1
|
Sevcikova Tomaskova Z, Mackova K. From function to structure: how myofibrillogenesis influences the transverse-axial tubular system development and its peculiarities. Front Physiol 2025; 16:1576133. [PMID: 40352140 PMCID: PMC12062141 DOI: 10.3389/fphys.2025.1576133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/21/2025] [Indexed: 05/14/2025] Open
Abstract
The transverse-axial tubular system (TATS) is the extension of sarcolemma growing to the cell interior, providing sufficient calcium signaling to induce calcium release from sarcoplasmic reticulum cisternae and stimulate the contraction of neighboring myofibrils. Interestingly, the development of TATS is delayed and matures during the post-partum period. It starts with small invaginations near the sarcolemma, proceeding to grow an irregular network that is later assembled into the notably transversally oriented tubular network. Accumulating evidence supports the idea that the development of TATS is linked to cell dimensions, calcium signaling, and increasing myofibrillar content orchestrated by electromechanical stimulation. However, the overall mechanism has not yet been described. The topic of this review is the development of TATS with an emphasis on the irregular phase of tubule growth. The traditional models of BIN1-related tubulation are also discussed. We summarized the recently described protein interactions during TATS development, mainly mediated by costameric and sarcomeric proteins, supporting the idea of the coupling sites between TATS and the myofibrils. We hypothesize that the formation and final organization of the tubular system is driven by the simultaneous development of the contractile apparatus under cycling electromechanical stimulus.
Collapse
Affiliation(s)
| | - Katarina Mackova
- Department of Biophysics and Electrophysiology, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
2
|
Bruge C, Bourg N, Pellier E, Tournois J, Polentes J, Benabides M, Grossi N, Bigot A, Brureau A, Richard I, Nissan X. High-throughput screening identifies bazedoxifene as a potential therapeutic for dysferlin-deficient limb girdle muscular dystrophy. Br J Pharmacol 2025. [PMID: 40108832 DOI: 10.1111/bph.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Limb-girdle muscular dystrophy R2 (LGMD R2) is a rare genetic disorder characterised by progressive weakness and wasting of proximal muscles. LGMD R2 is caused by the loss of function of dysferlin, a transmembrane protein crucial for plasma membrane repair in skeletal muscles. This study aimed to identify drugs that could improve the localisation and restore the function of an aggregated mutant form of dysferlin (DYSFL1341P). EXPERIMENTAL APPROACH We developed an in vitro high-throughput assay to monitor the expression and reallocation of aggregated mutant dysferlin (DYSFL1341P) in immortalised myoblasts. After screening 2239 clinically approved drugs and bioactive compounds, the ability of the more promising candidates to improve cell survival following hypo-osmotic shock was assessed. Their protective effects were evaluated on immortalised myoblasts carrying other dysferlin mutations and on dysferlin-deficient muscle fibres from Bla/J mice. KEY RESULTS We identified two compounds, saracatinib and bazedoxifene, that increase dysferlin content in cells carrying the DYSFL1341P mutation. Both drugs improved cell survival and plasma membrane resistance following osmotic shock. Whereas saracatinib acts specifically on misfolded L1341P dysferlin, bazedoxifene shows an additional protective effect on dysferlin KO immortalised myoblasts and mice muscle fibres. Further analysis revealed that bazedoxifene induces autophagy flux, which may enhance the survival of LGMD R2 myofibres. CONCLUSION AND IMPLICATIONS Our drug screening identified saracatinib and bazedoxifene as potential treatments for LGMD R2, especially for patients with the L1341P mutation. The widespread protective effect of bazedoxifene reveals a new avenue toward genotype-independent treatment of LGMD R2 patients.
Collapse
Affiliation(s)
- Celine Bruge
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Nathalie Bourg
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| | - Emilie Pellier
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Johana Tournois
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Jerome Polentes
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Manon Benabides
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Noella Grossi
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Anthony Brureau
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| | - Isabelle Richard
- INTEGRARE, Genethon, Inserm, Université d'Evry, Université Paris-Saclay, Evry, France
| | - Xavier Nissan
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- CECS, IStem, Corbeil-Essonnes, France
| |
Collapse
|
3
|
Lloyd EM, Hepburn MS, Li J, Mowla A, Jeong JH, Hwang Y, Choi YS, Jackaman C, Kennedy BF, Grounds MD. Multimodal three-dimensional characterization of murine skeletal muscle micro-scale elasticity, structure, and composition: Impact of dysferlinopathy, Duchenne muscular dystrophy, and age on three hind-limb muscles. J Mech Behav Biomed Mater 2024; 160:106751. [PMID: 39326249 DOI: 10.1016/j.jmbbm.2024.106751] [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: 05/27/2024] [Revised: 08/21/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
Skeletal muscle tissue function is governed by the mechanical properties and organization of its components, including myofibers, extracellular matrix, and adipose tissue, which can be modified by the onset and progression of many disorders. This study used a novel combination of quantitative micro-elastography and clearing-enhanced three-dimensional (3D) microscopy to assess 3D micro-scale elasticity and micro-architecture of muscles from two muscular dystrophies: dysferlinopathy and Duchenne muscular dystrophy, using male BLA/J and mdx mice, respectively, and their wild-type (WT) controls. We examined three muscles with varying proportions of slow- and fast-twitch myofibers: the soleus (predominantly slow), extensor digitorum longus (EDL; fast), and quadriceps (mixed), from BLA/J and WTBLA/J mice aged 3, 10, and 24 months, and mdx and WTmdx mice aged 10 months. Both dysferlin deficiency and age reduced the elasticity and variability of elasticity of the soleus and quadriceps, but not EDL. Overall, the BLA/J soleus was 20% softer than WT and less mechanically heterogeneous (-14% in standard deviation of elasticity). The BLA/J quadriceps at 24 months was 72% softer than WT and less mechanically heterogeneous (-59% in standard deviation), with substantial adipose tissue accumulation. While mdx muscles did not differ quantitatively from WT, regional heterogeneity was evident in micro-scale elasticity and micro-architecture of quadriceps (e.g., 11.2 kPa in a region with marked pathology vs 3.8 kPa in a less affected area). These results demonstrate differing biomechanical changes in hind-limb muscles of two distinct muscular dystrophies, emphasizing the potential for this novel multimodal technique to identify important differences between various myopathies.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Curtin Health Innovation Research Institute, Curtin Medical School, Faculty of Health Sciences, Curtin University, Kent St, Bentley, Western Australia, 6102, Australia.
| | - Matt S Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland.
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.
| | - Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia.
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, 31151, Republic of Korea.
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, 31151, Republic of Korea.
| | - Yu Suk Choi
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia.
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin Medical School, Faculty of Health Sciences, Curtin University, Kent St, Bentley, Western Australia, 6102, Australia.
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia.
| |
Collapse
|
4
|
Zhou Y, Liu Y, Luo H, Wen C, Cui Y, Du L, Kwaku OE, Li L, Xiong L, Zheng J, Ding X, Shen X, Zhou P, Hu H, Yue R. Myoferlin alleviates pressure overload-induced cardiac hypertrophy and dysfunction by inhibiting NLRP3-mediated pyroptosis. PeerJ 2024; 12:e18499. [PMID: 39553724 PMCID: PMC11568814 DOI: 10.7717/peerj.18499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
Myoferlin (MYOF) is a muscle-derived secretory protein. Recent studies have found that MYOF protects against cell damage. However, the role of MYOF in cardiac hypertrophy remains unclear. Increasing evidence suggests that NLRP3 (NOD-like receptor protein 3) and the pyroptosis cascade play critical roles in the development of cardiac hypertrophy and inflammation. To investigate the role of MYOF in cardiac hypertrophy, we conducted a transverse aortic constriction (TAC) experiment in a mouse model. We found that MYOF can improve cardiac hypertrophy and cardiac function. Furthermore, our study confirmed a connection between cardiac hypertrophy and myocardial pyroptosis. Cardiac hypertrophy significantly increased the proportion of apoptotic cells and upregulated apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, and gasdermin D (GSDMD). This suggests that pharmacological or genetic inhibition of NLRP3 can effectively reduce cardiac hypertrophy. An abnormal increase in NLRP3 can reverse the cardioprotective effects of MYOF. Our findings indicate that MYOF is a potential therapeutic agent for cardiac hypertrophy.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yanxu Liu
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hao Luo
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Cong Wen
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yangyang Cui
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Linqing Du
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ofe Eugene Kwaku
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Family Health University College and Hospital, Opposite Kofi Annan International Peace Keeping Training Center, Teshie- Accra, Ghana
| | - Lan Li
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lijuan Xiong
- Department of Cardiology, People’s Hospital of Guang ’an District, Guang ’an, Sichuan, China
| | - Jiankang Zheng
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xuefeng Ding
- Department of Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiufeng Shen
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Peng Zhou
- Department of Cardiology, People’s Hospital of Guang ’an District, Guang ’an, Sichuan, China
| | - Houxiang Hu
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Rongchuan Yue
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Cardiology, People’s Hospital of Guang ’an District, Guang ’an, Sichuan, China
| |
Collapse
|
5
|
Paulke NJ, Fleischhacker C, Wegener JB, Riedemann GC, Cretu C, Mushtaq M, Zaremba N, Möbius W, Zühlke Y, Wedemeyer J, Liebmann L, Gorshkova AA, Kownatzki-Danger D, Wagner E, Kohl T, Wichmann C, Jahn O, Urlaub H, Toischer K, Hasenfuß G, Moser T, Preobraschenski J, Lenz C, Rog-Zielinska EA, Lehnart SE, Brandenburg S. Dysferlin Enables Tubular Membrane Proliferation in Cardiac Hypertrophy. Circ Res 2024; 135:554-574. [PMID: 39011635 DOI: 10.1161/circresaha.124.324588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Cardiac hypertrophy compensates for increased biomechanical stress of the heart induced by prevalent cardiovascular pathologies but can result in heart failure if left untreated. Here, we hypothesized that the membrane fusion and repair protein dysferlin is critical for the integrity of the transverse-axial tubule (TAT) network inside cardiomyocytes and contributes to the proliferation of TAT endomembranes during pressure overload-induced cardiac hypertrophy. METHODS Stimulated emission depletion and electron microscopy were used to localize dysferlin in mouse and human cardiomyocytes. Data-independent acquisition mass spectrometry revealed the cardiac dysferlin interactome and proteomic changes of the heart in dysferlin-knockout mice. After transverse aortic constriction, we compared the hypertrophic response of wild-type versus dysferlin-knockout hearts and studied TAT network remodeling mechanisms inside cardiomyocytes by live-cell membrane imaging. RESULTS We localized dysferlin in a vesicular compartment in nanometric proximity to contact sites of the TAT network with the sarcoplasmic reticulum, a.k.a. junctional complexes for Ca2+-induced Ca2+ release. Interactome analyses demonstrated a novel protein interaction of dysferlin with the membrane-tethering sarcoplasmic reticulum protein juncophilin-2, a putative interactor of L-type Ca2+ channels and ryanodine receptor Ca2+ release channels in junctional complexes. Although the dysferlin-knockout caused a mild progressive phenotype of dilated cardiomyopathy, global proteome analysis revealed changes preceding systolic failure. Following transverse aortic constriction, dysferlin protein expression was significantly increased in hypertrophied wild-type myocardium, while dysferlin-knockout animals presented markedly reduced left-ventricular hypertrophy. Live-cell membrane imaging showed a profound reorganization of the TAT network in wild-type left-ventricular myocytes after transverse aortic constriction with robust proliferation of axial tubules, which critically depended on the increased expression of dysferlin within newly emerging tubule components. CONCLUSIONS Dysferlin represents a new molecular target in cardiac disease that protects the integrity of tubule-sarcoplasmic reticulum junctional complexes for regulated excitation-contraction coupling and controls TAT network reorganization and tubular membrane proliferation in cardiomyocyte hypertrophy induced by pressure overload.
Collapse
Affiliation(s)
- Nora Josefine Paulke
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Carolin Fleischhacker
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Justus B Wegener
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Gabriel C Riedemann
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Constantin Cretu
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience and InnerEarLab (C.C., J.P.), University Medical Center Göttingen, Germany
| | - Mufassra Mushtaq
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Nina Zaremba
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Electron Microscopy, City Campus (W.M.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Yannik Zühlke
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Jasper Wedemeyer
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Lorenz Liebmann
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Anastasiia A Gorshkova
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Daniel Kownatzki-Danger
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Now with Institute of Transfusion Medicine, University Hospital Schleswig-Holstein; Kiel, Germany (D.K.-D)
| | - Eva Wagner
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Tobias Kohl
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab and Center for Biostructural Imaging of Neurodegeneration (C.W.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Olaf Jahn
- Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy (O.J.), University Medical Center Göttingen, Germany
- Neuroproteomics Group, Department of Molecular Neurobiology (O.J.)
| | - Henning Urlaub
- Department of Clinical Chemistry (H.U., C.L.), University Medical Center Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany (H.U., C.L.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Karl Toischer
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany (K.T., G.H., S.E.L.)
| | - Gerd Hasenfuß
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany (K.T., G.H., S.E.L.)
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab (T.M.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Julia Preobraschenski
- Biochemistry of Membrane Dynamics Group, Institute for Auditory Neuroscience and InnerEarLab (C.C., J.P.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Christof Lenz
- Department of Clinical Chemistry (H.U., C.L.), University Medical Center Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany (H.U., C.L.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Faculty of Medicine, University of Freiburg, Germany (E.A.R.-Z.)
| | - Stephan E Lehnart
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany (K.T., G.H., S.E.L.)
| | - Sören Brandenburg
- Department of Cardiology and Pneumology (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., K.T., G.H., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center (N.J.P., C.F., J.B.W., G.C.R., M.M., N.Z., Y.Z., J.W., L.L., A.A.G., D.K.-D., E.W., T.K., S.E.L., S.B.), University Medical Center Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (W.M., C.W., H.U., K.T., G.H., T.M., J.P., C.L., S.E.L., S.B.)
| |
Collapse
|
6
|
Lloyd EM, Crew RC, Haynes VR, White RB, Mark PJ, Jackaman C, Papadimitriou JM, Pinniger GJ, Murphy RM, Watt MJ, Grounds MD. Pilot investigations into the mechanistic basis for adverse effects of glucocorticoids in dysferlinopathy. Skelet Muscle 2024; 14:19. [PMID: 39123261 PMCID: PMC11312411 DOI: 10.1186/s13395-024-00350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by gene mutations resulting in deficiency of the membrane-associated protein dysferlin. They manifest post-growth and are characterised by muscle wasting (primarily in the limb and limb-gridle muscles), inflammation, and replacement of myofibres with adipose tissue. The precise pathomechanism for dysferlinopathy is currently unclear; as such there are no treatments currently available. Glucocorticoids (GCs) are widely used to reduce inflammation and treat muscular dystrophies, but when administered to patients with dysferlinopathy, they have unexpected adverse effects, with accelerated loss of muscle strength. METHODS To investigate the mechanistic basis for the adverse effects of GCs in dysferlinopathy, the potent GC dexamethasone (Dex) was administered for 4-5 weeks (0.5-0.75 µg/mL in drinking water) to dysferlin-deficient BLA/J and normal wild-type (WT) male mice, sampled at 5 (Study 1) or 10 months (Study 2) of age. A wide range of analyses were conducted. Metabolism- and immune-related gene expression was assessed in psoas muscles at both ages and in quadriceps at 10 months of age. For the 10-month-old mice, quadriceps and psoas muscle histology was assessed. Additionally, we investigated the impact of Dex on the predominantly slow and fast-twitch soleus and extensor digitorum longus (EDL) muscles (respectively) in terms of contractile function, myofibre-type composition, and levels of proteins related to contractile function and metabolism, plus glycogen. RESULTS At both ages, many complement-related genes were highly expressed in BLA/J muscles, and WT mice were generally more responsive to Dex than BLA/J. The effects of Dex on BLA/J mice included (i) increased expression of inflammasome-related genes in muscles (at 5 months) and (ii) exacerbated histopathology of quadriceps and psoas muscles at 10 months. A novel observation was pronounced staining for glycogen in many myofibres of the damaged quadriceps muscles, with large pale vacuolated myofibres, suggesting possible myofibre death by oncosis. CONCLUSION These pilot studies provide a new focus for further investigation into the adverse effects of GCs on dysferlinopathic muscles.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - Rachael C Crew
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| | - Vanessa R Haynes
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Robert B White
- MD Education Unit, UWA Medical School, The University of Western Australia, Perth, WA, Australia
| | - Peter J Mark
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - John M Papadimitriou
- Department of Pathology and Laboratory Medicine, UWA Medical School, The University of Western Australia, Perth, WA, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
7
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
8
|
Poudel BH, Fletcher S, Wilton SD, Aung-Htut M. Limb Girdle Muscular Dystrophy Type 2B (LGMD2B): Diagnosis and Therapeutic Possibilities. Int J Mol Sci 2024; 25:5572. [PMID: 38891760 PMCID: PMC11171558 DOI: 10.3390/ijms25115572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Dysferlin is a large transmembrane protein involved in critical cellular processes including membrane repair and vesicle fusion. Mutations in the dysferlin gene (DYSF) can result in rare forms of muscular dystrophy; Miyoshi myopathy; limb girdle muscular dystrophy type 2B (LGMD2B); and distal myopathy. These conditions are collectively known as dysferlinopathies and are caused by more than 600 mutations that have been identified across the DYSF gene to date. In this review, we discuss the key molecular and clinical features of LGMD2B, the causative gene DYSF, and the associated dysferlin protein structure. We also provide an update on current approaches to LGMD2B diagnosis and advances in drug development, including splice switching antisense oligonucleotides. We give a brief update on clinical trials involving adeno-associated viral gene therapy and the current progress on CRISPR/Cas9 mediated therapy for LGMD2B, and then conclude by discussing the prospects of antisense oligomer-based intervention to treat selected mutations causing dysferlinopathies.
Collapse
Affiliation(s)
- Bal Hari Poudel
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
- Central Department of Biotechnology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
| | - May Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
9
|
Quinn CJ, Cartwright EJ, Trafford AW, Dibb KM. On the role of dysferlin in striated muscle: membrane repair, t-tubules and Ca 2+ handling. J Physiol 2024; 602:1893-1910. [PMID: 38615232 DOI: 10.1113/jp285103] [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: 06/27/2023] [Accepted: 03/05/2024] [Indexed: 04/15/2024] Open
Abstract
Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.
Collapse
Affiliation(s)
- C J Quinn
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - E J Cartwright
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - A W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - K M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| |
Collapse
|
10
|
Long AM, Kwon JM, Lee G, Reiser NL, Vaught LA, O'Brien JG, Page PGT, Hadhazy M, Reynolds JC, Crosbie RH, Demonbreun AR, McNally EM. The extracellular matrix differentially directs myoblast motility and differentiation in distinct forms of muscular dystrophy: Dystrophic matrices alter myoblast motility. Matrix Biol 2024; 129:44-58. [PMID: 38582404 PMCID: PMC11104166 DOI: 10.1016/j.matbio.2024.04.001] [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: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024]
Abstract
Extracellular matrix (ECM) pathologic remodeling underlies many disorders, including muscular dystrophy. Tissue decellularization removes cellular components while leaving behind ECM components. We generated "on-slide" decellularized tissue slices from genetically distinct dystrophic mouse models. The ECM of dystrophin- and sarcoglycan-deficient muscles had marked thrombospondin 4 deposition, while dysferlin-deficient muscle had excess decorin. Annexins A2 and A6 were present on all dystrophic decellularized ECMs, but annexin matrix deposition was excessive in dysferlin-deficient muscular dystrophy. Muscle-directed viral expression of annexin A6 resulted in annexin A6 in the ECM. C2C12 myoblasts seeded onto decellularized matrices displayed differential myoblast mobility and fusion. Dystrophin-deficient decellularized matrices inhibited myoblast mobility, while dysferlin-deficient decellularized matrices enhanced myoblast movement and differentiation. Myoblasts treated with recombinant annexin A6 increased mobility and fusion like that seen on dysferlin-deficient decellularized matrix and demonstrated upregulation of ECM and muscle cell differentiation genes. These findings demonstrate specific fibrotic signatures elicit effects on myoblast activity.
Collapse
Affiliation(s)
- Ashlee M Long
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jason M Kwon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - GaHyun Lee
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nina L Reiser
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lauren A Vaught
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph G O'Brien
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Patrick G T Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA; Department of Neurology David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA; Department of Neurology David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
11
|
Xiao J, Dong Y, Jin J, Yuan Z, Wang C, Xiang R, Guo Y. A missense variant in MYOF is associated with ARVC and sudden cardiac death. Gene 2024; 902:148193. [PMID: 38253296 DOI: 10.1016/j.gene.2024.148193] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 01/18/2024] [Indexed: 01/24/2024]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is rare autosomal dominant genetic disorder that leads to severe arrhythmia and sudden cardiac death. Although previous studies in clinical, pathological and genetics of ARVC established consensus diagnostic criteria and expanded the spectrum of pathogenic genes, there is still a proportion of patients with unclear causative factors. Here, whole-exome sequencing was employed to investigate the genetic etiology of a 15-year-old sudden cardiac death female caused by ARVC. A novel variant of MYOF (NM_013451.3: c.4723G > C: p.D1575H) was identified, which is a member of the Ferlin family of proteins is associated with cardiomyopathy. And the bioinformatics analysis predicted the pathogenicity of this variant. We report the first variant of MYOF in ARVC, which imply a vital role of MYOF in cardiomyopathy.
Collapse
Affiliation(s)
- Jiao Xiao
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Yi Dong
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China.
| | - Jieyuan Jin
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China.
| | - Zhuangzhuang Yuan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China.
| | - Chenyu Wang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China.
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China.
| | - Yadong Guo
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China.
| |
Collapse
|
12
|
Ning Y, Zhou X, Wang G, Zhang L, Wang J. Bioinformatics to Identify Biomarkers of Diabetic Nephropathy based on Sphingolipid Metabolism and their Molecular Mechanisms. Curr Diabetes Rev 2024; 21:e070524229720. [PMID: 38712372 DOI: 10.2174/0115733998297749240418071555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Diabetes mellitus (DM) frequently results in Diabetic Nephropathy (DN), which has a significant negative impact on the quality of life of diabetic patients. Sphingolipid metabolism is associated with diabetes, but its relationship with DN is unclear. Therefore, screening biomarkers related to sphingolipid metabolism is crucial for treating DN. METHODS To identify Differentially Expressed Genes (DEGs) in the GSE142153 dataset, we conducted a differential expression analysis (DN samples versus control samples). The intersection genes were obtained by overlapping DEGs and Sphingolipid Metabolism-Related Genes (SMRGs). Furthermore, The Least Absolute Shrinkage and Selection Operator (LASSO) and Support Vector Machine Recursive Feature Elimination (SVM-RFE) algorithms were used to filter biomarkers. We further analyzed the Gene Set Enrichment analysis (GSEA) and the immunoinfiltrational analysis based on biomarkers. RESULTS We identified 2,186 DEGs associated with DN. Then, five SMR-DEGs were obtained. Subsequently, biomarkers associated with sphingolipid metabolism (S1PR1 and SELL) were identified by applying machine learning and expression analysis. In addition, GSEA showed that these biomarkers were correlated with cytokine cytokine receptor interaction'. Significant variations in B cells, DCs, Tems, and Th2 cells between the two groups suggested that these cells might have a role in DN. CONCLUSION Overall, we obtained two sphingolipid metabolism-related biomarkers (S1PR1 and SELL) associated with DN, which laid a theoretical foundation for treating DN.
Collapse
Affiliation(s)
- Yaxian Ning
- Department of Nephrology, Second Hospital of Lanzhou University, Lanzhou 730030, Gansu, China
- Clinical Medical Research Center of Gansu Province(No. 21JR7RA436), Lanzhou 730030, Gansu, China
| | - Xiaochun Zhou
- Department of Nephrology, Second Hospital of Lanzhou University, Lanzhou 730030, Gansu, China
- Clinical Medical Research Center of Gansu Province(No. 21JR7RA436), Lanzhou 730030, Gansu, China
| | - Gouqin Wang
- Department of Nephrology, Second Hospital of Lanzhou University, Lanzhou 730030, Gansu, China
- Clinical Medical Research Center of Gansu Province(No. 21JR7RA436), Lanzhou 730030, Gansu, China
| | - Lili Zhang
- Department of Nephrology, Second Hospital of Lanzhou University, Lanzhou 730030, Gansu, China
- Clinical Medical Research Center of Gansu Province(No. 21JR7RA436), Lanzhou 730030, Gansu, China
| | - Jianqin Wang
- Department of Nephrology, Second Hospital of Lanzhou University, Lanzhou 730030, Gansu, China
- Clinical Medical Research Center of Gansu Province(No. 21JR7RA436), Lanzhou 730030, Gansu, China
| |
Collapse
|
13
|
Chen Y, Wu W, Wang P, Yip P, Wu Y, Lin Y, Lin W. Novel five nucleotide deletion in dysferlin leads to autosomal recessive limb-girdle muscular dystrophy. Physiol Rep 2023; 11:e15887. [PMID: 38110300 PMCID: PMC10727958 DOI: 10.14814/phy2.15887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/20/2023] Open
Abstract
Muscular dystrophy (MD) is a genetic disorder that causes progressive muscle weakness and degeneration. Limb-girdle muscular dystrophy (LGMD) is a type of MD that mainly causes muscle atrophy within the shoulder and pelvic girdles. LGMD is classified into autosomal dominant (LGMD-D) and autosomal recessive (LGMD-R) inheritance patterns. Mutations in the Dysferlin gene (DYSF) are common causes of LGMD-R. However, genetic screening of DYSF mutations is rare in Taiwan. Herein, we identified a novel c.2867_2871del ACCAG deletion and a previously reported c.937+1G>A mutation in DYSF from a Taiwanese family with LGMD. The primary symptoms of both siblings were difficulty climbing stairs, walking on the toes, and gradually worsening weakness in the proximal muscles and increased creatine kinase level. Through pedigree analysis and sequencing, two siblings from this family were found to have compound heterozygous DYSF mutations (c. 937+1G>A and c. 2867_2871del ACCAG) within the separated alleles. These mutations induced early stop codons; if translated, truncated DYSF proteins will be expressed. Or, the mRNA products of these two mutations will merit the nonsense-mediated decay, might result in no dysferlin protein expressed. To our knowledge, this is the first report of a novel c.2867_2871del ACCAG deletion in DYSF. Further research is required to examine the effects of the novel DYSF mutation in Taiwanese patients with LGMD.
Collapse
Affiliation(s)
- Yen‐Lin Chen
- Center for Precision Medicine and Genomics, Tri‐Service General HospitalMedical Defense Medical CenterTaipeiTaiwan
- Department of Pathology, Tri‐Service General HospitalMedical Defense Medical CenterTaipeiTaiwan
| | - Wen‐Bin Wu
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
| | - Pei Wang
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
| | - Ping‐Keung Yip
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
- Division of NeurologyCardinal Tien HospitalNew Taipei CityTaiwan
| | - Yi‐No Wu
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
| | - Ying‐Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical ScienceFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Wei‐Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical ScienceFu Jen Catholic UniversityNew Taipei CityTaiwan
| |
Collapse
|
14
|
Lloyd EM, Pinniger GJ, Murphy RM, Grounds MD. Slow or fast: Implications of myofibre type and associated differences for manifestation of neuromuscular disorders. Acta Physiol (Oxf) 2023; 238:e14012. [PMID: 37306196 DOI: 10.1111/apha.14012] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Many neuromuscular disorders can have a differential impact on a specific myofibre type, forming the central premise of this review. The many different skeletal muscles in mammals contain a spectrum of slow- to fast-twitch myofibres with varying levels of protein isoforms that determine their distinctive contractile, metabolic, and other properties. The variations in functional properties across the range of classic 'slow' to 'fast' myofibres are outlined, combined with exemplars of the predominantly slow-twitch soleus and fast-twitch extensor digitorum longus muscles, species comparisons, and techniques used to study these properties. Other intrinsic and extrinsic differences are discussed in the context of slow and fast myofibres. These include inherent susceptibility to damage, myonecrosis, and regeneration, plus extrinsic nerves, extracellular matrix, and vasculature, examined in the context of growth, ageing, metabolic syndrome, and sexual dimorphism. These many differences emphasise the importance of carefully considering the influence of myofibre-type composition on manifestation of various neuromuscular disorders across the lifespan for both sexes. Equally, understanding the different responses of slow and fast myofibres due to intrinsic and extrinsic factors can provide deep insight into the precise molecular mechanisms that initiate and exacerbate various neuromuscular disorders. This focus on the influence of different myofibre types is of fundamental importance to enhance translation for clinical management and therapies for many skeletal muscle disorders.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
15
|
Leclère JC, Dulon D. Otoferlin as a multirole Ca 2+ signaling protein: from inner ear synapses to cancer pathways. Front Cell Neurosci 2023; 17:1197611. [PMID: 37538852 PMCID: PMC10394277 DOI: 10.3389/fncel.2023.1197611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/28/2023] [Indexed: 08/05/2023] Open
Abstract
Humans have six members of the ferlin protein family: dysferlin, myoferlin, otoferlin, fer1L4, fer1L5, and fer1L6. These proteins share common features such as multiple Ca2+-binding C2 domains, FerA domains, and membrane anchoring through their single C-terminal transmembrane domain, and are believed to play a key role in calcium-triggered membrane fusion and vesicle trafficking. Otoferlin plays a crucial role in hearing and vestibular function. In this review, we will discuss how we see otoferlin working as a Ca2+-dependent mechanical sensor regulating synaptic vesicle fusion at the hair cell ribbon synapses. Although otoferlin is also present in the central nervous system, particularly in the cortex and amygdala, its role in brain tissues remains unknown. Mutations in the OTOF gene cause one of the most frequent genetic forms of congenital deafness, DFNB9. These mutations produce severe to profound hearing loss due to a defect in synaptic excitatory glutamatergic transmission between the inner hair cells and the nerve fibers of the auditory nerve. Gene therapy protocols that allow normal rescue expression of otoferlin in hair cells have just started and are currently in pre-clinical phase. In parallel, studies have linked ferlins to cancer through their effect on cell signaling and development, allowing tumors to form and cancer cells to adapt to a hostile environment. Modulation by mechanical forces and Ca2+ signaling are key determinants of the metastatic process. Although ferlins importance in cancer has not been extensively studied, data show that otoferlin expression is significantly associated with survival in specific cancer types, including clear cell and papillary cell renal carcinoma, and urothelial bladder cancer. These findings indicate a role for otoferlin in the carcinogenesis of these tumors, which requires further investigation to confirm and understand its exact role, particularly as it varies by tumor site. Targeting this protein may lead to new cancer therapies.
Collapse
Affiliation(s)
- Jean-Christophe Leclère
- Department of Head and Neck Surgery, Brest University Hospital, Brest, France
- Laboratory of Neurophysiologie de la Synapse Auditive, Université de Bordeaux, Bordeaux, France
| | - Didier Dulon
- Laboratory of Neurophysiologie de la Synapse Auditive, Université de Bordeaux, Bordeaux, France
- Institut de l’Audition, Institut Pasteur & INSERM UA06, Paris, France
| |
Collapse
|
16
|
Ballouhey O, Chapoton M, Alary B, Courrier S, Da Silva N, Krahn M, Lévy N, Weisleder N, Bartoli M. A Dysferlin Exon 32 Nonsense Mutant Mouse Model Shows Pathological Signs of Dysferlinopathy. Biomedicines 2023; 11:biomedicines11051438. [PMID: 37239109 DOI: 10.3390/biomedicines11051438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Dysferlinopathies are a group of autosomal recessive muscular dystrophies caused by pathogenic variants in the DYSF gene. While several animal models of dysferlinopathy have been developed, most of them involve major disruptions of the Dysf gene locus that are not optimal for studying human dysferlinopathy, which is often caused by single nucleotide substitutions. In this study, the authors describe a new murine model of dysferlinopathy that carries a nonsense mutation in Dysf exon 32, which has been identified in several patients with dysferlinopathy. This mouse model, called Dysf p.Y1159X/p.Y1159X, displays several molecular, histological, and functional defects observed in dysferlinopathy patients and other published mouse models. This mutant mouse model is expected to be useful for testing various therapeutic approaches such as termination codon readthrough, pharmacological approaches, and exon skipping. Therefore, the data presented in this study strongly support the use of this animal model for the development of preclinical strategies for the treatment of dysferlinopathies.
Collapse
Affiliation(s)
- Océane Ballouhey
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
| | - Marie Chapoton
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
| | - Benedicte Alary
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
| | | | - Nathalie Da Silva
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
| | - Martin Krahn
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
- Département de Génétique Médicale et de Biologie Cellulaire, AP-HM, Hôpital d'Enfants de la Timone, 13005 Marseille, France
| | - Nicolas Lévy
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
- Département de Génétique Médicale et de Biologie Cellulaire, AP-HM, Hôpital d'Enfants de la Timone, 13005 Marseille, France
| | - Noah Weisleder
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Marc Bartoli
- Aix Marseille University, INSERM, MMG, U1251, 13005 Marseille, France
| |
Collapse
|
17
|
Carpenter AP, Khuu P, Weidner T, Johnson CP, Roeters SJ, Baio JE. Orientation of the Dysferlin C2A Domain is Responsive to the Composition of Lipid Membranes. J Phys Chem B 2023; 127:577-589. [PMID: 36608331 DOI: 10.1021/acs.jpcb.2c06716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dysferlin is a 230 kD protein that plays a critical function in the active resealing of micron-sized injuries to the muscle sarcolemma by recruiting vesicles to patch the injured site via vesicle fusion. Muscular dystrophy is observed in humans when mutations disrupt this repair process or dysferlin is absent. While lipid binding by dysferlin's C2A domain (dysC2A) is considered fundamental to the membrane resealing process, the molecular mechanism of this interaction is not fully understood. By applying nonlinear surface-specific vibrational spectroscopy, we have successfully demonstrated that dysferlin's N-terminal C2A domain (dysC2A) alters its binding orientation in response to a membrane's lipid composition. These experiments reveal that dysC2A utilizes a generic electrostatic binding interaction to bind to most anionic lipid surfaces, inserting its calcium binding loops into the lipid surface while orienting its β-sheets 30-40° from surface normal. However, at lipid surfaces, where PI(4,5)P2 is present, dysC2A tilts its β-sheets more than 60° from surface normal to expose a polybasic face, while it binds to the PI(4,5)P2 surface. Both lipid binding mechanisms are shown to occur alongside dysC2A-induced lipid clustering. These different binding mechanisms suggest that dysC2A could provide a molecular cue to the larger dysferlin protein as to signal whether it is bound to the sarcolemma or another lipid surface.
Collapse
Affiliation(s)
- Andrew P Carpenter
- The School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon97331, United States
| | - Patricia Khuu
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon97331, United States
| | - Tobias Weidner
- Department of Chemistry, Aarhus University, 8000Aarhus C, Denmark
| | - Colin P Johnson
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon97331, United States
| | - Steven J Roeters
- Department of Chemistry, Aarhus University, 8000Aarhus C, Denmark
| | - Joe E Baio
- The School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon97331, United States
| |
Collapse
|
18
|
Lloyd EM, Pinniger GJ, Grounds MD, Murphy RM. Dysferlin Deficiency Results in Myofiber-Type Specific Differences in Abundances of Calcium-Handling and Glycogen Metabolism Proteins. Int J Mol Sci 2022; 24:ijms24010076. [PMID: 36613515 PMCID: PMC9820290 DOI: 10.3390/ijms24010076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by a genetic deficiency of the membrane-associated protein dysferlin, which usually manifest post-growth in young adults. The disease is characterized by progressive skeletal muscle wasting in the limb-girdle and limbs, inflammation, accumulation of lipid droplets in slow-twitch myofibers and, in later stages, replacement of muscles by adipose tissue. Previously we reported myofiber-type specific differences in muscle contractile function of 10-month-old dysferlin-deficient BLAJ mice that could not be fully accounted for by altered myofiber-type composition. In order to further investigate these findings, we examined the impact of dysferlin deficiency on the abundance of calcium (Ca2+) handling and glucose/glycogen metabolism-related proteins in predominantly slow-twitch, oxidative soleus and fast-twitch, glycolytic extensor digitorum longus (EDL) muscles of 10-month-old wild-type (WT) C57BL/6J and dysferlin-deficient BLAJ male mice. Additionally, we compared the Ca2+ activation properties of isolated slow- and fast-twitch myofibers from 3-month-old WT and BLAJ male mice. Differences were observed for some Ca2+ handling and glucose/glycogen metabolism-related protein levels between BLAJ soleus and EDL muscles (compared with WT) that may contribute to the previously reported differences in function in these BLAJ muscles. Dysferlin deficiency did not impact glycogen content of whole muscles nor Ca2+ activation of the myofilaments, although soleus muscle from 10-month-old BLAJ mice had more glycogen than EDL muscles. These results demonstrate a further impact of dysferlin deficiency on proteins associated with excitation-contraction coupling and glycogen metabolism in skeletal muscles, potentially contributing to altered contractile function in dysferlinopathy.
Collapse
Affiliation(s)
- Erin M. Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gavin J. Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Correspondence:
| | - Robyn M. Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
19
|
Guo Z, Wang L, Liu H, Xie Y. Innate Immune Memory in Monocytes and Macrophages: The Potential Therapeutic Strategies for Atherosclerosis. Cells 2022; 11:4072. [PMID: 36552836 PMCID: PMC9776628 DOI: 10.3390/cells11244072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerosis is a complex metabolic disease characterized by the dysfunction of lipid metabolism and chronic inflammation in the intimal space of the vessel. As the most abundant innate immune cells, monocyte-derived macrophages play a pivotal role in the inflammatory response, cholesterol metabolism, and foam cell formation. In recent decades, it has been demonstrated that monocytes and macrophages can establish innate immune memory (also termed trained immunity) via endogenous and exogenous atherogenic stimuli and exhibit a long-lasting proinflammatory phenotype. The important cellular metabolism processes, including glycolysis, oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, fatty acid synthesis, and cholesterol synthesis, are reprogrammed. Trained monocytes/macrophages with innate immune memory can be persistently hyperactivated and can undergo extensive epigenetic rewiring, which contributes to the pathophysiological development of atherosclerosis via increased proinflammatory cytokine production and lipid accumulation. Here, we provide an overview of the regulation of cellular metabolic processes and epigenetic modifications of innate immune memory in monocytes/macrophages as well as the potential endogenous and exogenous stimulations involved in the progression of atherosclerosis that have been reported recently. These elucidations might be beneficial for further understanding innate immune memory and the development of therapeutic strategies for inflammatory diseases and atherosclerosis.
Collapse
Affiliation(s)
- Zhigang Guo
- Huanghe Science and Technology College, Zhengzhou 450006, China
| | - Lixue Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an 271018, China
| | - Hongjian Liu
- Department of Pharmacy, The Second Affiliated Hospital of Shandong First Medical University, Tai’an 271000, China
| | - Yuhuai Xie
- Huanghe Science and Technology College, Zhengzhou 450006, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Gargan S, Dowling P, Zweyer M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic Identification of Markers of Membrane Repair, Regeneration and Fibrosis in the Aged and Dystrophic Diaphragm. Life (Basel) 2022; 12:1679. [PMID: 36362832 PMCID: PMC9696191 DOI: 10.3390/life12111679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 07/30/2023] Open
Abstract
Deficiency in the membrane cytoskeletal protein dystrophin is the underlying cause of the progressive muscle wasting disease named Duchenne muscular dystrophy. In order to detect novel disease marker candidates and confirm the complexity of the pathobiochemical signature of dystrophinopathy, mass spectrometric screening approaches represent ideal tools for comprehensive biomarker discovery studies. In this report, we describe the comparative proteomic analysis of young versus aged diaphragm muscles from wild type versus the dystrophic mdx-4cv mouse model of X-linked muscular dystrophy. The survey confirmed the drastic reduction of the dystrophin-glycoprotein complex in the mdx-4cv diaphragm muscle and concomitant age-dependent changes in key markers of muscular dystrophy, including proteins involved in cytoskeletal organization, metabolite transportation, the cellular stress response and excitation-contraction coupling. Importantly, proteomic markers of the regulation of membrane repair, tissue regeneration and reactive myofibrosis were detected by mass spectrometry and changes in key proteins were confirmed by immunoblotting. Potential disease marker candidates include various isoforms of annexin, the matricellular protein periostin and a large number of collagens. Alterations in these proteoforms can be useful to evaluate adaptive, compensatory and pathobiochemical changes in the intracellular cytoskeleton, myofiber membrane integrity and the extracellular matrix in dystrophin-deficient skeletal muscle tissues.
Collapse
Affiliation(s)
- Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children’s Hospital, German Center for Neurodegenerative Diseases, University of Bonn, D53127 Bonn, Germany
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, D09 E432 Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, D09 E432 Dublin, Ireland
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, D53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
| |
Collapse
|
21
|
Zhang X, He D, Xiang Y, Wang C, Liang B, Li B, Qi D, Deng Q, Yu H, Lu Z, Zheng F. DYSF promotes monocyte activation in atherosclerotic cardiovascular disease as a DNA methylation-driven gene. Transl Res 2022; 247:19-38. [PMID: 35460889 DOI: 10.1016/j.trsl.2022.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Dysferlin (DYSF) has drawn much attention due to its involvement in dysferlinopathy and was reported to affect monocyte functions in recent studies. However, the role of DYSF in the pathogenesis of atherosclerotic cardiovascular diseases (ASCVD) and the regulation mechanism of DYSF expression have not been fully studied. In this study, Gene Expression Omnibus (GEO) database and epigenome-wide association study (EWAS) literatures were searched to find the DNA methylation-driven genes (including DYSF) of ASCVD. The hub genes related to DYSF were also identified through weighted correlation network analysis (WGCNA). Regulation of DYSF expression through its promoter methylation status was verified using peripheral blood leucocytes (PBLs) from ASCVD patients and normal controls, and experiments on THP1 cells and Apoe-/- mice. Similarly, the expressions of DYSF related hub genes, mainly contained SELL, STAT3 and TMX1, were also validated. DYSF functions were then evaluated by phagocytosis, transwell and adhesion assays in DYSF knock-down and overexpressed THP1 cells. The results showed that DYSF promoter hypermethylation up-regulated its expression in clinical samples, THP1 cells and Apoe-/- mice, confirming DYSF as a DNA methylation-driven gene. The combination of DYSF expression and methylation status in PBLs had a considerable prediction value for ASCVD. Besides, DYSF could enhance the phagocytosis, migration and adhesion ability of THP1 cells. Among DYSF related hub genes, SELL was proven to be the downstream target of DYSF by wet experiments. In conclusion, DYSF promoter hypermethylation upregulated its expression and promoted monocytes activation, which further participated in the pathogenesis of ASCVD.
Collapse
Affiliation(s)
- Xiaokang Zhang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Dingdong He
- Department of Clinical Laboratory Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yang Xiang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chen Wang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Bin Liang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Boyu Li
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Daoxi Qi
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qianyun Deng
- Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510000, China
| | - Hong Yu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Zhibing Lu
- Institute of Myocardial Injury and Repair, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fang Zheng
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
22
|
Chernova ON, Chekmareva IA, Mavlikeev MO, Yakovlev IA, Kiyasov AP, Deev RV. Structural and ultrastructural changes in the skeletal muscles of dysferlin-deficient mice during postnatal ontogenesis. Ultrastruct Pathol 2022; 46:359-367. [PMID: 35880824 DOI: 10.1080/01913123.2022.2105464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
A number of sarcolemma proteins are responsible for muscle fiber repair. Dysferlin encoded by the DYSF gene is one of these proteins. Dysferlin promotes membrane repair in striated muscle fibers (MFs). Mutations in DYSF lead to loss of or decreased dysferlin expression, impaired membrane repair in MF, and its destruction, clinically manifesting as dysferlinopathy. Preclinical studies of cell and gene therapies aimed at restoring impaired muscle regeneration require well-characterized small animal models. Our investigation aimed to distinguish the histopathological features of a mouse strain lacking dysferlin expression (Bla/J strain). Ultrastructural changes in the sarcolemma, mitochondria and contractile apparatus were observed. It was shown that postnatal histogenesis of skeletal muscles in genetically determined dysferlin deficiency is characterized by a higher proportion of necrotic muscle fibers, compensatory hypertrophy of muscle fibers with their subsequent atrophy, and decreases in proliferative activity and the level of myogenic differentiation of myogenic progenitor cells compared to wild-type mice (C57Bl/6).
Collapse
Affiliation(s)
- O N Chernova
- Human Morphology Department, North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, Russian Federation.,Pathology and Forensic Medicine Department, Saint-Petersburg Medico-Social Institute, Saint-Petersburg, Russian Federation
| | - I A Chekmareva
- A.V. Vishnevsky National Medical Research Center of Surgery, Moscow, Russian Federation
| | - M O Mavlikeev
- Pathology Department, North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, Russian Federation
| | - I A Yakovlev
- Genotarget LLC, Moscow, Russian Federation.,Human Stem Cell Institute PJSC, Moscow, Russian Federation
| | - A P Kiyasov
- Morphology and General Pathology Department, Kazan (Volga region) Federal University, Kazan, Russian Federation
| | - R V Deev
- Pathology Department, North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, Russian Federation.,Human Stem Cell Institute PJSC, Moscow, Russian Federation
| |
Collapse
|
23
|
Abstract
In mammalian cardiac myocytes, the plasma membrane includes the surface sarcolemma but also a network of membrane invaginations called transverse (t-) tubules. These structures carry the action potential deep into the cell interior, allowing efficient triggering of Ca2+ release and initiation of contraction. Once thought to serve as rather static enablers of excitation-contraction coupling, recent work has provided a newfound appreciation of the plasticity of the t-tubule network's structure and function. Indeed, t-tubules are now understood to support dynamic regulation of the heartbeat across a range of timescales, during all stages of life, in both health and disease. This review article aims to summarize these concepts, with consideration given to emerging t-tubule regulators and their targeting in future therapies.
Collapse
Affiliation(s)
- Katharine M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo Norway
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
24
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
25
|
Pi R, Chen Y, Du Y, Dong S. Comprehensive Analysis of Myoferlin in Human Pancreatic Cancer via Bioinformatics. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2602322. [PMID: 34957301 PMCID: PMC8702316 DOI: 10.1155/2021/2602322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 01/11/2023]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related death and urgently needs biomarkers for clinical diagnosis and prognosis. It has been reported that myoferlin (MYOF) is implicated in the regulation of proliferation, invasion, and migration of tumor cells in many cancers including pancreatic cancer. To confirm the prognostic value of MYOF in pancreatic cancer, a comprehensive cancer versus healthy people analysis was conducted using public data. MYOF mRNA expression levels were compared in many kinds of cancers including pancreatic cancer via the Oncomine and Gene Expression Profiling Interactive Analysis (GEPIA) databases. The results have shown that MYOF mRNA expression levels were upregulated in most types of cancers, especially in pancreatic cancer, compared with healthy people's tissues. Data from the Cancer Cell Line Encyclopedia (CCLE) and European Bioinformatics Institute (EMBL-EML) database also revealed that MYOF mRNA is highly expressed in most cancer cells, particularly in pancreatic cancer cell lines. Furthermore, the prognostic value of MYOF was evaluated using GEPIA and Long-term Outcome and Gene Expression Profiling Database of pan-cancers (LOGpc) database. Higher expression of MYOF was associated with poorer overall survival, especially in the lower stage and lower grade. Coexpressed genes, possible regulators, and the correlation between MYOF expressions were analyzed via the GEPIA and LinkedOmics database. Nineteen coexpressed genes were identified, and most of these genes were related to cancer. The Tumor Immune Estimation Resource (TIMER) database was used to analyze the correlation between MYOF and immune response. Notably, we found that MYOF might have a potential novel immune regulatory role in tumor immunity. These results support that MYOF is a candidate prognostic biomarker for pancreatic cancer, which calls for further genomics research of pancreatic cancer and deeply functional studies on MYOF.
Collapse
Affiliation(s)
- Rou Pi
- Shanghai Engineering Research Centre of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yanmei Chen
- Shanghai Engineering Research Centre of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yijie Du
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Suzhen Dong
- Shanghai Engineering Research Centre of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| |
Collapse
|
26
|
Barefield DY, Sell JJ, Tahtah I, Kearns SD, McNally EM, Demonbreun AR. Loss of dysferlin or myoferlin results in differential defects in excitation-contraction coupling in mouse skeletal muscle. Sci Rep 2021; 11:15865. [PMID: 34354129 PMCID: PMC8342512 DOI: 10.1038/s41598-021-95378-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022] Open
Abstract
Muscular dystrophies are disorders characterized by progressive muscle loss and weakness that are both genotypically and phenotypically heterogenous. Progression of muscle disease arises from impaired regeneration, plasma membrane instability, defective membrane repair, and calcium mishandling. The ferlin protein family, including dysferlin and myoferlin, are calcium-binding, membrane-associated proteins that regulate membrane fusion, trafficking, and tubule formation. Mice lacking dysferlin (Dysf), myoferlin (Myof), and both dysferlin and myoferlin (Fer) on an isogenic inbred 129 background were previously demonstrated that loss of both dysferlin and myoferlin resulted in more severe muscle disease than loss of either gene alone. Furthermore, Fer mice had disordered triad organization with visibly malformed transverse tubules and sarcoplasmic reticulum, suggesting distinct roles of dysferlin and myoferlin. To assess the physiological role of disorganized triads, we now assessed excitation contraction (EC) coupling in these models. We identified differential abnormalities in EC coupling and ryanodine receptor disruption in flexor digitorum brevis myofibers isolated from ferlin mutant mice. We found that loss of dysferlin alone preserved sensitivity for EC coupling and was associated with larger ryanodine receptor clusters compared to wildtype myofibers. Loss of myoferlin alone or together with a loss of dysferlin reduced sensitivity for EC coupling, and produced disorganized and smaller ryanodine receptor cluster size compared to wildtype myofibers. These data reveal impaired EC coupling in Myof and Fer myofibers and slightly potentiated EC coupling in Dysf myofibers. Despite high homology, dysferlin and myoferlin have differential roles in regulating sarcotubular formation and maintenance resulting in unique impairments in calcium handling properties.
Collapse
Affiliation(s)
- David Y Barefield
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA.
| | - Jordan J Sell
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Ibrahim Tahtah
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Samuel D Kearns
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. .,Center for Genetic Medicine, Northwestern University, 303 E Superior Lurie 5-512, Chicago, IL, 60611, USA.
| |
Collapse
|
27
|
Begam M, Roche R, Hass JJ, Basel CA, Blackmer JM, Konja JT, Samojedny AL, Collier AF, Galen SS, Roche JA. The effects of concentric and eccentric training in murine models of dysferlin-associated muscular dystrophy. Muscle Nerve 2020; 62:393-403. [PMID: 32363622 DOI: 10.1002/mus.26906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/21/2020] [Accepted: 04/25/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Dysferlin-deficient murine muscle sustains severe damage after repeated eccentric contractions. METHODS With a robotic dynamometer, we studied the response of dysferlin-sufficient and dysferlin-deficient mice to 12 weeks of concentrically or eccentrically biased contractions. We also studied whether concentric contractions before or after eccentric contractions reduced muscle damage in dysferlin-deficient mice. RESULTS After 12 weeks of concentric training, there was no net gain in contractile force in dysferlin-sufficient or dysferlin-deficient mice, whereas eccentric training produced a net gain in force in both mouse strains. However, eccentric training induced more muscle damage in dysferlin-deficient vs dysferlin-sufficient mice. Although concentric training produced minimal muscle damage in dysferlin-deficient mice, it still led to a prominent increase in centrally nucleated fibers. Previous exposure to concentric contractions conferred slight protection on dysferlin-deficient muscle against damage from subsequent injurious eccentric contractions. DISCUSSION Concentric contractions may help dysferlin-deficient muscle derive the benefits of exercise without inducing damage.
Collapse
Affiliation(s)
- Morium Begam
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Renuka Roche
- Occupational Therapy Program, College of Health and Human Services, Eastern Michigan University, Ypsilanti, Michigan
| | - Joshua J Hass
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Chantel A Basel
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Jacob M Blackmer
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Jasmine T Konja
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Amber L Samojedny
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Alyssa F Collier
- Rehabilitation Department, Emory University Hospital, Atlanta, Georgia
| | - Sujay S Galen
- Department of Physical Therapy, Byrdine F. Lewis College of Nursing & Health Professions, Georgia State University, Atlanta, Georgia
| | - Joseph A Roche
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| |
Collapse
|
28
|
Abstract
Ferlins are multiple-C2-domain proteins involved in Ca2+-triggered membrane dynamics within the secretory, endocytic and lysosomal pathways. In bony vertebrates there are six ferlin genes encoding, in humans, dysferlin, otoferlin, myoferlin, Fer1L5 and 6 and the long noncoding RNA Fer1L4. Mutations in DYSF (dysferlin) can cause a range of muscle diseases with various clinical manifestations collectively known as dysferlinopathies, including limb-girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. A mutation in MYOF (myoferlin) was linked to a muscular dystrophy accompanied by cardiomyopathy. Mutations in OTOF (otoferlin) can be the cause of nonsyndromic deafness DFNB9. Dysregulated expression of any human ferlin may be associated with development of cancer. This review provides a detailed description of functions of the vertebrate ferlins with a focus on muscle ferlins and discusses the mechanisms leading to disease development.
Collapse
|
29
|
van Putten M, Lloyd EM, de Greef JC, Raz V, Willmann R, Grounds MD. Mouse models for muscular dystrophies: an overview. Dis Model Mech 2020; 13:dmm043562. [PMID: 32224495 PMCID: PMC7044454 DOI: 10.1242/dmm.043562] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Muscular dystrophies (MDs) encompass a wide variety of inherited disorders that are characterized by loss of muscle tissue associated with a progressive reduction in muscle function. With a cure lacking for MDs, preclinical developments of therapeutic approaches depend on well-characterized animal models that recapitulate the specific pathology in patients. The mouse is the most widely and extensively used model for MDs, and it has played a key role in our understanding of the molecular mechanisms underlying MD pathogenesis. This has enabled the development of therapeutic strategies. Owing to advancements in genetic engineering, a wide variety of mouse models are available for the majority of MDs. Here, we summarize the characteristics of the most commonly used mouse models for a subset of highly studied MDs, collated into a table. Together with references to key publications describing these models, this brief but detailed overview would be useful for those interested in, or working with, mouse models of MD.
Collapse
Affiliation(s)
- Maaike van Putten
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Erin M Lloyd
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| | - Jessica C de Greef
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Vered Raz
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | | | - Miranda D Grounds
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| |
Collapse
|
30
|
Quattrocelli M, Zelikovich AS, Jiang Z, Peek CB, Demonbreun AR, Kuntz NL, Barish GD, Haldar SM, Bass J, McNally EM. Pulsed glucocorticoids enhance dystrophic muscle performance through epigenetic-metabolic reprogramming. JCI Insight 2019; 4:132402. [PMID: 31852847 PMCID: PMC6975267 DOI: 10.1172/jci.insight.132402] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/13/2019] [Indexed: 12/23/2022] Open
Abstract
In humans, chronic glucocorticoid use is associated with side effects like muscle wasting, obesity, and metabolic syndrome. Intermittent steroid dosing has been proposed in Duchenne Muscular Dystrophy patients to mitigate the side effects seen with daily steroid intake. We evaluated biomarkers from Duchenne Muscular Dystrophy patients, finding that, compared with chronic daily steroid use, weekend steroid use was associated with reduced serum insulin, free fatty acids, and branched chain amino acids, as well as reduction in fat mass despite having similar BMIs. We reasoned that intermittent prednisone administration in dystrophic mice would alter muscle epigenomic signatures, and we identified the coordinated action of the glucocorticoid receptor, KLF15 and MEF2C as mediators of a gene expression program driving metabolic reprogramming and enhanced nutrient utilization. Muscle lacking Klf15 failed to respond to intermittent steroids. Furthermore, coadministration of the histone acetyltransferase inhibitor anacardic acid with steroids in mdx mice eliminated steroid-specific epigenetic marks and abrogated the steroid response. Together, these findings indicate that intermittent, repeated exposure to glucocorticoids promotes performance in dystrophic muscle through an epigenetic program that enhances nutrient utilization.
Collapse
MESH Headings
- Anacardic Acids/administration & dosage
- Animals
- Biomarkers/blood
- Biomarkers/metabolism
- Child
- Cross-Sectional Studies
- Disease Models, Animal
- Drug Therapy, Combination
- Epigenesis, Genetic/drug effects
- Epigenomics
- Gene Expression Regulation/drug effects
- Glucocorticoids/administration & dosage
- Histone Acetyltransferases/antagonists & inhibitors
- Histone Acetyltransferases/metabolism
- Humans
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- MEF2 Transcription Factors/metabolism
- Male
- Metabolomics
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/blood
- Muscular Dystrophy, Duchenne/diagnosis
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Nutrients/blood
- Nutrients/metabolism
- Prednisone/administration & dosage
- Pulse Therapy, Drug
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
| | - Aaron S. Zelikovich
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
| | - Zhen Jiang
- Gladstone Institutes, San Francisco, California, USA
- Amgen Research, South San Francisco, California, USA
| | - Clara Bien Peek
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, NU, Chicago, Illinois, USA
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
| | - Nancy L. Kuntz
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Grant D. Barish
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, NU, Chicago, Illinois, USA
| | - Saptarsi M. Haldar
- Gladstone Institutes, San Francisco, California, USA
- Amgen Research, South San Francisco, California, USA
- Cardiology Division, Department of Medicine, UCSF School of Medicine, San Francisco, California, USA
| | - Joseph Bass
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, NU, Chicago, Illinois, USA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University (NU), Chicago, Illinois, USA
| |
Collapse
|
31
|
Báez-Matus X, Figueroa-Cares C, Gónzalez-Jamett AM, Almarza-Salazar H, Arriagada C, Maldifassi MC, Guerra MJ, Mouly V, Bigot A, Caviedes P, Cárdenas AM. Defects in G-Actin Incorporation into Filaments in Myoblasts Derived from Dysferlinopathy Patients Are Restored by Dysferlin C2 Domains. Int J Mol Sci 2019; 21:ijms21010037. [PMID: 31861684 PMCID: PMC6981584 DOI: 10.3390/ijms21010037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/23/2022] Open
Abstract
Dysferlin is a transmembrane C-2 domain-containing protein involved in vesicle trafficking and membrane remodeling in skeletal muscle cells. However, the mechanism by which dysferlin regulates these cellular processes remains unclear. Since actin dynamics is critical for vesicle trafficking and membrane remodeling, we studied the role of dysferlin in Ca2+-induced G-actin incorporation into filaments in four different immortalized myoblast cell lines (DYSF2, DYSF3, AB320, and ER) derived from patients harboring mutations in the dysferlin gene. As compared with immortalized myoblasts obtained from a control subject, dysferlin expression and G-actin incorporation were significantly decreased in myoblasts from dysferlinopathy patients. Stable knockdown of dysferlin with specific shRNA in control myoblasts also significantly reduced G-actin incorporation. The impaired G-actin incorporation was restored by the expression of full-length dysferlin as well as dysferlin N-terminal or C-terminal regions, both of which contain three C2 domains. DYSF3 myoblasts also exhibited altered distribution of annexin A2, a dysferlin partner involved in actin remodeling. However, dysferlin N-terminal and C-terminal regions appeared to not fully restore such annexin A2 mislocation. Then, our results suggest that dysferlin regulates actin remodeling by a mechanism that does to not involve annexin A2.
Collapse
Affiliation(s)
- Ximena Báez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Cindel Figueroa-Cares
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Arlek M. Gónzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Hugo Almarza-Salazar
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Christian Arriagada
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | - María Constanza Maldifassi
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - María José Guerra
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, UMRS 974, Center for Research in Myology, 75013 Paris, France; (V.M.); (A.B.)
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, UMRS 974, Center for Research in Myology, 75013 Paris, France; (V.M.); (A.B.)
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile;
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago 8370456, Chile
| | - Ana M. Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
- Correspondence: ; Tel.: +56-322-508-052
| |
Collapse
|
32
|
Myoferlin, a Membrane Protein with Emerging Oncogenic Roles. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7365913. [PMID: 31828126 PMCID: PMC6885792 DOI: 10.1155/2019/7365913] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/02/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022]
Abstract
Myoferlin (MYOF), initially identified in muscle cells, is a member of the Ferlin family involved in membrane fusion, membrane repair, and membrane trafficking. Dysfunction of this protein is associated with muscular dysfunction. Recently, a growing body of studies have identified MYOF as an oncogenic protein. It is overexpressed in a variety of human cancers and promotes tumorigenesis, tumor cell motility, proliferation, migration, epithelial to mesenchymal transition, angiogenesis as well as metastasis. Clinically, MYOF overexpression is associated with poor outcome in various cancers. It can serve as a prognostic marker of human malignant disease. MYOF drives the progression of cancer in various processes, including surface receptor transportation, endocytosis, exocytosis, intercellular communication, fit mitochondrial structure maintenance and cell metabolism. Depletion of MYOF demonstrates significant antitumor effects both in vitro and in vivo, suggesting that targeting MYOF may produce promising clinical benefits in the treatment of malignant disease. In the present article, we reviewed the physiological function of MYOF as well as its role in cancer, thus providing a general understanding for further exploration of this protein.
Collapse
|
33
|
Demonbreun AR, Fallon KS, Oosterbaan CC, Bogdanovic E, Warner JL, Sell JJ, Page PG, Quattrocelli M, Barefield DY, McNally EM. Recombinant annexin A6 promotes membrane repair and protects against muscle injury. J Clin Invest 2019; 129:4657-4670. [PMID: 31545299 PMCID: PMC6819108 DOI: 10.1172/jci128840] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022] Open
Abstract
Membrane repair is essential to cell survival. In skeletal muscle, injury often associates with plasma membrane disruption. Additionally, muscular dystrophy is linked to mutations in genes that produce fragile membranes or reduce membrane repair. Methods to enhance repair and reduce susceptibility to injury could benefit muscle in both acute and chronic injury settings. Annexins are a family of membrane-associated Ca2+-binding proteins implicated in repair, and annexin A6 was previously identified as a genetic modifier of muscle injury and disease. Annexin A6 forms the repair cap over the site of membrane disruption. To elucidate how annexins facilitate repair, we visualized annexin cap formation during injury. We found that annexin cap size positively correlated with increasing Ca2+ concentrations. We also found that annexin overexpression promoted external blebs enriched in Ca2+ and correlated with a reduction of intracellular Ca2+ at the injury site. Annexin A6 overexpression reduced membrane injury, consistent with enhanced repair. Treatment with recombinant annexin A6 protected against acute muscle injury in vitro and in vivo. Moreover, administration of recombinant annexin A6 in a model of muscular dystrophy reduced serum creatinine kinase, a biomarker of disease. These data identify annexins as mediators of membrane-associated Ca2+ release during membrane repair and annexin A6 as a therapeutic target to enhance membrane repair capacity.
Collapse
Affiliation(s)
- Alexis R. Demonbreun
- Center for Genetic Medicine, and
- Department of Pharmacology, Northwestern University, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Han S, Cui C, He H, Shen X, Chen Y, Wang Y, Li D, Zhu Q, Yin H. Myoferlin Regulates Wnt/β-Catenin Signaling-Mediated Skeletal Muscle Development by Stabilizing Dishevelled-2 Against Autophagy. Int J Mol Sci 2019; 20:ijms20205130. [PMID: 31623157 PMCID: PMC6829482 DOI: 10.3390/ijms20205130] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 12/02/2022] Open
Abstract
Myoferlin (MyoF), which is a calcium/phospholipid-binding protein expressed in cardiac and muscle tissues, belongs to the ferlin family. While MyoF promotes myoblast differentiation, the underlying mechanisms remain poorly understood. Here, we found that MyoF not only promotes C2C12 myoblast differentiation, but also inhibits muscle atrophy and autophagy. In the present study, we found that myoblasts fail to develop into mature myotubes due to defective differentiation in the absence of MyoF. Meanwhile, MyoF regulates the expression of atrophy-related genes (Atrogin-1 and MuRF1) to rescue muscle atrophy. Furthermore, MyoF interacts with Dishevelled-2 (Dvl-2) to activate canonical Wnt signaling. MyoF facilitates Dvl-2 ubiquitination resistance by reducing LC3-labeled Dvl-2 levels and antagonizing the autophagy system. In conclusion, we found that MyoF plays an important role in myoblast differentiation during skeletal muscle atrophy. At the molecular level, MyoF protects Dvl-2 against autophagy-mediated degradation, thus promoting activation of the Wnt/β-catenin signaling pathway. Together, our findings suggest that MyoF, through stabilizing Dvl-2 and preventing autophagy, regulates Wnt/β-catenin signaling-mediated skeletal muscle development.
Collapse
Affiliation(s)
- Shunshun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Can Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Haorong He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Xiaoxu Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yuqi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
35
|
Kitmitto A, Baudoin F, Cartwright EJ. Cardiomyocyte damage control in heart failure and the role of the sarcolemma. J Muscle Res Cell Motil 2019; 40:319-333. [PMID: 31520263 PMCID: PMC6831538 DOI: 10.1007/s10974-019-09539-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 01/07/2023]
Abstract
The cardiomyocyte plasma membrane, termed the sarcolemma, is fundamental for regulating a myriad of cellular processes. For example, the structural integrity of the cardiomyocyte sarcolemma is essential for mediating cardiac contraction by forming microdomains such as the t-tubular network, caveolae and the intercalated disc. Significantly, remodelling of these sarcolemma microdomains is a key feature in the development and progression of heart failure (HF). However, despite extensive characterisation of the associated molecular and ultrastructural events there is a lack of clarity surrounding the mechanisms driving adverse morphological rearrangements. The sarcolemma also provides protection, and is the cell's first line of defence, against external stresses such as oxygen and nutrient deprivation, inflammation and oxidative stress with a loss of sarcolemma viability shown to be a key step in cell death via necrosis. Significantly, cumulative cell death is also a feature of HF, and is linked to disease progression and loss of cardiac function. Herein, we will review the link between structural and molecular remodelling of the sarcolemma associated with the progression of HF, specifically considering the evidence for: (i) Whether intrinsic, evolutionary conserved, plasma membrane injury-repair mechanisms are in operation in the heart, and (ii) if deficits in key 'wound-healing' proteins (annexins, dysferlin, EHD2 and MG53) may play a yet to be fully appreciated role in triggering sarcolemma microdomain remodelling and/or necrosis. Cardiomyocytes are terminally differentiated with very limited regenerative capability and therefore preserving cell viability and cardiac function is crucially important. This review presents a novel perspective on sarcolemma remodelling by considering whether targeting proteins that regulate sarcolemma injury-repair may hold promise for developing new strategies to attenuate HF progression.
Collapse
Affiliation(s)
- Ashraf Kitmitto
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK.
| | - Florence Baudoin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, AV Hill, Dover Street, Manchester, M13 9PL, UK
| |
Collapse
|
36
|
Nader M. The SLMAP/Striatin complex: An emerging regulator of normal and abnormal cardiac excitation-contraction coupling. Eur J Pharmacol 2019; 858:172491. [PMID: 31233748 DOI: 10.1016/j.ejphar.2019.172491] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 12/01/2022]
Abstract
The excitation-contraction (E-C) module involves a harmonized correspondence between the sarcolemma and the sarcoplasmic reticulum. This is provided by membrane proteins, which primarily shape the caveolae, the T-tubule/Sarcoplasmic reticulum (TT/SR) junction, and the intercalated discs (ICDs). Distortion of either one of these structures impairs myocardial contraction, and subsequently translates into cardiac failure. Thus, detailed studies on the molecular cues of the E-C module are becoming increasingly necessary to pharmacologically eradicate cardiac failure Herein we reviewed the organization of caveolae, TT/SR junctions, and the ICDs in the heart, with special attention to the Sarcolemma Membrane Associated Protein (SLMAP) and striatin (STRN) in cardiac membranes biology and cardiomyocyte contraction. We emphasized on their in vivo and in vitro signaling in cardiac function/dysfunction. SLMAP is a cardiac membrane protein that plays an important role in E-C coupling and the adrenergic response of the heart. Similarly, STRN is a dynamic protein that is also involved in cardiac E-C coupling and ICD-related cardiomyopathies. Both SLMAP and STRN are linked to cardiac conditions, including heart failure, and their role in cardiomyocyte function was elucidated in our laboratory. They interact together in a protein complex that holds therapeutic potentials for cardiac dysfunction. This review is the first of its kind to conceptualize the role of the SLMAP/STRN complex in cardiac function and failure. It provides in depth information on the signaling of these two proteins and projects their interaction as a novel therapeutic target for cardiac failure.
Collapse
Affiliation(s)
- Moni Nader
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, 11533, P.O. Box 50927, Saudi Arabia; Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
37
|
Kiselev A, Vaz R, Knyazeva A, Sergushichev A, Dmitrieva R, Khudiakov A, Jorholt J, Smolina N, Sukhareva K, Fomicheva Y, Mikhaylov E, Mitrofanova L, Predeus A, Sjoberg G, Rudenko D, Sejersen T, Lindstrand A, Kostareva A. Truncating Variant in Myof Gene Is Associated With Limb-Girdle Type Muscular Dystrophy and Cardiomyopathy. Front Genet 2019; 10:608. [PMID: 31297131 PMCID: PMC6607695 DOI: 10.3389/fgene.2019.00608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/11/2019] [Indexed: 11/13/2022] Open
Abstract
Even though genetic studies of individuals with neuromuscular diseases have uncovered the molecular background of many cardiac disorders such as cardiomyopathies and inherited arrhythmic syndromes, the genetic cause of a proportion of cardiomyopathies associated with neuromuscular phenotype still remains unknown. Here, we present an individual with a combination of cardiomyopathy and limb-girdle type muscular dystrophy where whole exome sequencing identified myoferlin (MYOF)-a member of the Ferlin protein family and close homolog of DYSF-as the most likely candidate gene. The disease-causative role of the identified variant c.[2576delG; 2575G>C], p.G859QfsTer8 is supported by functional studies in vitro using the primary patient's skeletal muscle mesenchymal progenitor cells, including both RNA sequencing and morphological studies, as well as recapitulating the muscle phenotype in vivo in zebrafish. We provide the first evidence supporting a role of MYOF in human muscle disease.
Collapse
Affiliation(s)
- Artem Kiselev
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Raquel Vaz
- Department of Molecular Medicine and Surgery, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Knyazeva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Renata Dmitrieva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Aleksandr Khudiakov
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - John Jorholt
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Smolina
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ksenia Sukhareva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Yulia Fomicheva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Evgeny Mikhaylov
- Arrhythmia Department, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Lubov Mitrofanova
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Alexander Predeus
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia.,Bioinformatics Institute, Saint Petersburg, Russia
| | - Gunnar Sjoberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Dmitriy Rudenko
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Thomas Sejersen
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Kostareva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
38
|
Haynes VR, Keenan SN, Bayliss J, Lloyd EM, Meikle PJ, Grounds MD, Watt MJ. Dysferlin deficiency alters lipid metabolism and remodels the skeletal muscle lipidome in mice. J Lipid Res 2019; 60:1350-1364. [PMID: 31203232 DOI: 10.1194/jlr.m090845] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 05/14/2019] [Indexed: 12/15/2022] Open
Abstract
Defects in the gene coding for dysferlin, a membrane-associated protein, affect many tissues, including skeletal muscles, with a resultant myopathy called dysferlinopathy. Dysferlinopathy manifests postgrowth with a progressive loss of skeletal muscle function, early intramyocellular lipid accumulation, and a striking later replacement of selective muscles by adipocytes. To better understand the changes underpinning this disease, we assessed whole-body energy homeostasis, skeletal muscle fatty acid metabolism, lipolysis in adipose tissue, and the skeletal muscle lipidome using young adult dysferlin-deficient male BLAJ mice and age-matched C57Bl/6J WT mice. BLAJ mice had increased lean mass and reduced fat mass associated with increased physical activity and increased adipose tissue lipolysis. Skeletal muscle fatty acid metabolism was remodeled in BLAJ mice, characterized by a partitioning of fatty acids toward storage rather than oxidation. Lipidomic analysis identified marked changes in almost all lipid classes examined in the skeletal muscle of BLAJ mice, including sphingolipids, phospholipids, cholesterol, and most glycerolipids but, surprisingly, not triacylglycerol. These observations indicate that an early manifestation of dysferlin deficiency is the reprogramming of skeletal muscle and adipose tissue lipid metabolism, which is likely to contribute to the progressive adverse histopathology in dysferlinopathies.
Collapse
Affiliation(s)
- Vanessa R Haynes
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Stacey N Keenan
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Jackie Bayliss
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Erin M Lloyd
- School of Human Sciences University of Western Australia, Perth, Australia
| | - Peter J Meikle
- Metabolomics Laboratory Baker Heart Institute, Melbourne, Australia
| | - Miranda D Grounds
- School of Human Sciences University of Western Australia, Perth, Australia
| | - Matthew J Watt
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| |
Collapse
|
39
|
An expanded proteome of cardiac t-tubules. Cardiovasc Pathol 2019; 42:15-20. [PMID: 31202980 DOI: 10.1016/j.carpath.2019.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/29/2019] [Accepted: 05/17/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Transverse tubules (t-tubules) are important structural elements, derived from sarcolemma, found on all striated myocytes. These specialized organelles create a scaffold for many proteins crucial to the effective propagation of signal in cardiac excitation-contraction coupling. The full protein composition of this region is unknown. METHODS We characterized the t-tubule subproteome using 52,033 immunohistochemical images covering 13,203 proteins from the Human Protein Atlas (HPA) cardiac tissue microarrays. We used HPASubC, a suite of Python tools, to rapidly review and classify each image for a specific t-tubule staining pattern. The tools Gene Cards, String 11, and Gene Ontology Consortium as well as literature searches were used to understand pathways and relationships between the proteins. RESULTS There were 96 likely t-tubule proteins identified by HPASubC. Of these, 12 were matrisome proteins and 3 were mitochondrial proteins. A separate literature search identified 50 known t-tubule proteins. A comparison of the 2 lists revealed only 17 proteins in common, including 8 of the matrisome proteins. String11 revealed that 94 of 127 combined t-tubule proteins generated a single interconnected network. CONCLUSION Using HPASubC and the HPA, we identified 78 novel, putative t-tubule proteins and validated 17 within the literature. This expands and improves our knowledge of this important subcellular structure of the cardiac myocyte. This information can be used to identify new structural targets involved in excitation-contraction coupling that may be altered in disease.
Collapse
|
40
|
Dysferlin-deficiency has greater impact on function of slow muscles, compared with fast, in aged BLAJ mice. PLoS One 2019; 14:e0214908. [PMID: 30970035 PMCID: PMC6457631 DOI: 10.1371/journal.pone.0214908] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/24/2019] [Indexed: 12/26/2022] Open
Abstract
Dysferlinopathies are a form of muscular dystrophy caused by gene mutations resulting in deficiency of the protein dysferlin. Symptoms manifest later in life in a muscle specific manner, although the pathomechanism is not well understood. This study compared the impact of dysferlin-deficiency on in vivo and ex vivo muscle function, and myofibre type composition in slow (soleus) and fast type (extensor digitorum longus; EDL) muscles using male dysferlin-deficient (dysf-/-) BLAJ mice aged 10 months, compared with wild type (WT) C57Bl/6J mice. There was a striking increase in muscle mass of BLAJ soleus (+25%) (p<0.001), with no strain differences in EDL mass, compared with WT. In vivo measures of forelimb grip strength and wheel running capacity showed no strain differences. Ex vivo measures showed the BLAJ soleus had faster twitch contraction (-21%) and relaxation (-20%) times, and delayed post fatigue recovery (ps<0.05); whereas the BLAJ EDL had a slower relaxation time (+11%) and higher maximum rate of force production (+25%) (ps<0.05). Similar proportions of MHC isoforms were evident in the soleus muscles of both strains (ps>0.05); however, for the BLAJ EDL, there was an increased proportion of type IIx MHC isoform (+5.5%) and decreased type IIb isoform (-5.5%) (ps<0.01). This identification of novel differences in the impact of dysferlin-deficiency on slow and fast twitch muscles emphasises the importance of evaluating myofibre type specific effects to provide crucial insight into the mechanisms responsible for loss of function in dysferlinopathies; this is critical for the development of targeted future clinical therapies.
Collapse
|
41
|
Bonventre JA, Holman C, Manchanda A, Codding SJ, Chau T, Huegel J, Barton C, Tanguay R, Johnson CP. Fer1l6 is essential for the development of vertebrate muscle tissue in zebrafish. Mol Biol Cell 2018; 30:293-301. [PMID: 30516436 PMCID: PMC6589578 DOI: 10.1091/mbc.e18-06-0401] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The precise spatial and temporal expression of genes is essential for proper organismal development. Despite their importance, however, many developmental genes have yet to be identified. We have determined that Fer1l6, a member of the ferlin family of genes, is a novel factor in zebrafish development. We find that Fer1l6 is expressed broadly in the trunk and head of zebrafish larvae and is more restricted to gills and female gonads in adult zebrafish. Using both genetic mutant and morpholino knockdown models, we found that loss of Fer1l6 led to deformation of striated muscle tissues, delayed development of the heart, and high morbidity. Further, expression of genes associated with muscle cell proliferation and differentiation were affected. Fer1l6 was also detected in the C2C12 cell line, and unlike other ferlin homologues, we found Fer1l6 expression was independent of the myoblast-to-myotube transition. Finally, analysis of cell and recombinant protein-based assays indicate that Fer1l6 colocalizes with syntaxin 4 and vinculin, and that the putative C2 domains interact with lipid membranes. We conclude that Fer1l6 has diverged from other vertebrate ferlins to play an essential role in zebrafish skeletal and cardiac muscle development.
Collapse
Affiliation(s)
- Josephine A Bonventre
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| | - Chelsea Holman
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| | - Aayushi Manchanda
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, OR 97331
| | - Sara J Codding
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| | - Trisha Chau
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| | - Jacob Huegel
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| | - Carrie Barton
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Robert Tanguay
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Colin P Johnson
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331.,Molecular and Cellular Biology Program, Oregon State University, Corvallis, OR 97331
| |
Collapse
|
42
|
De La Mata A, Tajada S, O'Dwyer S, Matsumoto C, Dixon RE, Hariharan N, Moreno CM, Santana LF. BIN1 Induces the Formation of T-Tubules and Adult-Like Ca 2+ Release Units in Developing Cardiomyocytes. Stem Cells 2018; 37:54-64. [PMID: 30353632 PMCID: PMC6312737 DOI: 10.1002/stem.2927] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/29/2018] [Accepted: 09/22/2018] [Indexed: 12/26/2022]
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) are at the center of new cell-based therapies for cardiac disease, but may also serve as a useful in vitro model for cardiac cell development. An intriguing feature of hESC-CMs is that although they express contractile proteins and have sarcomeres, they do not develop transverse-tubules (T-tubules) with adult-like Ca2+ release units (CRUs). We tested the hypothesis that expression of the protein BIN1 in hESC-CMs promotes T-tubules formation, facilitates CaV 1.2 channel clustering along the tubules, and results in the development of stable CRUs. Using electrophysiology, [Ca2+ ]i imaging, and super resolution microscopy, we found that BIN1 expression induced T-tubule development in hESC-CMs, while increasing differentiation toward a more ventricular-like phenotype. Voltage-gated CaV 1.2 channels clustered along the surface sarcolemma and T-tubules of hESC-CM. The length and width of the T-tubules as well as the expression and size of CaV 1.2 clusters grew, as BIN1 expression increased and cells matured. BIN1 expression increased CaV 1.2 channel activity and the probability of coupled gating within channel clusters. Interestingly, BIN1 clusters also served as sites for sarcoplasmic reticulum (SR) anchoring and stabilization. Accordingly, BIN1-expressing cells had more CaV 1.2-ryanodine receptor junctions than control cells. This was associated with larger [Ca2+ ]i transients during excitation-contraction coupling. Our data support the view that BIN1 is a key regulator of T-tubule formation and CaV 1.2 channel delivery. By studying the role of BIN1 during the differentiation of hESC-CMs, we show that BIN1 is also important for CaV 1.2 channel clustering, junctional SR organization, and the establishment of excitation-contraction coupling. Stem Cells 2019;37:54-64.
Collapse
Affiliation(s)
- Ana De La Mata
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| | - Sendoa Tajada
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| | - Samantha O'Dwyer
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| | - Collin Matsumoto
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| | - Rose E Dixon
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| | - Nirmala Hariharan
- Department of Pharmacology, University of California School of Medicine, Davis, California, USA
| | - Claudia M Moreno
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| | - Luis Fernando Santana
- Department of Physiology & Membrane Biology, University of California School of Medicine, Davis, California, USA
| |
Collapse
|
43
|
Maani N, Sabha N, Rezai K, Ramani A, Groom L, Eltayeb N, Mavandadnejad F, Pang A, Russo G, Brudno M, Haucke V, Dirksen RT, Dowling JJ. Tamoxifen therapy in a murine model of myotubular myopathy. Nat Commun 2018; 9:4849. [PMID: 30451841 PMCID: PMC6242823 DOI: 10.1038/s41467-018-07057-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Myotubular myopathy (MTM) is a severe X-linked disease without existing therapies. Here, we show that tamoxifen ameliorates MTM-related histopathological and functional abnormalities in mice, and nearly doubles survival. The beneficial effects of tamoxifen are mediated primarily via estrogen receptor signaling, as demonstrated through in vitro studies and in vivo phenotypic rescue with estradiol. RNA sequencing and protein expression analyses revealed that rescue is mediated in part through post-transcriptional reduction of dynamin-2, a known MTM modifier. These findings demonstrate an unexpected ability of tamoxifen to improve the murine MTM phenotype, providing preclinical evidence to support clinical translation.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Dynamin II/genetics
- Dynamin II/metabolism
- Estradiol/metabolism
- Estradiol/pharmacology
- Excitation Contraction Coupling/drug effects
- Female
- Gene Expression/drug effects
- High-Throughput Nucleotide Sequencing
- Humans
- Longevity/drug effects
- Male
- Mice
- Mice, Knockout
- Motor Activity/drug effects
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myofibrils/drug effects
- Myofibrils/metabolism
- Myofibrils/ultrastructure
- Myopathies, Structural, Congenital/drug therapy
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/pathology
- Protective Agents/pharmacology
- Protein Tyrosine Phosphatases, Non-Receptor/deficiency
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Tamoxifen/pharmacology
Collapse
Affiliation(s)
- Nika Maani
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Medical Science Building, Room 4386, 1 King's College Cir, Toronto, ON, CAN M5S 1A8, Canada
| | - Nesrin Sabha
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Medical Science Building, Room 4386, 1 King's College Cir, Toronto, ON, CAN M5S 1A8, Canada
- Department of Paediatrics, University of Toronto, Room 1436D, 555 University Avenue, Toronto, ON, CAN M5G 1X8, Canada
| | - Kamran Rezai
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Medical Science Building, Room 4386, 1 King's College Cir, Toronto, ON, CAN M5S 1A8, Canada
| | - Arun Ramani
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
- Department of Computer Science, University of Toronto, Pratt Building Room 286C, 6 King's College Rd, Toronto, ON, CAN M5S 3G4, Canada
- Centre for Computational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center School of Medicine and Dentistry, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA
| | - Nadine Eltayeb
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
| | - Faranak Mavandadnejad
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
| | - Andrea Pang
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
| | - Giulia Russo
- Department of Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Michael Brudno
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
- Department of Computer Science, University of Toronto, Pratt Building Room 286C, 6 King's College Rd, Toronto, ON, CAN M5S 3G4, Canada
- Centre for Computational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center School of Medicine and Dentistry, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON, CAN M5G 0A4, Canada.
- Department of Molecular Genetics, University of Toronto, Medical Science Building, Room 4386, 1 King's College Cir, Toronto, ON, CAN M5S 1A8, Canada.
- Department of Paediatrics, University of Toronto, Room 1436D, 555 University Avenue, Toronto, ON, CAN M5G 1X8, Canada.
| |
Collapse
|
44
|
Murphy S, Zweyer M, Mundegar RR, Swandulla D, Ohlendieck K. Chemical crosslinking analysis of β-dystroglycan in dystrophin-deficient skeletal muscle. HRB Open Res 2018; 1:17. [PMID: 35528858 PMCID: PMC9039762 DOI: 10.12688/hrbopenres.12846.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2018] [Indexed: 12/19/2022] Open
Abstract
Background: In Duchenne muscular dystrophy, primary abnormalities in the membrane cytoskeletal protein dystrophin trigger the loss of sarcolemmal linkage between the extracellular matrix component laminin-211 and the intracellular cortical actin membrane cytoskeleton. The disintegration of the dystrophin-associated glycoprotein complex renders the plasma membrane of contractile fibres more susceptible to micro-rupturing, which is associated with abnormal calcium handling and impaired cellular signalling in dystrophinopathy. Methods: The oligomerisation pattern of β-dystroglycan, an integral membrane protein belonging to the core dystrophin complex, was studied using immunoprecipitation and chemical crosslinking analysis. A homo-bifunctional and non-cleavable agent with water-soluble and amine-reactive properties was employed to study protein oligomerisation in normal versus dystrophin-deficient skeletal muscles. Crosslinker-induced protein oligomerisation was determined by a combination of gel-shift analysis and immunoblotting. Results: Although proteomics was successfully applied for the identification of dystroglycan as a key component of the dystrophin-associated glycoprotein complex in the muscle membrane fraction, mass spectrometric analysis did not efficiently recognize this relatively low-abundance protein after immunoprecipitation or chemical crosslinking. As an alternative approach, comparative immunoblotting was used to evaluate the effects of chemical crosslinking. Antibody decoration of the crosslinked microsomal protein fraction from wild type versus the
mdx-4cv mouse model of dystrophinopathy revealed oligomers that contain β-dystroglycan. The protein exhibited a comparable reduction in gel electrophoretic mobility in both normal and dystrophic samples. The membrane repair proteins dysferlin and myoferlin, which are essential components of fibre regeneration, as well as the caveolae-associated protein cavin-1, were also shown to exist in high-molecular mass complexes. Conclusions: The muscular dystrophy-related reduction in the concentration of β-dystroglycan, which forms in conjunction with its extracellular binding partner α-dystroglycan a critical plasmalemmal receptor for laminin-211, does not appear to alter its oligomeric status. Thus, independent of direct interactions with dystrophin, this sarcolemmal glycoprotein appears to exist in a supramolecular assembly in muscle.
Collapse
Affiliation(s)
- Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- Institute of Physiology II, University of Bonn, Bonn, D‑53115, Germany
| | | | - Dieter Swandulla
- Institute of Physiology II, University of Bonn, Bonn, D‑53115, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
45
|
Murphy S, Zweyer M, Mundegar RR, Swandulla D, Ohlendieck K. Chemical crosslinking analysis of β-dystroglycan in dystrophin-deficient skeletal muscle. HRB Open Res 2018; 1:17. [PMID: 35528858 PMCID: PMC9039762 DOI: 10.12688/hrbopenres.12846.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2018] [Indexed: 07/30/2023] Open
Abstract
Background: In Duchenne muscular dystrophy, primary abnormalities in the membrane cytoskeletal protein dystrophin trigger the loss of sarcolemmal linkage between the extracellular matrix component laminin-211 and the intracellular cortical actin membrane cytoskeleton. The disintegration of the dystrophin-associated glycoprotein complex renders the plasma membrane of contractile fibres more susceptible to micro-rupturing, which is associated with abnormal calcium handling and impaired cellular signalling in dystrophinopathy. Methods: The oligomerisation pattern of β-dystroglycan, an integral membrane protein belonging to the core dystrophin complex, was studied using immunoprecipitation and chemical crosslinking analysis. A homo-bifunctional and non-cleavable agent with water-soluble and amine-reactive properties was employed to study protein oligomerisation in normal versus dystrophin-deficient skeletal muscles. Crosslinker-induced protein oligomerisation was determined by a combination of gel-shift analysis and immunoblotting. Results: Although proteomics was successfully applied for the identification of dystroglycan as a key component of the dystrophin-associated glycoprotein complex in the muscle membrane fraction, mass spectrometric analysis did not efficiently recognize this relatively low-abundance protein after immunoprecipitation or chemical crosslinking. As an alternative approach, comparative immunoblotting was used to evaluate the effects of chemical crosslinking. Antibody decoration of the crosslinked microsomal protein fraction from wild type versus the mdx-4cv mouse model of dystrophinopathy revealed oligomers that contain β-dystroglycan. The protein exhibited a comparable reduction in gel electrophoretic mobility in both normal and dystrophic samples. The membrane repair proteins dysferlin and myoferlin, which are essential components of fibre regeneration, as well as the caveolae-associated protein cavin-1, were also shown to exist in high-molecular mass complexes. Conclusions: The muscular dystrophy-related reduction in the concentration of β-dystroglycan, which forms in conjunction with its extracellular binding partner α-dystroglycan a critical plasmalemmal receptor for laminin-211, does not appear to alter its oligomeric status. Thus, independent of direct interactions with dystrophin, this sarcolemmal glycoprotein appears to exist in a supramolecular assembly in muscle.
Collapse
Affiliation(s)
- Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- Institute of Physiology II, University of Bonn, Bonn, D‑53115, Germany
| | | | - Dieter Swandulla
- Institute of Physiology II, University of Bonn, Bonn, D‑53115, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
46
|
Begam M, Collier AF, Mueller AL, Roche R, Galen SS, Roche JA. Diltiazem improves contractile properties of skeletal muscle in dysferlin-deficient BLAJ mice, but does not reduce contraction-induced muscle damage. Physiol Rep 2018; 6:e13727. [PMID: 29890050 PMCID: PMC5995314 DOI: 10.14814/phy2.13727] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/06/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
B6.A-Dysfprmd /GeneJ (BLAJ) mice model human limb-girdle muscular dystrophy 2B (LGMD2B), which is linked to mutations in the dysferlin (DYSF) gene. We tested the hypothesis that, the calcium ion (Ca2+ ) channel blocker diltiazem (DTZ), reduces contraction-induced skeletal muscle damage, in BLAJ mice. We randomly assigned mice (N = 12; 3-4 month old males) to one of two groups - DTZ (N = 6) or vehicle (VEH, distilled water, N = 6). We conditioned mice with either DTZ or VEH for 1 week, after which, their tibialis anterior (TA) muscles were tested for contractile torque and susceptibility to injury from forced eccentric contractions. We continued dosing with DTZ or VEH for 3 days following eccentric contractions, and then studied torque recovery and muscle damage. We analyzed contractile torque before eccentric contractions, immediately after eccentric contractions, and at 3 days after eccentric contractions; and counted damaged fibers in the injured and uninjured TA muscles. We found that DTZ improved contractile torque before and immediately after forced eccentric contractions, but did not reduce delayed-onset muscle damage that was observed at 3 days after eccentric contractions.
Collapse
Affiliation(s)
- Morium Begam
- Physical Therapy ProgramDepartment of Health Care SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichigan
| | - Alyssa F. Collier
- Program in Physical TherapyWashington University in St. Louis School of MedicineSt. LouisMissouri
| | - Amber L. Mueller
- Program in Molecular MedicineUniversity of Maryland School of MedicineBaltimoreMaryland
| | - Renuka Roche
- Eastern Michigan University School of Health SciencesYpsilantiMichigan
| | - Sujay S. Galen
- Physical Therapy ProgramDepartment of Health Care SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichigan
| | - Joseph A. Roche
- Physical Therapy ProgramDepartment of Health Care SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichigan
| |
Collapse
|
47
|
Mahdy MAA. Glycerol-induced injury as a new model of muscle regeneration. Cell Tissue Res 2018; 374:233-241. [DOI: 10.1007/s00441-018-2846-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/24/2018] [Indexed: 01/15/2023]
|
48
|
Murphy S, Zweyer M, Mundegar RR, Swandulla D, Ohlendieck K. Comparative gel-based proteomic analysis of chemically crosslinked complexes in dystrophic skeletal muscle. Electrophoresis 2018; 39:1735-1744. [PMID: 29679381 PMCID: PMC6099379 DOI: 10.1002/elps.201800028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 03/08/2018] [Accepted: 04/19/2018] [Indexed: 12/18/2022]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disease with a complex pathophysiology that is based on primary abnormalities in the dystrophin gene. In order to study potential changes in the oligomerization of high-molecular-mass protein complexes in dystrophic skeletal muscle, chemical crosslinking was combined with mass spectrometric analysis. The biochemical stabilization of protein interactions was carried out with the homo-bifunctional and amine-reactive agent bis[sulfosuccinimidyl]suberate, followed by protein shift analysis in one-dimensional gels. The proteomic approach identified 11 and 15 protein species in wild type versus dystrophic microsomal fractions, respectively, as well as eight common proteins, with an electrophoretic mobility shift to very high molecular mass following chemical crosslinking. In dystrophin-deficient preparations, several protein species with an increased tendency of oligomerisation were identified as components of the sarcolemma and its associated intra- and extracellular structures, as well as mitochondria. This included the sarcolemmal proteins myoferlin and caveolin, the cytoskeletal components vimentin and tubulin, extracellular collagen alpha-1(XII) and the mitochondrial trifunctional enzyme and oxoglutarate dehydrogenase. These changes are probably related to structural and metabolic adaptations, especially cellular repair processes, which agrees with the increased oligomerisation of myosin-3, myosin-9 and actin, and their role in cellular regeneration and structural adjustments in dystrophinopathy.
Collapse
Affiliation(s)
- Sandra Murphy
- Department of BiologyMaynooth UniversityNational University of IrelandMaynoothCo. KildareIreland
| | - Margit Zweyer
- Institute of Physiology IIUniversity of BonnBonnGermany
| | | | | | - Kay Ohlendieck
- Department of BiologyMaynooth UniversityNational University of IrelandMaynoothCo. KildareIreland
| |
Collapse
|
49
|
Schiaffino S. Knockout of human muscle genes revealed by large scale whole-exome studies. Mol Genet Metab 2018; 123:411-415. [PMID: 29452748 DOI: 10.1016/j.ymgme.2018.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
Large scale whole-exome sequence studies have revealed that a number of individuals from different populations have predicted loss-of-function of different genes due to nonsense, frameshift, or canonical splice-site mutations. Surprisingly, many of these mutations do not apparently show the deleterious phenotypic consequences expected from gene knockout. These homozygous null mutations, when confirmed, can provide insight into human gene function and suggest novel approaches to correct gene dysfunction, as the lack of the expected disease phenotype may reflect the existence of modifier genes that reveal potential therapeutic targets. Human knockouts complement the information derived from mouse knockouts, which are not always good models of human disease. We have examined human knockout datasets searching for genes expressed exclusively or predominantly in striated muscle. A number of well-known muscle genes was found in one or more datasets, including genes coding for sarcomeric myosins, components of the sarcomeric cytoskeleton, sarcoplasmic reticulum and plasma membrane, and enzymes involved in muscle metabolism. The surprising absence of phenotype in some of these human knockouts is critically discussed, focusing on the comparison with the corresponding mouse knockouts.
Collapse
|
50
|
Murphy S, Zweyer M, Henry M, Meleady P, Mundegar RR, Swandulla D, Ohlendieck K. Proteomic analysis of the sarcolemma-enriched fraction from dystrophic mdx-4cv skeletal muscle. J Proteomics 2018; 191:212-227. [PMID: 29408692 DOI: 10.1016/j.jprot.2018.01.015] [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: 10/16/2017] [Revised: 01/12/2018] [Accepted: 01/28/2018] [Indexed: 02/07/2023]
Abstract
The highly progressive neuromuscular disorder dystrophinopathy is triggered by primary abnormalities in the Dmd gene, which causes cytoskeletal instability and loss of sarcolemmal integrity. Comparative organellar proteomics was employed to identify sarcolemma-associated proteins with an altered concentration in dystrophic muscle tissue from the mdx-4cv mouse model of dystrophinopathy. A lectin agglutination method was used to prepare a sarcolemma-enriched fraction and resulted in the identification of 190 significantly changed protein species. Proteomics established differential expression patterns for key components of the muscle plasma membrane, cytoskeletal network, extracellular matrix, metabolic pathways, cellular stress response, protein synthesis, immune response and neuromuscular junction. The deficiency in dystrophin and drastic reduction in dystrophin-associated proteins appears to trigger (i) enhanced membrane repair involving myoferlin, dysferlin and annexins, (ii) increased protein synthesis and the compensatory up-regulation of cytoskeletal proteins, (iii) the decrease in the scaffolding protein periaxin and myelin PO involved in myelination of motor neurons, (iv) complex changes in bioenergetic pathways, (v) elevated levels of molecular chaperones to prevent proteotoxic effects, (vi) increased collagen deposition causing reactive myofibrosis, (vii) disturbed ion homeostasis at the sarcolemma and associated membrane systems, and (viii) a robust inflammatory response by the innate immune system in response to chronic muscle damage. SIGNIFICANCE: Duchenne muscular dystrophy is a devastating muscle wasting disease and represents the most frequently inherited neuromuscular disorder in humans. Genetic abnormalities in the Dmd gene cause a loss of sarcolemmal integrity and highly progressive muscle fibre degeneration. Changes in the neuromuscular system are associated with necrosis, fibrosis and inflammation. In order to evaluate secondary changes in the sarcolemma membrane system due to the lack of the membrane cytoskeletal protein dystrophin, comparative organellar proteomics was used to study the mdx-4cv mouse model of dystrophinopathy. Mass spectrometric analyses identified a variety of altered components of the extracellular matrix-sarcolemma-cytoskeleton axis in dystrophic muscles. This included proteins involved in membrane repair, cytoskeletal restoration, calcium homeostasis, cellular signalling, stress response, neuromuscular transmission and reactive myofibrosis, as well as immune cell infiltration. These pathobiochemical alterations agree with the idea of highly complex secondary changes in X-linked muscular dystrophy and support the concept that micro-rupturing of the dystrophin-deficient plasma membrane is at the core of muscle wasting pathology.
Collapse
Affiliation(s)
- Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- Institute of Physiology II, University of Bonn, D-53115 Bonn, Germany
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Rustam R Mundegar
- Institute of Physiology II, University of Bonn, D-53115 Bonn, Germany
| | - Dieter Swandulla
- Institute of Physiology II, University of Bonn, D-53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|