellular organization. However, IFs have been revealed as much more than just rigid structures. Their dynamics is regulated by multiple signaling cascades and appears to contribute to signaling events in response to cell stress and to dynamic cellular functions such as mitosis, apoptosis, and migration.
Collapse
Affiliation(s)
- Sandrine Etienne-Manneville
- Institut Pasteur Paris, CNRS UMR 3691, Cell Polarity, Migration and Cancer Unit, Equipe Labellisée Ligue Contre le Cancer, Paris Cedex 15, France;
| |
Collapse
|
39
|
Tsikitis M, Galata Z, Mavroidis M, Psarras S, Capetanaki Y. Intermediate filaments in cardiomyopathy. Biophys Rev 2018; 10:1007-1031. [PMID: 30027462 DOI: 10.1007/s12551-018-0443-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/05/2018] [Indexed: 12/20/2022] Open
Abstract
Intermediate filament (IF) proteins are critical regulators in health and disease. The discovery of hundreds of mutations in IF genes and posttranslational modifications has been linked to a plethora of human diseases, including, among others, cardiomyopathies, muscular dystrophies, progeria, blistering diseases of the epidermis, and neurodegenerative diseases. The major IF proteins that have been linked to cardiomyopathies and heart failure are the muscle-specific cytoskeletal IF protein desmin and the nuclear IF protein lamin, as a subgroup of the known desminopathies and laminopathies, respectively. The studies so far, both with healthy and diseased heart, have demonstrated the importance of these IF protein networks in intracellular and intercellular integration of structure and function, mechanotransduction and gene activation, cardiomyocyte differentiation and survival, mitochondrial homeostasis, and regulation of metabolism. The high coordination of all these processes is obviously of great importance for the maintenance of proper, life-lasting, and continuous contraction of this highly organized cardiac striated muscle and consequently a healthy heart. In this review, we will cover most known information on the role of IFs in the above processes and how their deficiency or disruption leads to cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527, Athens, Greece
| | - Zoi Galata
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527, Athens, Greece
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527, Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527, Athens, Greece
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527, Athens, Greece.
| |
Collapse
|
40
|
Brodehl A, Gaertner-Rommel A, Milting H. Molecular insights into cardiomyopathies associated with desmin (DES) mutations. Biophys Rev 2018; 10:983-1006. [PMID: 29926427 DOI: 10.1007/s12551-018-0429-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Increasing usage of next-generation sequencing techniques pushed during the last decade cardiogenetic diagnostics leading to the identification of a huge number of genetic variants in about 170 genes associated with cardiomyopathies, channelopathies, or syndromes with cardiac involvement. Because of the biochemical and cellular complexity, it is challenging to understand the clinical meaning or even the relevant pathomechanisms of the majority of genetic sequence variants. However, detailed knowledge about the associated molecular pathomechanism is essential for the development of efficient therapeutic strategies in future and genetic counseling. Mutations in DES, encoding the muscle-specific intermediate filament protein desmin, have been identified in different kinds of cardiac and skeletal myopathies. Here, we review the functions of desmin in health and disease with a focus on cardiomyopathies. In addition, we will summarize the genetic and clinical literature about DES mutations and will explain relevant cell and animal models. Moreover, we discuss upcoming perspectives and consequences of novel experimental approaches like genome editing technology, which might open a novel research field contributing to the development of efficient and mutation-specific treatment options.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany.
| | - Anna Gaertner-Rommel
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development, Heart and Diabetes Centre NRW, Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany.
| |
Collapse
|
41
|
Goldmann WH. Intermediate filaments and cellular mechanics. Cell Biol Int 2018; 42:132-138. [PMID: 28980752 DOI: 10.1002/cbin.10879] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/30/2017] [Indexed: 12/16/2022]
Abstract
Intermediate filaments (IFs) are one of the three types of cytoskeletal polymers that resist tensile and compressive forces in cells. They crosslink each other as well as with actin filaments and microtubules by proteins, which include desmin, filamin C, plectin, and lamin (A/C). Mutations in these proteins can lead to a wide range of pathologies, some of which exhibit mechanical failure of the skin, skeletal, or heart muscle.
Collapse
Affiliation(s)
- Wolfgang H Goldmann
- Department of Physics, Biophysics Group, Friedrich-Alexander-University Erlangen-Nuremberg, D-91052 Erlangen, Germany
| |
Collapse
|
42
|
Diermeier S, Buttgereit A, Schürmann S, Winter L, Xu H, Murphy RM, Clemen CS, Schröder R, Friedrich O. Preaged remodeling of myofibrillar cytoarchitecture in skeletal muscle expressing R349P mutant desmin. Neurobiol Aging 2017; 58:77-87. [PMID: 28715662 DOI: 10.1016/j.neurobiolaging.2017.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/14/2022]
Abstract
The majority of hereditary and acquired myopathies are clinically characterized by progressive muscle weakness. We hypothesized that ongoing derangement of skeletal muscle cytoarchitecture at the single fiber level may precede and be responsible for the progressive muscle weakness. Here, we analyzed the effects of aging in wild-type (wt) and heterozygous (het) and homozygous (hom) R349P desmin knock-in mice. The latter harbor the ortholog of the most frequently encountered human R350P desmin missense mutation. We quantitatively analyzed the subcellular cytoarchitecture of fast- and slow-twitch muscles from young, intermediate, and aged wt as well as desminopathy mice. We recorded multiphoton second harmonic generation and nuclear fluorescence signals in single muscle fibers to compare aging-related effects in all genotypes. The analysis of wt mice revealed that the myofibrillar cytoarchitecture remained stable with aging in fast-twitch muscles, whereas slow-twitch muscle fibers displayed structural derangements during aging. In contrast, the myofibrillar cytoarchitecture and nuclear density were severely compromised in fast- and slow-twitch muscle fibers of hom R349P desmin mice at all ages. Het mice only showed a clear degradation in their fiber structure in fast-twitch muscles from the adult to the presenescent age bin. Our study documents distinct signs of normal and R349P mutant desmin-related remodeling of the 3D myofibrillar architecture during aging, which provides a structural basis for the progressive muscle weakness.
Collapse
Affiliation(s)
- Stefanie Diermeier
- Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; SAOT, Erlangen Graduate School in Advanced Optical Technologies, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Buttgereit
- Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Schürmann
- Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; SAOT, Erlangen Graduate School in Advanced Optical Technologies, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Lilli Winter
- Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Hongyang Xu
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Australia
| | - Christoph S Clemen
- Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany; Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany; Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; SAOT, Erlangen Graduate School in Advanced Optical Technologies, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; Muscle Research Center Erlangen (MURCE), Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
43
|
Early signs of architectural and biomechanical failure in isolated myofibers and immortalized myoblasts from desmin-mutant knock-in mice. Sci Rep 2017; 7:1391. [PMID: 28469177 PMCID: PMC5431221 DOI: 10.1038/s41598-017-01485-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/29/2017] [Indexed: 01/25/2023] Open
Abstract
In striated muscle, desmin intermediate filaments interlink the contractile myofibrillar apparatus with mitochondria, nuclei, and the sarcolemma. The desmin network’s pivotal role in myocytes is evident since mutations in the human desmin gene cause severe myopathies and cardiomyopathies. Here, we investigated skeletal muscle pathology in myofibers and myofibrils isolated from young hetero- and homozygous R349P desmin knock-in mice, which carry the orthologue of the most frequent human desmin missense mutation R350P. We demonstrate that mutant desmin alters myofibrillar cytoarchitecture, markedly disrupts the lateral sarcomere lattice and distorts myofibrillar angular axial orientation. Biomechanical assessment revealed a high predisposition to stretch-induced damage in fiber bundles of R349P mice. Notably, Ca2+-sensitivity and passive myofibrillar tension were decreased in heterozygous fiber bundles, but increased in homozygous fiber bundles compared to wildtype mice. In a parallel approach, we generated and subsequently subjected immortalized heterozygous R349P desmin knock-in myoblasts to magnetic tweezer experiments that revealed a significantly increased sarcolemmal lateral stiffness. Our data suggest that mutated desmin already markedly impedes myocyte structure and function at pre-symptomatic stages of myofibrillar myopathies.
Collapse
|
44
|
Batonnet-Pichon S, Behin A, Cabet E, Delort F, Vicart P, Lilienbaum A. Myofibrillar Myopathies: New Perspectives from Animal Models to Potential Therapeutic Approaches. J Neuromuscul Dis 2017; 4:1-15. [PMID: 28269794 PMCID: PMC5345645 DOI: 10.3233/jnd-160203] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Myofibrillar myopathies (MFMs) are muscular disorders involving proteins that play a role in the structure, maintenance processes and protein quality control mechanisms closely related to the Z-disc in the muscular fibers. MFMs share common histological characteristics including progressive disorganization of the interfibrillar network and protein aggregation. Currently no treatment is available. In this review, we describe first clinical symptoms associated with mutations of the six genes (DES, CRYAB, MYOT, ZASP, FLNC and BAG3) primary involved in MFM and defining the origin of this pathology. As mechanisms determining the aetiology of the disease remain unclear yet, several research teams have developed animal models from invertebrates to mammalians species. Thus we describe here these different models that often recapitulate human clinical symptoms. Therefore they are very useful for deeper studies to understand early molecular and progressive mechanisms determining the pathology. Finally in the last part, we emphasize on the potential therapeutic approaches for MFM that could be conducted in the future. In conclusion, this review offers a link from patients to future therapy through the use of MFMs animal models.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Drosophila
- Humans
- Mice
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Mutation
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/pathology
- Myopathies, Structural, Congenital/physiopathology
- Myopathies, Structural, Congenital/therapy
- Oryzias
Collapse
Affiliation(s)
- Sabrina Batonnet-Pichon
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR, Paris, France
| | - Anthony Behin
- Centre de Référence de Pathologie Neuromusculaire Paris-Est, groupe hospitalier Pitié-Salpêtrière, institut de Myologie, AP-HP, boulevard de l’Hôpital, Paris cedex 13, France
| | - Eva Cabet
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR, Paris, France
| | - Florence Delort
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR, Paris, France
| | - Patrick Vicart
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR, Paris, France
| | - Alain Lilienbaum
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR, Paris, France
| |
Collapse
|
45
|
Abstract
Proteins of the intermediate filament (IF) supergene family are ubiquitous structural components that comprise, in a cell type-specific manner, the cytoskeleton proper in animal tissues. All IF proteins show a distinctly organized, extended α-helical conformation prone to form two-stranded coiled coils, which are the basic building blocks of these highly flexible, stress-resistant cytoskeletal filaments. IF proteins are highly charged, thus representing versatile polyampholytes with multiple functions. Taking vimentin, keratins, and the nuclear lamins as our prime examples, we present an overview of their molecular and structural parameters. These, in turn, document the ability of IF proteins to form distinct, highly diverse supramolecular assemblies and biomaterials found, for example, at the inner nuclear membrane, throughout the cytoplasm, and in highly complex extracellular appendages, such as hair and nails, of vertebrate organisms. Ultimately, our aim is to set the stage for a more rational understanding of the immediate effects that missense mutations in IF genes have on cellular functions and for their far-reaching impact on the development of the numerous IF diseases caused by them.
Collapse
Affiliation(s)
- Harald Herrmann
- Functional Architecture of the Cell (B065), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany, and Institute of Neuropathology, University Hospital Erlangen, D-91054 Erlangen, Germany
| | - Ueli Aebi
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| |
Collapse
|
46
|
|
47
|
Mutant desmin substantially perturbs mitochondrial morphology, function and maintenance in skeletal muscle tissue. Acta Neuropathol 2016; 132:453-73. [PMID: 27393313 PMCID: PMC4992032 DOI: 10.1007/s00401-016-1592-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/18/2022]
Abstract
Secondary mitochondrial dysfunction is a feature in a wide variety of human protein aggregate diseases caused by mutations in different proteins, both in the central nervous system and in striated muscle. The functional relationship between the expression of a mutated protein and mitochondrial dysfunction is largely unknown. In particular, the mechanism how this dysfunction drives the disease process is still elusive. To address this issue for protein aggregate myopathies, we performed a comprehensive, multi-level analysis of mitochondrial pathology in skeletal muscles of human patients with mutations in the intermediate filament protein desmin and in muscles of hetero- and homozygous knock-in mice carrying the R349P desmin mutation. We demonstrate that the expression of mutant desmin causes disruption of the extrasarcomeric desmin cytoskeleton and extensive mitochondrial abnormalities regarding subcellular distribution, number and shape. At the molecular level, we uncovered changes in the abundancy and assembly of the respiratory chain complexes and supercomplexes. In addition, we revealed a marked reduction of mtDNA- and nuclear DNA-encoded mitochondrial proteins in parallel with large-scale deletions in mtDNA and reduced mtDNA copy numbers. Hence, our data demonstrate that the expression of mutant desmin causes multi-level damage of mitochondria already in early stages of desminopathies.
Collapse
|
48
|
Durmuş H, Ayhan Ö, Çırak S, Deymeer F, Parman Y, Franke A, Eiber N, Chevessier F, Schlötzer-Schrehardt U, Clemen CS, Hashemolhosseini S, Schröder R, Hemmrich-Stanisak G, Tolun A, Serdaroğlu-Oflazer P. Neuromuscular endplate pathology in recessive desminopathies: Lessons from man and mice. Neurology 2016; 87:799-805. [PMID: 27440146 DOI: 10.1212/wnl.0000000000003004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 05/17/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the clinical, genetic, and myopathologic findings in 2 cousins with lack of desmin, the response to salbutamol in one patient, and the neuromuscular endplate pathology in a knock-in mouse model for recessive desminopathy. METHODS We performed clinical investigations in the patients, genetic studies for linkage mapping, exome sequencing, and qPCR for transcript quantification, assessment of efficacy of (3-month oral) salbutamol administration by muscle strength assessment, 6-minute walking test (6MWT), and forced vital capacity, analysis of neuromuscular endplate pathology in a homozygous R349P desmin knock-in mouse by immunofluorescence staining of the hind limb muscles, and quantitative 3D morphometry and expression studies of acetylcholine receptor genes by quantitative PCR. RESULTS Both patients had infantile-onset weakness and fatigability, facial weakness with bilateral ptosis and ophthalmoparesis, generalized muscle weakness, and a decremental response over 10% on repetitive nerve stimulation. Salbutamol improved 6MWT and subjective motor function in the treated patient. Genetic analysis revealed previously unreported novel homozygous truncating desmin mutation c.345dupC leading to protein truncation and consequent fast degradation of the mutant mRNA. In the recessive desminopathy mouse with low expression of the mutant desmin protein, we demonstrated fragmented motor endplates with increased surface areas, volumes, and fluorescence intensities in conjunction with increased α and γ acetylcholine receptor subunit expression in oxidative soleus muscle. CONCLUSIONS The patients were desmin-null and had myopathy, cardiomyopathy, and a congenital myasthenic syndrome. The data from man and mouse demonstrate that the complete lack as well as the markedly decreased expression of mutant R349P desmin impair the structural and functional integrity of neuromuscular endplates.
Collapse
Affiliation(s)
- Hacer Durmuş
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany.
| | - Özgecan Ayhan
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Sebahattin Çırak
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Feza Deymeer
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Yeşim Parman
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Andre Franke
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Nane Eiber
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Frederic Chevessier
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Ursula Schlötzer-Schrehardt
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Christoph S Clemen
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Said Hashemolhosseini
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Rolf Schröder
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Georg Hemmrich-Stanisak
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Aslıhan Tolun
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| | - Piraye Serdaroğlu-Oflazer
- From the Department of Neurology (H.D., F.D., Y.P., P.S.-O.), Faculty of Medicine, Istanbul University; Department of Molecular Biology and Genetics (Ö.A., A.T.), Boğaziçi University, Istanbul, Turkey; Children's National Medical Center (S.Ç.), Research Center for Genetic Medicine, Washington, DC; Department of Pediatrics, Institute for Human Genetics, and Center for Molecular Medicine, University Hospital Cologne; Institute of Clinical Molecular Biology (A.F., G.H.-S.), Christian-Albrechts-University of Kiel; Institute of Biochemistry (N.E., S.H.), Institute of Neuropathology (F.C., R.S.), and Department of Ophthalmology (U.S.-S.), Friedrich-Alexander-University of Erlangen-Nuremberg; and Center for Biochemistry, Institute of Biochemistry I, Medical Faculty (C.S.C.), University of Cologne, Germany
| |
Collapse
|
49
|
Bang ML. Animal Models of Congenital Cardiomyopathies Associated With Mutations in Z-Line Proteins. J Cell Physiol 2016; 232:38-52. [PMID: 27171814 DOI: 10.1002/jcp.25424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/10/2016] [Indexed: 01/15/2023]
Abstract
The cardiac Z-line at the boundary between sarcomeres is a multiprotein complex connecting the contractile apparatus with the cytoskeleton and the extracellular matrix. The Z-line is important for efficient force generation and transmission as well as the maintenance of structural stability and integrity. Furthermore, it is a nodal point for intracellular signaling, in particular mechanosensing and mechanotransduction. Mutations in various genes encoding Z-line proteins have been associated with different cardiomyopathies, including dilated cardiomyopathy, hypertrophic cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, restrictive cardiomyopathy, and left ventricular noncompaction, and mutations even within the same gene can cause widely different pathologies. Animal models have contributed to a great advancement in the understanding of the physiological function of Z-line proteins and the pathways leading from mutations in Z-line proteins to cardiomyopathy, although genotype-phenotype prediction remains a great challenge. This review presents an overview of the currently available animal models for Z-line and Z-line associated proteins involved in human cardiomyopathies with special emphasis on knock-in and transgenic mouse models recapitulating the clinical phenotypes of human cardiomyopathy patients carrying mutations in Z-line proteins. Pros and cons of mouse models will be discussed and a future outlook will be given. J. Cell. Physiol. 232: 38-52, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council and Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
50
|
Bengoechea R, Pittman SK, Tuck EP, True HL, Weihl CC. Myofibrillar disruption and RNA-binding protein aggregation in a mouse model of limb-girdle muscular dystrophy 1D. Hum Mol Genet 2015; 24:6588-602. [PMID: 26362252 PMCID: PMC4634370 DOI: 10.1093/hmg/ddv363] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022] Open
Abstract
Limb-girdle muscular dystrophy type 1D (LGMD1D) is caused by dominantly inherited missense mutations in DNAJB6, an Hsp40 co-chaperone. LGMD1D muscle has rimmed vacuoles and inclusion bodies containing DNAJB6, Z-disc proteins and TDP-43. DNAJB6 is expressed as two isoforms; DNAJB6a and DNAJB6b. Both isoforms contain LGMD1D mutant residues and are expressed in human muscle. To identify which mutant isoform confers disease pathogenesis and generate a mouse model of LGMD1D, we evaluated DNAJB6 expression and localization in skeletal muscle as well as generating DNAJB6 isoform specific expressing transgenic mice. DNAJB6a localized to myonuclei while DNAJB6b was sarcoplasmic. LGMD1D mutations in DNAJB6a or DNAJB6b did not alter this localization in mouse muscle. Transgenic mice expressing the LGMD1D mutant, F93L, in DNAJB6b under a muscle-specific promoter became weak, had early lethality and developed muscle pathology consistent with myopathy after 2 months; whereas mice expressing the same F93L mutation in DNAJB6a or overexpressing DNAJB6a or DNAJB6b wild-type transgenes remained unaffected after 1 year. DNAJB6b localized to the Z-disc and DNAJB6b-F93L expressing mouse muscle had myofibrillar disorganization and desmin inclusions. Consistent with DNAJB6 dysfunction, keratin 8/18, a DNAJB6 client also accumulated in DNAJB6b-F93L expressing mouse muscle. The RNA-binding proteins hnRNPA1 and hnRNPA2/B1 accumulated and co-localized with DNAJB6 at sarcoplasmic stress granules suggesting that these proteins maybe novel DNAJB6b clients. Similarly, hnRNPA1 and hnRNPA2/B1 formed sarcoplasmic aggregates in patients with LGMD1D. Our data support that LGMD1D mutations in DNAJB6 disrupt its sarcoplasmic function suggesting a role for DNAJB6b in Z-disc organization and stress granule kinetics.
Collapse
Affiliation(s)
| | | | | | - Heather L True
- Department of Cell Biology and Physiology and and The Hope Center for Neurological Diseases, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Conrad C Weihl
- Department of Neurology, The Hope Center for Neurological Diseases, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|