1
|
Wang X, Wang X, Ma J, Zhang S, Fang W, Xu F, Du J, Liang H, Duan W, Li Z, Liu J. GPR30 Agonist G1 Mitigates Sepsis-Induced Cardiac Dysfunction by Inhibiting ACE2/c-FOS-Mediated Necroptosis in Female Mice. ACS Infect Dis 2024; 10:3797-3809. [PMID: 39377746 DOI: 10.1021/acsinfecdis.4c00319] [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] [Indexed: 10/09/2024]
Abstract
Sepsis is a severe inflammatory syndrome with high mortality and morbidity. Sepsis-induced myocardial dysfunction (SIMD) is a common cause of death in sepsis. The female sex is less susceptible to sepsis-related organ dysfunction, although the underlying mechanism of this sex difference remains unclear. This study explored the role of estrogen receptor G protein-coupled estrogen receptor 30 (GPR30) in septic cardiac dysfunction. Results from the present study indicated that GPR30 activation by the G1 agonist protected female mouse hearts against SIMD exposed to lipopolysaccharides. However, this beneficial effect was absent in female ACE2-knockout mice, as demonstrated by poorer cardiac contractility, myocardial injury, and necroptosis. We also demonstrated that the Stat6 transcription factor induced ace2 transcription by enhancing its promoter activity under GPR30 activation in septic hearts. The adenovirus-mediated inhibition of ACE2 targeting c-FOS expression reversed the deterioration, restored cardiac function, and improved survival in female ACE2-knockout mice. These results demonstrate the essential role of GPR30/STAT6/ACE2/c-FOS-mediated necroptosis in G1-mediated protection and provide novel insight into the pathogenesis of sepsis-related organ damage.
Collapse
Affiliation(s)
- Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Xiaoya Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Jipeng Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Shuaishuai Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Weiyi Fang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
- Graduate School, Xi'an Medical University, Xi'an 710021, PR China
| | - Fujie Xu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
- Graduate School, Xi'an Medical University, Xi'an 710021, PR China
| | - Jun Du
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
- Graduate School, Xi'an Medical University, Xi'an 710021, PR China
| | - Hongliang Liang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, United States
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Zilin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| |
Collapse
|
2
|
Zarén P, Gawlik KI. Thrombospondin-4 deletion does not exacerbate muscular dystrophy in β-sarcoglycan-deficient and laminin α2 chain-deficient mice. Sci Rep 2024; 14:14757. [PMID: 38926599 PMCID: PMC11208443 DOI: 10.1038/s41598-024-65473-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024] Open
Abstract
Muscular dystrophy is a group of genetic disorders that lead to muscle wasting and loss of muscle function. Identifying genetic modifiers that alleviate symptoms or enhance the severity of a primary disease helps to understand mechanisms behind disease pathology and facilitates discovery of molecular targets for therapy. Several muscular dystrophies are caused by genetic defects in the components of the dystrophin-glycoprotein adhesion complex (DGC). Thrombospondin-4 overexpression has been shown to mitigate dystrophic disease in mouse models for Duchenne muscular dystrophy (dystrophin deficiency) and limb-girdle muscular dystrophy type 2F (LGMD2F, δ-sarcoglycan deficiency), while deletion of the thrombospondin-4 gene exacerbated the diseases. Hence, thrombospondin-4 has been considered a candidate molecule for therapy of muscular dystrophies involving the DGC. We have investigated whether thrombospondin-4 could act as a genetic modifier for other DGC-associated diseases: limb-girdle muscular dystrophy type 2E (LGMD2E, β-sarcoglycan deficiency) and laminin α2 chain-deficient muscular dystrophy (LAMA2-RD). Deletion of the thrombospondin-4 gene in mouse models for LGMD2E and LAMA2-RD, respectively, did not result in worsening of the dystrophic phenotype. Loss of thrombospondin-4 did not enhance sarcolemma damage and did not impair trafficking of transmembrane receptors integrin α7β1 and dystroglycan in double knockout muscles. Our results suggest that thrombospondin-4 might not be a relevant therapeutic target for all muscular dystrophies involving the DGC. This data also demonstrates that molecular pathology between very similar diseases like LGMD2E and 2F can differ significantly.
Collapse
Affiliation(s)
- Paula Zarén
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, BMC C12, 221 84, Lund, Sweden
| | - Kinga I Gawlik
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, BMC C12, 221 84, Lund, Sweden.
| |
Collapse
|
3
|
Elasbali AM, Al-Soud WA, Anwar S, Alhassan HH, Adnan M, Hassan MI. A review on mechanistic insights into structure and function of dystrophin protein in pathophysiology and therapeutic targeting of Duchenne muscular dystrophy. Int J Biol Macromol 2024; 264:130544. [PMID: 38428778 DOI: 10.1016/j.ijbiomac.2024.130544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/09/2024] [Accepted: 02/28/2024] [Indexed: 03/03/2024]
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive genetic disorder characterized by progressive and severe muscle weakening and degeneration. Among the various forms of muscular dystrophy, it stands out as one of the most common and impactful, predominantly affecting boys. The condition arises due to mutations in the dystrophin gene, a key player in maintaining the structure and function of muscle fibers. The manuscript explores the structural features of dystrophin protein and their pivotal roles in DMD. We present an in-depth analysis of promising therapeutic approaches targeting dystrophin and their implications for the therapeutic management of DMD. Several therapies aiming to restore dystrophin protein or address secondary pathology have obtained regulatory approval, and many others are ongoing clinical development. Notably, recent advancements in genetic approaches have demonstrated the potential to restore partially functional dystrophin forms. The review also provides a comprehensive overview of the status of clinical trials for major therapeutic genetic approaches for DMD. In addition, we have summarized the ongoing therapeutic approaches and advanced mechanisms of action for dystrophin restoration and the challenges associated with DMD therapeutics.
Collapse
Affiliation(s)
- Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Waleed Abu Al-Soud
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Sakaka, Saudi Arabia; Molekylärbiologi, Klinisk Mikrobiologi och vårdhygien, Region Skåne, Sölvegatan 23B, 221 85 Lund, Sweden
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Hassan H Alhassan
- Department of Clinical Laboratory Science, College of Applied Sciences-Sakaka, Jouf University, Sakaka, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
4
|
Bencze M, Periou B, Punzón I, Barthélémy I, Taglietti V, Hou C, Zaidan L, Kefi K, Blot S, Agbulut O, Gervais M, Derumeaux G, Authier F, Tiret L, Relaix F. Receptor interacting protein kinase-3 mediates both myopathy and cardiomyopathy in preclinical animal models of Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2023; 14:2520-2531. [PMID: 37909859 PMCID: PMC10751447 DOI: 10.1002/jcsm.13265] [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: 09/06/2022] [Revised: 03/27/2023] [Accepted: 04/24/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive muscle degenerative disorder, culminating in a complete loss of ambulation, hypertrophic cardiomyopathy and a fatal cardiorespiratory failure. Necroptosis is the form of necrosis that is dependent upon the receptor-interacting protein kinase (RIPK) 3; it is involved in several inflammatory and neurodegenerative conditions. We previously identified RIPK3 as a key player in the acute myonecrosis affecting the hindlimb muscles of the mdx dystrophic mouse model. Whether necroptosis also mediates respiratory and heart disorders in DMD is currently unknown. METHODS Evidence of activation of the necroptotic axis was examined in dystrophic tissues from Golden retriever muscular dystrophy (GRMD) dogs and R-DMDdel52 rats. A functional assessment of the involvement of necroptosis in dystrophic animals was performed on mdx mice that were genetically depleted for RIPK3. Dystrophic mice aged from 12 to 18 months were analysed by histology and molecular biology to compare the phenotype of muscles from mdxRipk3+/+ and mdxRipk3-/- mice. Heart function was also examined by echocardiography in 40-week-old mice. RESULTS RIPK3 expression in sartorius and biceps femoris muscles from GRMD dogs positively correlated to myonecrosis levels (r = 0.81; P = 0.0076). RIPK3 was also found elevated in the diaphragm (P ≤ 0.05). In the slow-progressing heart phenotype of GRMD dogs, the phosphorylated form of RIPK1 at the Serine 161 site was dramatically increased in cardiomyocytes. A similar p-RIPK1 upregulation characterized the cardiomyocytes of the severe DMDdel52 rat model, associated with a marked overexpression of Ripk1 (P = 0.007) and Ripk3 (P = 0.008), indicating primed activation of the necroptotic pathway in the dystrophic heart. MdxRipk3-/- mice displayed decreased compensatory hypertrophy of the heart (P = 0.014), and echocardiography showed a 19% increase in the relative wall thickness (P < 0.05) and 29% reduction in the left ventricle mass (P = 0.0144). Besides, mdxRipk3-/- mice presented no evidence of a regenerative default or sarcopenia in skeletal muscles, moreover around 50% less affected by fibrosis (P < 0.05). CONCLUSIONS Our data highlight molecular and histological evidence that the necroptotic pathway is activated in degenerative tissues from dystrophic animal models, including the diaphragm and the heart. We also provide the genetic proof of concept that selective inhibition of necroptosis in dystrophic condition improves both histological features of muscles and cardiac function, suggesting that prevention of necroptosis is susceptible to providing multiorgan beneficial effects for DMD.
Collapse
Affiliation(s)
- Maximilien Bencze
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Baptiste Periou
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Isabel Punzón
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Inès Barthélémy
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Valentina Taglietti
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Cyrielle Hou
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Louai Zaidan
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Kaouthar Kefi
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Stéphane Blot
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Onnik Agbulut
- Institut de Biologie Paris‐Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and AgeingSorbonne UniversitéParisFrance
| | - Marianne Gervais
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Geneviève Derumeaux
- Team Derumeaux, Department of Physiology, Henri Mondor Hospital, FHU‐SENEC, AP‐HPU955‐IMRB, Université Paris‐Est Créteil (UPEC)CréteilFrance
| | - François‐Jérôme Authier
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Laurent Tiret
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| | - Fréderic Relaix
- Team Relaix, Biology of the Neuromuscular SystemU955‐IMRB, Inserm, UPEC, ENVA, EFSCréteilFrance
| |
Collapse
|
5
|
Kakimoto T, Ogasawara A, Ishikawa K, Kurita T, Yoshida K, Harada S, Nonaka T, Inoue Y, Uchida K, Tateoka T, Ohta T, Kumagai S, Sasaki T, Aihara H. A Systemically Administered Unconjugated Antisense Oligonucleotide Targeting DUX4 Improves Muscular Injury and Motor Function in FSHD Model Mice. Biomedicines 2023; 11:2339. [PMID: 37760780 PMCID: PMC10525656 DOI: 10.3390/biomedicines11092339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/02/2023] [Accepted: 08/13/2023] [Indexed: 09/29/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD), one of the most common muscular dystrophies, is caused by an abnormal expression of the DUX4 gene in skeletal muscles, resulting in muscle weakness. In this study, we investigated MT-DUX4-ASO, a novel gapmer antisense oligonucleotide (ASO). MT-DUX4-ASO decreased the expression of DUX4 and its target genes in FSHD patient-derived myoblasts. For the first time, we demonstrated that a systemically administered ASO, even without a ligand for drug delivery, could significantly improve muscle injury and motor function in the ACTA1-MCM/FLExDUX4 (DUX4-TG) mouse model of FSHD. Tamoxifen (TMX) injection transiently induces skeletal-muscle-specific DUX4 expression in DUX4-TG mice, while the skeletal muscles of TMX-untreated DUX4-TG mice have leaky DUX4 expression in a small subset of myofibers similar to those of FSHD patients. Subcutaneous 10 mg/kg of MT-DUX4-ASO at two-week intervals significantly suppressed muscular DUX4 target gene expression, histological muscle injury, and blood muscle injury marker elevation in TMX-untreated DUX4-TG mice. Notably, MT-DUX4-ASO at 10 mg/kg every other week significantly prevented the TMX-induced declines in treadmill test running speed and muscle force in DUX4-TG mice. Thus, the systemically administered unconjugated MT-DUX4-ASO suppressed disease progression in DUX4-TG mice, extending the potential of unconjugated ASOs as a promising FSHD treatment strategy.
Collapse
Affiliation(s)
- Tetsuhiro Kakimoto
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-26-1 Muraoka-Higashi, Fujisawa-shi, Kanagawa 251-8555, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Mirouse V. Evolution and developmental functions of the dystrophin-associated protein complex: beyond the idea of a muscle-specific cell adhesion complex. Front Cell Dev Biol 2023; 11:1182524. [PMID: 37384252 PMCID: PMC10293626 DOI: 10.3389/fcell.2023.1182524] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
The Dystrophin-Associated Protein Complex (DAPC) is a well-defined and evolutionarily conserved complex in animals. DAPC interacts with the F-actin cytoskeleton via dystrophin, and with the extracellular matrix via the membrane protein dystroglycan. Probably for historical reasons that have linked its discovery to muscular dystrophies, DAPC function is often described as limited to muscle integrity maintenance by providing mechanical robustness, which implies strong cell-extracellular matrix adhesion properties. In this review, phylogenetic and functional data from different vertebrate and invertebrate models will be analyzed and compared to explore the molecular and cellular functions of DAPC, with a specific focus on dystrophin. These data reveals that the evolution paths of DAPC and muscle cells are not intrinsically linked and that many features of dystrophin protein domains have not been identified yet. DAPC adhesive properties also are discussed by reviewing the available evidence of common key features of adhesion complexes, such as complex clustering, force transmission, mechanosensitivity and mechanotransduction. Finally, the review highlights DAPC developmental roles in tissue morphogenesis and basement membrane (BM) assembly that may indicate adhesion-independent functions.
Collapse
Affiliation(s)
- Vincent Mirouse
- Institute of Genetics, Reproduction and Development (iGReD), Université Clermont Auvergne-UMR CNRS 6293-INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| |
Collapse
|
7
|
Sekine S, Mayama S, Nishijima N, Kojima T, Endo-Takahashi Y, Ishii Y, Shiono H, Akiyama S, Sakurai A, Sashida S, Hamano N, Tada R, Suzuki R, Maruyama K, Negishi Y. Development of a Gene and Nucleic Acid Delivery System for Skeletal Muscle Administration via Limb Perfusion Using Nanobubbles and Ultrasound. Pharmaceutics 2023; 15:1665. [PMID: 37376113 PMCID: PMC10302710 DOI: 10.3390/pharmaceutics15061665] [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: 04/27/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Strategies for gene and nucleic acid delivery to skeletal muscles have been extensively explored to treat Duchenne muscular dystrophy (DMD) and other neuromuscular diseases. Of these, effective intravascular delivery of naked plasmid DNA (pDNA) and nucleic acids into muscles is an attractive approach, given the high capillary density in close contact with myofibers. We developed lipid-based nanobubbles (NBs) using polyethylene-glycol-modified liposomes and an echo-contrast gas and found that these NBs could improve tissue permeability by ultrasound (US)-induced cavitation. Herein, we delivered naked pDNA or antisense phosphorodiamidate morpholino oligomers (PMOs) into the regional hindlimb muscle via limb perfusion using NBs and US exposure. pDNA encoding the luciferase gene was injected with NBs via limb perfusion into normal mice with application of US. High luciferase activity was achieved in a wide area of the limb muscle. DMD model mice were administered PMOs, designed to skip the mutated exon 23 of the dystrophin gene, with NBs via intravenous limb perfusion, followed by US exposure. The number of dystrophin-positive fibers increased in the muscles of mdx mice. Combining NBs and US exposure, which can be widely delivered to the hind limb muscles via the limb vein, could be an effective therapeutic approach for DMD and other neuromuscular disorders.
Collapse
Affiliation(s)
- Shohko Sekine
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Sayaka Mayama
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Nobuaki Nishijima
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Takuo Kojima
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Yoko Endo-Takahashi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Yuko Ishii
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Hitomi Shiono
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Saki Akiyama
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Akane Sakurai
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Sanae Sashida
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Nobuhito Hamano
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Rui Tada
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan;
- Advanced Comprehensive Research Organization (ACRO), Teikyo University, Tokyo 173-8605, Japan;
| | - Kazuo Maruyama
- Advanced Comprehensive Research Organization (ACRO), Teikyo University, Tokyo 173-8605, Japan;
- Laboratory of Ultrasound Theranostics, Faculty of Pharma-Sciences, Teikyo University, Tokyo 173-8605, Japan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.E.-T.); (N.H.); (R.T.)
| |
Collapse
|
8
|
Russell AJ, DuVall M, Barthel B, Qian Y, Peter AK, Newell-Stamper BL, Hunt K, Lehman S, Madden M, Schlachter S, Robertson B, Van Deusen A, Rodriguez HM, Vera C, Su Y, Claflin DR, Brooks SV, Nghiem P, Rutledge A, Juehne TI, Yu J, Barton ER, Luo YE, Patsalos A, Nagy L, Sweeney HL, Leinwand LA, Koch K. Modulating fast skeletal muscle contraction protects skeletal muscle in animal models of Duchenne muscular dystrophy. J Clin Invest 2023; 133:e153837. [PMID: 36995778 PMCID: PMC10178848 DOI: 10.1172/jci153837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal muscle disease caused by absence of the protein dystrophin, which acts as a structural link between the basal lamina and contractile machinery to stabilize muscle membranes in response to mechanical stress. In DMD, mechanical stress leads to exaggerated membrane injury and fiber breakdown, with fast fibers being the most susceptible to damage. A major contributor to this injury is muscle contraction, controlled by the motor protein myosin. However, how muscle contraction and fast muscle fiber damage contribute to the pathophysiology of DMD has not been well characterized. We explored the role of fast skeletal muscle contraction in DMD with a potentially novel, selective, orally active inhibitor of fast skeletal muscle myosin, EDG-5506. Surprisingly, even modest decreases of contraction (<15%) were sufficient to protect skeletal muscles in dystrophic mdx mice from stress injury. Longer-term treatment also decreased muscle fibrosis in key disease-implicated tissues. Importantly, therapeutic levels of myosin inhibition with EDG-5506 did not detrimentally affect strength or coordination. Finally, in dystrophic dogs, EDG-5506 reversibly reduced circulating muscle injury biomarkers and increased habitual activity. This unexpected biology may represent an important alternative treatment strategy for Duchenne and related myopathies.
Collapse
Affiliation(s)
- Alan J. Russell
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Mike DuVall
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Ben Barthel
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Ying Qian
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Angela K. Peter
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | | | - Kevin Hunt
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Sarah Lehman
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Molly Madden
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Stephen Schlachter
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Ben Robertson
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Ashleigh Van Deusen
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | | | - Carlos Vera
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Yu Su
- Molecular and Integrative Physiology and
| | - Dennis R. Claflin
- Department of Surgery, Section of Plastic Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Peter Nghiem
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Alexis Rutledge
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Twlya I. Juehne
- Genome Technology Access Center, Department of Genetics, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Jinsheng Yu
- Genome Technology Access Center, Department of Genetics, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Elisabeth R. Barton
- Department of Applied Physiology and Kinesiology and Myology Institute, University of Florida College of Health and Human Performance, Gainesville, Florida, USA
| | - Yangyi E. Luo
- Department of Applied Physiology and Kinesiology and Myology Institute, University of Florida College of Health and Human Performance, Gainesville, Florida, USA
| | - Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics and Myology Institute, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Leslie A. Leinwand
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Kevin Koch
- Edgewise Therapeutics, BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
9
|
Fahrner A, Luca E, Krützfeldt J. microRNA-501 controls myogenin +/CD74 + myogenic progenitor cells during muscle regeneration. Mol Metab 2023; 71:101704. [PMID: 36907509 PMCID: PMC10033748 DOI: 10.1016/j.molmet.2023.101704] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/14/2023] Open
Abstract
OBJECTIVE Skeletal muscle regeneration is markedly impaired during aging. How adult muscle stem cells contribute to this decrease in regenerative capacity is incompletely understood. We investigated mechanisms of age-related changes in myogenic progenitor cells using the tissue-specific microRNA 501. METHODS Young and old C57Bl/6 mice were used (3 months or 24 months of age, respectively) with or without global or tissue-specific genetic deletion of miR-501. Muscle regeneration was induced using intramuscular cardiotoxin injection or treadmill exercise and analysed using single cell and bulk RNA sequencing, qRT-PCR and immunofluorescence. Muscle fiber damage was assessed with Evan`s blue dye (EBD). In vitro analysis was performed in primary muscle cells obtained from mice and humans. RESULTS Single cell sequencing revealed myogenic progenitor cells in miR-501 knockout mice at day 6 after muscle injury that are characterized by high levels of myogenin and CD74. In control mice these cells were less in number and already downregulated after day 3 of muscle injury. Muscle from knockout mice had reduced myofiber size and reduced myofiber resilience to injury and exercise. miR-501 elicits this effect by regulating sarcomeric gene expression through its target gene estrogen-related receptor gamma (Esrrg). Importantly, in aged skeletal muscle where miR-501 was significantly downregulated and its target Esrrg significantly upregulated, the number of myog+/CD74+ cells during regeneration was upregulated to similar levels as observed in 501 knockout mice. Moreover, myog+/CD74+-aged skeletal muscle exhibited a similar decrease in the size of newly formed myofibers and increased number of necrotic myofibers after injury as observed in mice lacking miR-501. CONCLUSIONS miR-501 and Esrrg are regulated in muscle with decreased regenerative capacity and loss of miR-501 is permissive to the appearance of CD74+ myogenic progenitors. Our data uncover a novel link between the metabolic transcription factor Esrrg and sarcomere formation and demonstrate that stem cell heterogeneity in skeletal muscle during aging is under miRNA control. Targeting Esrrg or myog+/CD74+ progenitor cells might improve fiber size and myofiber resilience to exercise in aged skeletal muscle.
Collapse
Affiliation(s)
- Alexandra Fahrner
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Zurich, Switzerland; Life Science Zurich Graduate School, Biomedicine, University of Zurich, 8057, Zurich, Switzerland
| | - Edlira Luca
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Jan Krützfeldt
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Zurich, Switzerland; Life Science Zurich Graduate School, Biomedicine, University of Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
10
|
Effect of heme oxygenase-1 on the differentiation of human myoblasts and the regeneration of murine skeletal muscles after acute and chronic injury. Pharmacol Rep 2023; 75:397-410. [PMID: 36918494 DOI: 10.1007/s43440-023-00475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Impaired muscle regeneration is a hallmark of Duchenne muscular dystrophy (DMD), a neuromuscular disorder caused by mutations in the DMD gene encoding dystrophin. The lack of heme oxygenase-1 (HO-1, Hmox1), a known anti-inflammatory and cytoprotective enzyme, was shown to aggravate DMD pathology. METHODS We evaluated the role of HO-1 overexpression in human induced pluripotent stem cell (hiPSC)-derived skeletal muscle cells (hiPSC-SkM) in vitro and in the regeneration process in vivo in wild-type mice. Furthermore, the effect of cobalt protoporphyrin IX (CoPP), a pharmacological inducer of HO-1 expression, on regeneration markers during myogenic hiPSC differentiation and progression of the dystrophic phenotype was analysed in the mdx mouse DMD model. RESULTS HO-1 has an impact on hiPSC-SkM generation by decreasing cell fusion capacity and the expression of myogenic regulatory factors and muscle-specific microRNAs (myomiRs). Also, strong induction of HO-1 by CoPP totally abolished hiPSC-SkM differentiation. Injection of HO-1-overexpressing hiPSC-SkM into the cardiotoxin (CTX)-injured muscle of immunodeficient wild-type mice was associated with decreased expression of miR-206 and Myh3 and lower number of regenerating fibers, suggesting some advanced regeneration. However, the very potent induction of HO-1 by CoPP did not exert any protective effect on necrosis, leukocyte infiltration, fibrosis, myofiber regeneration biomarkers, and exercise capacity of mdx mice. CONCLUSIONS In summary, HO-1 inhibits the expression of differentiation markers in human iPSC-derived myoblasts. Although moderate overexpression of HO-1 in the injected myoblast was associated with partially advanced muscle regeneration, the high systemic induction of HO-1 did not improve muscle regeneration. The appropriate threshold of HO-1 expression must be established for the therapeutic effect of HO-1 on muscle regeneration.
Collapse
|
11
|
Jama A, Alshudukhi AA, Burke S, Dong L, Kamau JK, Voss AA, Ren H. Lipin1 plays complementary roles in myofibre stability and regeneration in dystrophic muscles. J Physiol 2023; 601:961-978. [PMID: 36715084 PMCID: PMC9992338 DOI: 10.1113/jp284085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder caused by dystrophin mutations, leading to the loss of sarcolemmal integrity, and resulting in progressive myofibre necrosis and impaired muscle function. Our previous studies suggest that lipin1 is important for skeletal muscle regeneration and myofibre integrity. Additionally, we discovered that mRNA expression levels of lipin1 were significantly reduced in skeletal muscle of DMD patients and the mdx mouse model. To understand the role of lipin1 in dystrophic muscle, we generated dystrophin/lipin1 double knockout (DKO) mice, and compared the limb muscle pathology and function of wild-type B10, muscle-specific lipin1 deficient (lipin1Myf5cKO ), mdx and DKO mice. We found that further knockout of lipin1 in dystrophic muscle exhibited a more severe phenotype characterized by increased necroptosis, fibrosis and exacerbated membrane damage in DKO compared to mdx mice. In barium chloride-induced muscle injury, both lipin1Myf5cKO and DKO showed prolonged regeneration at day 14 post-injection, suggesting that lipin1 is critical for muscle regeneration. In situ contractile function assays showed that lipin1 deficiency in dystrophic muscle led to reduced specific force production. Using a cell culture system, we found that lipin1 deficiency led to elevated expression levels of necroptotic markers and medium creatine kinase, which could be a result of sarcolemmal damage. Most importantly, restoration of lipin1 inhibited the elevation of necroptotic markers in differentiated primary lipin1-deficient myoblasts. Overall, our data suggests that lipin1 plays complementary roles in myofibre stability and muscle function in dystrophic muscles, and overexpression of lipin1 may serve as a potential therapeutic strategy for dystrophic muscles. KEY POINTS: We identified that lipin1 mRNA expression levels are significantly reduced in skeletal muscles of Duchenne muscular dystrophy patients and mdx mice. We found that further depletion of lipin1 in skeletal muscles of mdx mice induces more severe dystrophic phenotypes, including enhanced myofibre sarcolemma damage, muscle necroptosis, inflammation, fibrosis and reduced specific force production. Lipin1 deficiency leads to elevated expression levels of necroptotic markers, whereas restoration of lipin1 inhibits their expression. Our results suggest that lipin1 is functionally complementary to dystrophin in muscle membrane integrity and muscle regeneration.
Collapse
Affiliation(s)
- Abdulrahman Jama
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Abdullah A. Alshudukhi
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Steve Burke
- Department of Biological Sciences, Wright State University, Dayton, OH, USA
| | - Lixin Dong
- Mumetel LLC, University Technology Park at IIT, Chicago, IL, USA
| | - John Karanja Kamau
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Andrew Alvin Voss
- Department of Biological Sciences, Wright State University, Dayton, OH, USA
| | - Hongmei Ren
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| |
Collapse
|
12
|
Kiriaev L, Baumann CW, Lindsay A. Eccentric contraction-induced strength loss in dystrophin-deficient muscle: Preparations, protocols, and mechanisms. J Gen Physiol 2023; 155:213810. [PMID: 36651896 PMCID: PMC9856740 DOI: 10.1085/jgp.202213208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/01/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
The absence of dystrophin hypersensitizes skeletal muscle of lower and higher vertebrates to eccentric contraction (ECC)-induced strength loss. Loss of strength can be accompanied by transient and reversible alterations to sarcolemmal excitability and disruption, triad dysfunction, and aberrations in calcium kinetics and reactive oxygen species production. The degree of ECC-induced strength loss, however, appears dependent on several extrinsic and intrinsic factors such as vertebrate model, skeletal muscle preparation (in vivo, in situ, or ex vivo), skeletal muscle hierarchy (single fiber versus whole muscle and permeabilized versus intact), strength production, fiber branching, age, and genetic background, among others. Consistent findings across research groups show that dystrophin-deficient fast(er)-twitch muscle is hypersensitive to ECCs relative to wildtype muscle, but because preparations are highly variable and sensitivity to ECCs are used repeatedly to determine efficacy of many preclinical treatments, it is critical to evaluate the impact of skeletal muscle preparations on sensitivity to ECC-induced strength loss in dystrophin-deficient skeletal muscle. Here, we review and discuss variations in skeletal muscle preparations to evaluate the factors responsible for variations and discrepancies between research groups. We further highlight that dystrophin-deficiency, or loss of the dystrophin-glycoprotein complex in skeletal muscle, is not a prerequisite for accelerated strength loss-induced by ECCs.
Collapse
Affiliation(s)
- Leonit Kiriaev
- Muscle Research Group, Murdoch Children’s Research Institute, Parkville, Victoria, Australia,School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Cory W. Baumann
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, OH, USA,Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia,Correspondence to Angus Lindsay:
| |
Collapse
|
13
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
14
|
Dowling P, Gargan S, Swandulla D, Ohlendieck K. Proteomic profiling of impaired excitation-contraction coupling and abnormal calcium handling in muscular dystrophy. Proteomics 2022; 22:e2200003. [PMID: 35902360 PMCID: PMC10078611 DOI: 10.1002/pmic.202200003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/06/2022]
Abstract
The X-linked inherited neuromuscular disorder Duchenne muscular dystrophy is characterised by primary abnormalities in the membrane cytoskeletal component dystrophin. The almost complete absence of the Dp427-M isoform of dystrophin in skeletal muscles renders contractile fibres more susceptible to progressive degeneration and a leaky sarcolemma membrane. This in turn results in abnormal calcium homeostasis, enhanced proteolysis and impaired excitation-contraction coupling. Biochemical and mass spectrometry-based proteomic studies of both patient biopsy specimens and genetic animal models of dystrophinopathy have demonstrated significant changes in the concentration and/or physiological function of essential calcium-regulatory proteins in dystrophin-lacking voluntary muscles. Abnormalities include dystrophinopathy-associated changes in voltage sensing receptors, calcium release channels, calcium pumps and calcium binding proteins. This review article provides an overview of the importance of the sarcolemmal dystrophin-glycoprotein complex and the wider dystrophin complexome in skeletal muscle and its linkage to depolarisation-induced calcium-release mechanisms and the excitation-contraction-relaxation cycle. Besides chronic inflammation, fat substitution and reactive myofibrosis, a major pathobiochemical hallmark of X-linked muscular dystrophy is represented by the chronic influx of calcium ions through the damaged plasmalemma in conjunction with abnormal intracellular calcium fluxes and buffering. Impaired calcium handling proteins should therefore be included in an improved biomarker signature of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
15
|
Iwasaki T, Terrill JR, Kawarai K, Miyata Y, Tagami T, Maeda N, Hasegawa Y, Watanabe T, Grounds MD, Arthur PG. The location of protein oxidation in dystrophic skeletal muscle from the mdx mouse model of Duchenne muscular dystrophy. Acta Histochem 2022; 124:151959. [DOI: 10.1016/j.acthis.2022.151959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/01/2022]
|
16
|
Riddell DO, Hildyard JCW, Harron RCM, Hornby NL, Wells DJ, Piercy RJ. Serum inflammatory cytokines as disease biomarkers in the DE50-MD dog model of Duchenne muscular dystrophy. Dis Model Mech 2022; 15:dmm049394. [PMID: 36444978 PMCID: PMC9789403 DOI: 10.1242/dmm.049394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease, caused by mutations in the dystrophin gene, characterised by cycles of muscle degeneration, inflammation and regeneration. Recently, there has been renewed interest specifically in drugs that ameliorate muscle inflammation in DMD patients. The DE50-MD dog is a model of DMD that closely mimics the human DMD phenotype. We quantified inflammatory proteins in serum from wild-type (WT) and DE50-MD dogs aged 3-18 months to identify biomarkers for future pre-clinical trials. Significantly higher concentrations of C-C motif chemokine ligand 2 (CCL2), granulocyte-macrophage colony-stimulating factor (GM-CSF or CSF2), keratinocyte chemotactic-like (KC-like, homologous to mouse CXCL1), TNFα (or TNF), and interleukins IL2, IL6, IL7, IL8 (CXCL8), IL10, IL15 and IL18 were detected in DE50-MD serum compared to WT serum. Of these, CCL2 best differentiated the two genotypes. The relative level of CCL2 mRNA was greater in the vastus lateralis muscle of DE50-MD dogs than in that of WT dogs, and CCL2 was expressed both within and at the periphery of damaged myofibres. Serum CCL2 concentration was significantly associated with acid phosphatase staining in vastus lateralis biopsy samples in DE50-MD dogs. In conclusion, the serum cytokine profile suggests that inflammation is a feature of the DE50-MD phenotype. Quantification of serum CCL2 in particular is a useful non-invasive biomarker of the DE50-MD phenotype.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Natasha L. Hornby
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| |
Collapse
|
17
|
Guhathakurta P, Carter AL, Thompson AR, Kurila D, LaFrence J, Zhang L, Trask JR, Vanderheyden B, Muretta JM, Ervasti JM, Thomas DD. Enhancing interaction of actin and actin-binding domain 1 of dystrophin with modulators: Toward improved gene therapy for Duchenne muscular dystrophy. J Biol Chem 2022; 298:102675. [PMID: 36372234 PMCID: PMC9731851 DOI: 10.1016/j.jbc.2022.102675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
Duchenne muscular dystrophy is a lethal muscle disease, caused by mutations in the gene encoding dystrophin, an actin-binding cytoskeletal protein. Absence of functional dystrophin results in muscle weakness and degeneration, eventually leading to cardiac and respiratory failure. Strategies to replace the missing dystrophin via gene therapy have been intensively pursued. However, the dystrophin gene is too large for current gene therapy approaches. Currently available micro-dystrophin constructs lack the actin-binding domain 2 and show decreased actin-binding affinity in vitro compared to full-length dystrophin. Thus, increasing the actin-binding affinity of micro-dystrophin, using small molecules, could be a beneficial therapeutic approach. Here, we have developed and validated a novel high-throughput screening (HTS) assay to discover small molecules that increase the binding affinity of dystrophin's actin-binding domain 1 (ABD1). We engineered a novel FRET biosensor, consisting of the mClover3, fluorescent protein (donor) attached to the C-terminus of dystrophin ABD1, and Alexa Fluor 568 (acceptor) attached to the C-terminal cysteine of actin. We used this biosensor in small-molecule screening, using a unique high-precision, HTS fluorescence lifetime assay, identifying several compounds from an FDA-approved library that significantly increase the binding between actin and ABD1. This HTS assay establishes feasibility for the discovery of small-molecule modulators of the actin-dystrophin interaction, with the ultimate goal of developing therapies for muscular dystrophy.
Collapse
|
18
|
Lloyd EM, Hepburn MS, Li J, Mowla A, Hwang Y, Choi YS, Grounds MD, Kennedy BF. Three-dimensional mechanical characterization of murine skeletal muscle using quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:5879-5899. [PMID: 36733728 PMCID: PMC9872891 DOI: 10.1364/boe.471062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 06/18/2023]
Abstract
Skeletal muscle function is governed by both the mechanical and structural properties of its constituent tissues, which are both modified by disease. Characterizing the mechanical properties of skeletal muscle tissue at an intermediate scale, i.e., between that of cells and organs, can provide insight into diseases such as muscular dystrophies. In this study, we use quantitative micro-elastography (QME) to characterize the micro-scale elasticity of ex vivo murine skeletal muscle in three-dimensions in whole muscles. To address the challenge of achieving high QME image quality with samples featuring uneven surfaces and geometry, we encapsulate the muscles in transparent hydrogels with flat surfaces. Using this method, we study aging and disease in quadriceps tissue by comparing normal wild-type (C57BL/6J) mice with dysferlin-deficient BLAJ mice, a model for the muscular dystrophy dysferlinopathy, at 3, 10, and 24 months of age (sample size of three per group). We observe a 77% decrease in elasticity at 24 months in dysferlin-deficient quadriceps compared to wild-type quadriceps.
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
- These authors contributed equally to this work
| | - Matt S. Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and 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
- These authors contributed equally to this work
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and 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 and 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
| | - 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
| | - 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
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and 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
| |
Collapse
|
19
|
Cardiac troponin T and autoimmunity in skeletal muscle aging. GeroScience 2022; 44:2025-2045. [PMID: 35034279 PMCID: PMC9616986 DOI: 10.1007/s11357-022-00513-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/05/2022] [Indexed: 01/03/2023] Open
Abstract
Age-related muscle mass and strength decline (sarcopenia) impairs the performance of daily living activities and can lead to mobility disability/limitation in older adults. Biological pathways in muscle that lead to mobility problems have not been fully elucidated. Immunoglobulin G (IgG) infiltration in muscle is a known marker of increased fiber membrane permeability and damage vulnerability, but whether this translates to impaired function is unknown. Here, we report that IgG1 and IgG4 are abundantly present in the skeletal muscle (vastus lateralis) of ~ 50% (11 out of 23) of older adults (> 65 years) examined. Skeletal muscle IgG1 was inversely correlated with physical performance (400 m walk time: r = 0.74, p = 0.005; SPPB score: r = - 0.73, p = 0.006) and muscle strength (r = - 0.6, p = 0.05). In a murine model, IgG was found to be higher in both muscle and blood of older, versus younger, C57BL/6 mice. Older mice with a higher level of muscle IgG had lower motor activity. IgG in mouse muscle co-localized with cardiac troponin T (cTnT) and markers of complement activation and apoptosis/necroptosis. Skeletal muscle-inducible cTnT knockin mice also showed elevated IgG in muscle and an accelerated muscle degeneration and motor activity decline with age. Most importantly, anti-cTnT autoantibodies were detected in the blood of cTnT knockin mice, old mice, and older humans. Our findings suggest a novel cTnT-mediated autoimmune response may be an indicator of sarcopenia.
Collapse
|
20
|
Size-tunable PEG-grafted copolymers as a polymeric nanoruler for passive targeting muscle tissues. J Control Release 2022; 347:607-614. [PMID: 35613686 DOI: 10.1016/j.jconrel.2022.05.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022]
Abstract
Muscle-targeted drug delivery is a major challenge in nanomedicine. The extravasation of nanomedicines (or nanoparticles) from the bloodstream into muscle tissues is hindered by the continuous endothelium, the so-called blood-muscle barrier. This study aimed to evaluate the optimal size of macromolecular drugs for extravasation (or passive targeting) into muscle tissues. We constructed a size-tunable polymeric delivery platform as a polymeric nanoruler by grafting poly(ethylene glycol)s (PEGs) onto the poly(aspartic acid) (PAsp) backbone. A series of PEG-grafted copolymers (gPEGs) with a narrow size distribution between 11 and 32 nm in hydrodynamic diameter (DH) were prepared by changing the molecular weight of the PEGs. Biodistribution analyses revealed that accumulation amounts of gPEGs in the muscle tissues of normal mice tended to decrease above their size of ~15 nm (or ~ 11 nm for the heart). The gPEGs accumulated in the skeletal muscles of Duchenne muscular dystrophy model mice (mdx mice) at a 2-3-fold higher level than in the skeletal muscles of normal mice. At the same time, there was a reduced accumulation of gPEGs in the spleen and liver. Intravital confocal laser scanning microscopy and immunohistochemical analysis showed extravasation and locally enhanced accumulation of gPEGs in the skeletal muscle of mdx mice. This study outlined the pivotal role of macromolecular drug size in muscle-targeted drug delivery and demonstrated the enhanced permeability of 11-32 nm-sized macromolecular drugs in mdx mice.
Collapse
|
21
|
Smith SJ, Fabian L, Sheikh A, Noche R, Cui X, Moore SA, Dowling JJ. Lysosomes and the pathogenesis of merosin-deficient congenital muscular dystrophy. Hum Mol Genet 2022; 31:733-747. [PMID: 34568901 PMCID: PMC9989739 DOI: 10.1093/hmg/ddab278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/01/2021] [Accepted: 09/14/2021] [Indexed: 11/14/2022] Open
Abstract
Congenital muscular dystrophy type 1A (MDC1A), the most common congenital muscular dystrophy in Western countries, is caused by recessive mutations in LAMA2, the gene encoding laminin alpha 2. Currently, no cure or disease modifying therapy has been successfully developed for MDC1A. Examination of patient muscle biopsies revealed altered distribution of lysosomes. We hypothesized that this redistribution was a novel and potentially druggable aspect of disease pathogenesis. We explored this hypothesis using candyfloss (caf), a zebrafish model of MDC1A. We found that lysosome distribution in caf zebrafish was also abnormal. This altered localization was significantly associated with fiber detachment and could be prevented by blocking myofiber detachment. Overexpression of transcription factor EB, a transcription factor that promotes lysosomal biogenesis, led to increased lysosome content and decreased fiber detachment. We conclude that genetic manipulation of the lysosomal compartment is able to alter the caf zebrafish disease process, suggesting that lysosome function may be a target for disease modification.
Collapse
Affiliation(s)
- Sarah J Smith
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Family Medicine, University of Calgary, Calgary T2R 0X7, Alberta
| | - Lacramioara Fabian
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Adeel Sheikh
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Ramil Noche
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiucheng Cui
- Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Steven A Moore
- Department of Pathology, University of Iowa Medical Center, Iowa City, IA, USA
| | - James J Dowling
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Neurology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
22
|
Mamsa H, Stark RL, Shin KM, Beedle AM, Crosbie RH. Sarcospan increases laminin-binding capacity of α-dystroglycan to ameliorate DMD independent of Galgt2. Hum Mol Genet 2022; 31:718-732. [PMID: 34581784 PMCID: PMC8895749 DOI: 10.1093/hmg/ddab276] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 08/27/2021] [Accepted: 09/13/2021] [Indexed: 11/14/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), mutations in dystrophin result in a loss of the dystrophin-glycoprotein complex (DGC) at the myofiber membrane, which functions to connect the extracellular matrix with the intracellular actin cytoskeleton. The dystroglycan subcomplex interacts with dystrophin and spans the sarcolemma where its extensive carbohydrates (matriglycan and CT2 glycan) directly interact with the extracellular matrix. In the current manuscript, we show that sarcospan overexpression enhances the laminin-binding capacity of dystroglycan in DMD muscle by increasing matriglycan glycosylation of α-dystroglycan. Furthermore, we find that this modification is not affected by loss of Galgt2, a glycotransferase, which catalyzes the CT2 glycan. Our findings reveal that the matriglycan carbohydrates, and not the CT2 glycan, are necessary for sarcospan-mediated amelioration of DMD. Overexpression of Galgt2 in the DMD mdx murine model prevents muscle pathology by increasing CT2 modified α-dystroglycan. Galgt2 also increases expression of utrophin, which compensates for the loss of dystrophin in DMD muscle. We found that combined loss of Galgt2 and dystrophin reduced utrophin expression; however, it did not interfere with sarcospan rescue of disease. These data reveal a partial dependence of sarcospan on Galgt2 for utrophin upregulation. In addition, sarcospan alters the cross-talk between the adhesion complexes by decreasing the association of integrin β1D with dystroglycan complexes. In conclusion, sarcospan functions to re-wire the cell to matrix connections by strengthening the cellular adhesion and signaling, which, in turn, increases the resilience of the myofiber membrane.
Collapse
Affiliation(s)
- Hafsa Mamsa
- Department of Integrative Biology & Physiology, University of California, Los Angeles 90095, USA
| | - Rachelle L Stark
- Department of Integrative Biology & Physiology, University of California, Los Angeles 90095, USA
| | - Kara M Shin
- Department of Integrative Biology & Physiology, University of California, Los Angeles 90095, USA
| | - Aaron M Beedle
- Department of Pharmaceutical Sciences, Binghamton University State University of New York, New York 13902, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology & Physiology, University of California, Los Angeles 90095, USA
- Broad Stem Cell Institute, University of California, Los Angeles 90095, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles 90095, USA
- Molecular Biology Institute, University of California, Los Angeles 90095, USA
| |
Collapse
|
23
|
Bourg N, Vu Hong A, Lostal W, Jaber A, Guerchet N, Tanniou G, Bordier F, Bertil-Froidevaux E, Georger C, Daniele N, Richard I, Israeli D. Co-Administration of Simvastatin Does Not Potentiate the Benefit of Gene Therapy in the mdx Mouse Model for Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23042016. [PMID: 35216132 PMCID: PMC8878028 DOI: 10.3390/ijms23042016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common and cureless muscle pediatric genetic disease, which is caused by the lack or the drastically reduced expression of dystrophin. Experimental therapeutic approaches for DMD have been mainly focused in recent years on attempts to restore the expression of dystrophin. While significant progress was achieved, the therapeutic benefit of treated patients is still unsatisfactory. Efficiency in gene therapy for DMD is hampered not only by incompletely resolved technical issues, but likely also due to the progressive nature of DMD. It is indeed suspected that some of the secondary pathologies, which are evolving over time in DMD patients, are not fully corrected by the restoration of dystrophin expression. We recently identified perturbations of the mevalonate pathway and of cholesterol metabolism in DMD patients. Taking advantage of the mdx model for DMD, we then demonstrated that some of these perturbations are improved by treatment with the cholesterol-lowering drug, simvastatin. In the present investigation, we tested whether the combination of the restoration of dystrophin expression with simvastatin treatment could have an additive beneficial effect in the mdx model. We confirmed the positive effects of microdystrophin, and of simvastatin, when administrated separately, but detected no additive effect by their combination. Thus, the present study does not support an additive beneficial effect by combining dystrophin restoration with a metabolic normalization by simvastatin.
Collapse
Affiliation(s)
- Nathalie Bourg
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Ai Vu Hong
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - William Lostal
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Abbass Jaber
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Nicolas Guerchet
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Guillaume Tanniou
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Fanny Bordier
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Emilie Bertil-Froidevaux
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Christophe Georger
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Nathalie Daniele
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Isabelle Richard
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - David Israeli
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
- Correspondence: ; Tel.: + 33-1-6947-2967
| |
Collapse
|
24
|
Valentine WJ, Mostafa SA, Tokuoka SM, Hamano F, Inagaki NF, Nordin JZ, Motohashi N, Kita Y, Aoki Y, Shimizu T, Shindou H. Lipidomic Analyses Reveal Specific Alterations of Phosphatidylcholine in Dystrophic Mdx Muscle. Front Physiol 2022; 12:698166. [PMID: 35095541 PMCID: PMC8791236 DOI: 10.3389/fphys.2021.698166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), lack of dystrophin increases the permeability of myofiber plasma membranes to ions and larger macromolecules, disrupting calcium signaling and leading to progressive muscle wasting. Although the biological origin and meaning are unclear, alterations of phosphatidylcholine (PC) are reported in affected skeletal muscles of patients with DMD that may include higher levels of fatty acid (FA) 18:1 chains and lower levels of FA 18:2 chains, possibly reflected in relatively high levels of PC 34:1 (with 16:0_18:1 chain sets) and low levels of PC 34:2 (with 16:0_18:2 chain sets). Similar PC alterations have been reported to occur in the mdx mouse model of DMD. However, altered ratios of PC 34:1 to PC 34:2 have been variably reported, and we also observed that PC 34:2 levels were nearly equally elevated as PC 34:1 in the affected mdx muscles. We hypothesized that experimental factors that often varied between studies; including muscle types sampled, mouse ages, and mouse diets; may strongly impact the PC alterations detected in dystrophic muscle of mdx mice, especially the PC 34:1 to PC 34:2 ratios. In order to test our hypothesis, we performed comprehensive lipidomic analyses of PC and phosphatidylethanolamine (PE) in several muscles (extensor digitorum longus, gastrocnemius, and soleus) and determined the mdx-specific alterations. The alterations in PC 34:1 and PC 34:2 were closely monitored from the neonate period to the adult, and also in mice raised on several diets that varied in their fats. PC 34:1 was naturally high in neonate’s muscle and decreased until age ∼3-weeks (disease onset age), and thereafter remained low in WT muscles but was higher in regenerated mdx muscles. Among the muscle types, soleus showed a distinctive phospholipid pattern with early and diminished mdx alterations. Diet was a major factor to impact PC 34:1/PC 34:2 ratios because mdx-specific alterations of PC 34:2 but not PC 34:1 were strictly dependent on diet. Our study identifies high PC 34:1 as a consistent biochemical feature of regenerated mdx-muscle and indicates nutritional approaches are also effective to modify the phospholipid compositions.
Collapse
Affiliation(s)
- William J. Valentine
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- *Correspondence: William J. Valentine,
| | - Sherif A. Mostafa
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Suzumi M. Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Fumie Hamano
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Natsuko F. Inagaki
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
| | - Joel Z. Nordin
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
- Department of Laboratory Medicine, Centre for Biomolecular and Cellular Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Norio Motohashi
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
| | - Yoshihiro Kita
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Center for Neurology and Psychiatry (NCNP), National Institute of Neuroscience, Kodaira, Tokyo, Japan
- Yoshitsugu Aoki,
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Hideo Shindou,
| |
Collapse
|
25
|
Mariot V, Joubert R, Le Gall L, Sidlauskaite E, Hourde C, Duddy W, Voit T, Bencze M, Dumonceaux J. RIPK3-mediated cell death is involved in DUX4-mediated toxicity in facioscapulohumeral dystrophy. J Cachexia Sarcopenia Muscle 2021; 12:2079-2090. [PMID: 34687171 PMCID: PMC8718031 DOI: 10.1002/jcsm.12813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 06/14/2021] [Accepted: 09/07/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Facioscapulohumeral dystrophy (FSHD) is caused by mutations leading to the aberrant expression of the DUX4 transcription factor in muscles. DUX4 was proposed to induce cell death, but the involvement of different death pathways is still discussed. A possible pro-apoptotic role of DUX4 was proposed, but as FSHD muscles are characterized by necrosis and inflammatory infiltrates, non-apoptotic pathways may be also involved. METHODS We explored DUX4-mediated cell death by focusing on the role of one regulated necrosis pathway called necroptosis, which is regulated by RIPK3. We investigated the effect of necroptosis on cell death in vitro and in vivo experiments using RIPK3 inhibitors and a RIPK3-deficient transgenic mouse model. RESULTS We showed in vitro that DUX4 expression causes a caspase-independent and RIPK3-mediated cell death in both myoblasts and myotubes. In vivo, RIPK3-deficient animals present improved body and muscle weights, a reduction of the aberrant activation of the DUX4 network genes, and an improvement of muscle histology. CONCLUSIONS These results provide evidence for a role of RIPK3 in DUX4-mediated cell death and open new avenues of research.
Collapse
Affiliation(s)
- Virginie Mariot
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Romain Joubert
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK.,United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Laura Le Gall
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Eva Sidlauskaite
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Christophe Hourde
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Savoie Mont Blanc, Chambéry, France
| | - William Duddy
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, Northern Ireland, UK
| | - Thomas Voit
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | - Maximilien Bencze
- University Paris Est Créteil, INSERM, IMRB, Créteil, France.,The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Julie Dumonceaux
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK.,Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, Northern Ireland, UK
| |
Collapse
|
26
|
Molecular and cellular basis of genetically inherited skeletal muscle disorders. Nat Rev Mol Cell Biol 2021; 22:713-732. [PMID: 34257452 PMCID: PMC9686310 DOI: 10.1038/s41580-021-00389-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
Neuromuscular disorders comprise a diverse group of human inborn diseases that arise from defects in the structure and/or function of the muscle tissue - encompassing the muscle cells (myofibres) themselves and their extracellular matrix - or muscle fibre innervation. Since the identification in 1987 of the first genetic lesion associated with a neuromuscular disorder - mutations in dystrophin as an underlying cause of Duchenne muscular dystrophy - the field has made tremendous progress in understanding the genetic basis of these diseases, with pathogenic variants in more than 500 genes now identified as underlying causes of neuromuscular disorders. The subset of neuromuscular disorders that affect skeletal muscle are referred to as myopathies or muscular dystrophies, and are due to variants in genes encoding muscle proteins. Many of these proteins provide structural stability to the myofibres or function in regulating sarcolemmal integrity, whereas others are involved in protein turnover, intracellular trafficking, calcium handling and electrical excitability - processes that ensure myofibre resistance to stress and their primary activity in muscle contraction. In this Review, we discuss how defects in muscle proteins give rise to muscle dysfunction, and ultimately to disease, with a focus on pathologies that are most common, best understood and that provide the most insight into muscle biology.
Collapse
|
27
|
Tian M, Chen K, Huang J, Chu D, Li J, Huang K, Ma C. Asiatic acid inhibits angiogenesis and vascular permeability through the VEGF/VEGFR2 signaling pathway to inhibit the growth and metastasis of breast cancer in mice. Phytother Res 2021; 35:6389-6400. [PMID: 34541711 DOI: 10.1002/ptr.7292] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/28/2021] [Accepted: 09/04/2021] [Indexed: 12/28/2022]
Abstract
Anti-angiogenic medicines have been evaluated as anticancer therapies, however, their use remains limited in clinical practice due to associated adverse effects. Asiatic acid (AA) is known to have broad-spectrum anticancer properties, however, its effects on angiogenesis in breast cancer remain to be fully established. In this study, we analyzed the inhibitory effects of AA on angiogenesis using human umbilical vein endothelial cells (HUVECs) cultured in vitro and on the growth and metastasis of a subcutaneous breast cancer 4T1 tumor model and a lung metastasis model in vivo. AA significantly inhibited HUVECs proliferation, migration, and tube formation in vitro. In vivo, AA significantly reduced the microvascular density and blood vascular permeability in breast cancer tumors and inhibited growth and lung metastasis. AA inhibited the expression of vascular endothelial growth factor (VEGF) in HUVECs and subsequently downregulated the phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2) and its downstream target proteins including ERK1/2, Src, and FAK. These results indicate that AA significantly inhibits angiogenesis and blood vessel permeability through the VEGF/VEGFR2 signal axis to inhibit the growth and metastasis of breast cancer. Our data strongly demonstrate the potential applications of AA in the treatment of breast cancer.
Collapse
Affiliation(s)
- Miaomiao Tian
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.,Graduated School of Jinzhou Medical University, Jinzhou, China
| | - Kan Chen
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jianhua Huang
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.,Life Science Institute of Jinzhou Medical University, Jinzhou, China
| | - Dongqing Chu
- Graduated School of Jinzhou Medical University, Jinzhou, China
| | - Jialin Li
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.,Graduated School of Jinzhou Medical University, Jinzhou, China
| | - Keqiang Huang
- Second Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chunyu Ma
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
28
|
Uezumi A, Ikemoto-Uezumi M, Zhou H, Kurosawa T, Yoshimoto Y, Nakatani M, Hitachi K, Yamaguchi H, Wakatsuki S, Araki T, Morita M, Yamada H, Toyoda M, Kanazawa N, Nakazawa T, Hino J, Fukada SI, Tsuchida K. Mesenchymal Bmp3b expression maintains skeletal muscle integrity and decreases in age-related sarcopenia. J Clin Invest 2021; 131:139617. [PMID: 33170806 DOI: 10.1172/jci139617] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Age-related sarcopenia constitutes an important health problem associated with adverse outcomes. Sarcopenia is closely associated with fat infiltration in muscle, which is attributable to interstitial mesenchymal progenitors. Mesenchymal progenitors are nonmyogenic in nature but are required for homeostatic muscle maintenance. However, the underlying mechanism of mesenchymal progenitor-dependent muscle maintenance is not clear, nor is the precise role of mesenchymal progenitors in sarcopenia. Here, we show that mice genetically engineered to specifically deplete mesenchymal progenitors exhibited phenotypes markedly similar to sarcopenia, including muscle weakness, myofiber atrophy, alterations of fiber types, and denervation at neuromuscular junctions. Through searching for genes responsible for mesenchymal progenitor-dependent muscle maintenance, we found that Bmp3b is specifically expressed in mesenchymal progenitors, whereas its expression level is significantly decreased during aging or adipogenic differentiation. The functional importance of BMP3B in maintaining myofiber mass as well as muscle-nerve interaction was demonstrated using knockout mice and cultured cells treated with BMP3B. Furthermore, the administration of recombinant BMP3B in aged mice reversed their sarcopenic phenotypes. These results reveal previously unrecognized mechanisms by which the mesenchymal progenitors ensure muscle integrity and suggest that age-related changes in mesenchymal progenitors have a considerable impact on the development of sarcopenia.
Collapse
Affiliation(s)
- Akiyoshi Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Madoka Ikemoto-Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Heying Zhou
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Tamaki Kurosawa
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Yuki Yoshimoto
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Masashi Nakatani
- Faculty of Rehabilitation and Care, Seijoh University, Tokai, Japan
| | - Keisuke Hitachi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Japan
| | - Hisateru Yamaguchi
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Japan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Mitsuhiro Morita
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Japan
| | - Harumoto Yamada
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Japan
| | | | - Nobuo Kanazawa
- Department of Surgery, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology (TMGHIG), Tokyo, Japan
| | | | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Japan
| |
Collapse
|
29
|
Ben Larbi S, Saclier M, Fessard A, Juban G, Chazaud B. Histological Analysis of Tibialis Anterior Muscle of DMDmdx4Cv Mice from 1 to 24 Months. J Neuromuscul Dis 2021; 8:513-524. [PMID: 33843691 DOI: 10.3233/jnd-200562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The mdx-C57/B6 mouse model does not show the clinical signs of Duchenne muscular dystrophy (DMD), although muscles exhibit hallmarks of permanent regeneration and alterations in muscle function. The DMDmdx4Cv strain exhibits very few revertant dystrophin positive myofibers, making that model suitable for studies on gene and cell therapies. OBJECTIVE The study appraises the histological evolution of the Tibialis Anterior muscle of WT and DMD mdx4Cv mutant from 1 to 24 months. METHODS Histological analysis included a series of immunostainings of muscle sections for assessing tissue features (fibrosis, lipid deposition, necrosis) and cellular characteristics (size of myofibers, number and distribution of myonuclei, number of satellite cells, vessels, macrophages). RESULTS None of the investigated cell types (satellite cells, endothelial cells, macrophages) showed variations in their density within the tissue in both WT and DMD mdx4Cv muscle. However, analyzing their number per myofiber showed that in DMD mdx4Cv, myofiber capillarization was increased from 1 to 6 months as compared with WT muscle, then dropped from 12 months. Macrophage number did not vary in WT muscle and peaked at 6 months in DMD mdx4Cv muscle. The number of satellite cells per myofiber did not vary in WT muscle while it remained high in DMD mdx4Cv muscle, starting to decrease from 12 months and being significantly lower at 24 months of age. Myofiber size was not different in DMD mdx4Cv from WT except at 24 months, when it strongly decreased in DMD mdx4Cv muscle. Necrosis and lipid deposition were rare in DMD mdx4Cv muscle. Fibrosis did not increase with age in DMD mdx4Cv muscle and was higher than in WT at 6 and 12 months of age. CONCLUSIONS As a whole, the results show a strong decrease of the myofiber size at 24 months, and an increased capillarization until 6 months of age in DMD mdx4Cv as compared with the WT. Thus, DMD mdx4Cv mice poorly recapitulates histological DMD features, and its use should take into account the age of the animals according to the purpose of the investigation.
Collapse
Affiliation(s)
- Sabrina Ben Larbi
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| | | | - Aurélie Fessard
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| |
Collapse
|
30
|
Jelinkova S, Sleiman Y, Fojtík P, Aimond F, Finan A, Hugon G, Scheuermann V, Beckerová D, Cazorla O, Vincenti M, Amedro P, Richard S, Jaros J, Dvorak P, Lacampagne A, Carnac G, Rotrekl V, Meli AC. Dystrophin Deficiency Causes Progressive Depletion of Cardiovascular Progenitor Cells in the Heart. Int J Mol Sci 2021; 22:ijms22095025. [PMID: 34068508 PMCID: PMC8125982 DOI: 10.3390/ijms22095025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating condition shortening the lifespan of young men. DMD patients suffer from age-related dilated cardiomyopathy (DCM) that leads to heart failure. Several molecular mechanisms leading to cardiomyocyte death in DMD have been described. However, the pathological progression of DMD-associated DCM remains unclear. In skeletal muscle, a dramatic decrease in stem cells, so-called satellite cells, has been shown in DMD patients. Whether similar dysfunction occurs with cardiac muscle cardiovascular progenitor cells (CVPCs) in DMD remains to be explored. We hypothesized that the number of CVPCs decreases in the dystrophin-deficient heart with age and disease state, contributing to DCM progression. We used the dystrophin-deficient mouse model (mdx) to investigate age-dependent CVPC properties. Using quantitative PCR, flow cytometry, speckle tracking echocardiography, and immunofluorescence, we revealed that young mdx mice exhibit elevated CVPCs. We observed a rapid age-related CVPC depletion, coinciding with the progressive onset of cardiac dysfunction. Moreover, mdx CVPCs displayed increased DNA damage, suggesting impaired cardiac muscle homeostasis. Overall, our results identify the early recruitment of CVPCs in dystrophic hearts and their fast depletion with ageing. This latter depletion may participate in the fibrosis development and the acceleration onset of the cardiomyopathy.
Collapse
MESH Headings
- Aging/genetics
- Aging/pathology
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiovascular System/metabolism
- Cardiovascular System/pathology
- DNA Damage/genetics
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Gene Expression Regulation/genetics
- Humans
- Mice
- Mice, Inbred mdx/genetics
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proto-Oncogene Proteins c-kit/genetics
- Stem Cells/metabolism
- Stem Cells/pathology
Collapse
Affiliation(s)
- Sarka Jelinkova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Yvonne Sleiman
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Petr Fojtík
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Franck Aimond
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Amanda Finan
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Gerald Hugon
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Valerie Scheuermann
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Deborah Beckerová
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
| | - Olivier Cazorla
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Marie Vincenti
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Pediatric and Adult Congenital Cardiology Department, M3C Regional Reference CHD Center, CHU Montpellier, 371 Avenue du Doyen Giraud, 34295 Montpellier, France
| | - Pascal Amedro
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Pediatric and Adult Congenital Cardiology Department, M3C Regional Reference CHD Center, CHU Montpellier, 371 Avenue du Doyen Giraud, 34295 Montpellier, France
| | - Sylvain Richard
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Josef Jaros
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5/A1, 62500 Brno, Czech Republic
| | - Petr Dvorak
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Gilles Carnac
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A3, 62500 Brno, Czech Republic; (S.J.); (P.F.); (D.B.); (P.D.)
- ICRC, St Anne’s University Hospital, Pekařská 53, 65691 Brno, Czech Republic;
- Correspondence: (V.R.); (A.C.M.); Tel.: +420-549-498-002 (V.R.); +33-4-67-41-52-44 (A.C.M.); Fax: +420-549-491-327 (V.R.); +33-4-67-41-52-42 (A.C.M.)
| | - Albano C. Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France; (Y.S.); (F.A.); (A.F.); (G.H.); (V.S.); (O.C.); (M.V.); (P.A.); (S.R.); (A.L.); (G.C.)
- Correspondence: (V.R.); (A.C.M.); Tel.: +420-549-498-002 (V.R.); +33-4-67-41-52-44 (A.C.M.); Fax: +420-549-491-327 (V.R.); +33-4-67-41-52-42 (A.C.M.)
| |
Collapse
|
31
|
Seo K, Kim EK, Choi J, Kim DS, Shin JH. Functional recovery of a novel knockin mouse model of dysferlinopathy by readthrough of nonsense mutation. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:702-709. [PMID: 34141825 PMCID: PMC8181533 DOI: 10.1016/j.omtm.2021.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/27/2021] [Indexed: 12/04/2022]
Abstract
Biallelic mutations in the dysferlin gene cause limb-girdle muscular dystrophy 2B or Miyoshi distal myopathy. We found that nonsense mutations are the most common mutation type among Korean patients with dysferlinopathy; more than half of the patients have at least one nonsense allele, which may be amenable to readthrough therapy. We generated a knockin mouse, dqx, harboring DYSF p.Q832∗ mutation. Homozygous dqx mice lacked dysferlin in skeletal muscle, while 2 weeks of oral ataluren restored dysferlin expression and ameliorated skeletal muscle pathology. Their physical performance improved, and protection against eccentric contractions was noted. The improvement was most evident in mice treated with oral ataluren of 0.9 mg/mL. These improvements were sustained for 8 weeks in ataluren-treated dqx mice, while the parameters of A/J mice treated with ataluren over the same period did not improve. These results support that readthrough therapy by oral ataluren may also be applicable to dysferlinopathy patients with nonsense mutation.
Collapse
Affiliation(s)
- Kyowon Seo
- Neurology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnamdo 50612, Republic of Korea
| | - Eun Kyoung Kim
- Neurology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnamdo 50612, Republic of Korea
| | - Jaeil Choi
- Neurology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnamdo 50612, Republic of Korea
| | - Dae-Seong Kim
- Neurology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnamdo 50612, Republic of Korea
| | - Jin-Hong Shin
- Neurology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnamdo 50612, Republic of Korea
| |
Collapse
|
32
|
Neyroud D, Nosacka RL, Callaway CS, Trevino JG, Hu H, Judge SM, Judge AR. FoxP1 is a transcriptional repressor associated with cancer cachexia that induces skeletal muscle wasting and weakness. J Cachexia Sarcopenia Muscle 2021; 12:421-442. [PMID: 33527776 PMCID: PMC8061399 DOI: 10.1002/jcsm.12666] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/05/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Skeletal muscle wasting is a devastating consequence of cancer that affects up to 80% of cancer patients and associates with reduced survival. Herein, we investigated the biological significance of Forkhead box P1 (FoxP1), a transcriptional repressor that we demonstrate is up-regulated in skeletal muscle in multiple models of cancer cachexia and in cachectic cancer patients. METHODS Inducible, skeletal muscle-specific FoxP1 over-expressing (FoxP1iSkmTg/Tg ) mice were generated through crossing conditional Foxp1a transgenic mice with HSA-MCM mice that express tamoxifen-inducible Cre recombinase under control of the skeletal muscle actin promoter. To determine the requirement of FoxP1 for cancer-induced skeletal muscle wasting, FoxP1-shRNA was packaged and targeted to muscles using AAV9 delivery prior to inoculation of mice with Colon-26 Adenocarcinoma (C26) cells. RESULTS Up-regulation of FoxP1 in adult skeletal muscle was sufficient to induce features of cachexia, including 15% reduction in body mass (P < 0.05), and a 16-27% reduction in skeletal muscle mass (P < 0.05) that was characterized by a 20% reduction in muscle fibre cross-sectional area of type IIX/B muscle fibres (P = 0.020). Skeletal muscles from FoxP1iSkmTg/Tg mice also showed significant damage and myopathy characterized by the presence of centrally nucleated myofibres, extracellular matrix expansion, and were 19-26% weaker than controls (P < 0.05). Transcriptomic analysis revealed FoxP1 as a potent transcriptional repressor of skeletal muscle gene expression, with enrichment of pathways related to skeletal muscle structure and function, growth signalling, and cell quality control. Because FoxP1 functions, at least in part, as a transcriptional repressor through its interaction with histone deacetylase proteins, we treated FoxP1iSkmTg/Tg mice with Trichostatin A, and found that this completely prevented the loss of muscle mass (p = 0.007) and fibre atrophy (P < 0.001) in the tibialis anterior. In the context of cancer, FoxP1 knockdown blocked the cancer-induced repression of myocyte enhancer factor 2 (MEF2)-target genes critical to muscle differentiation and repair, improved muscle ultrastructure, and attenuated muscle fibre atrophy by 50% (P < 0.05). CONCLUSIONS In summary, we identify FoxP1 as a novel repressor of skeletal muscle gene expression that is increased in cancer cachexia, whose up-regulation is sufficient to induce skeletal muscle wasting and weakness, and required for the normal wasting response to cancer.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | | | | | | | - Hui Hu
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Sarah M. Judge
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | - Andrew R. Judge
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
33
|
Muscle Diversity, Heterogeneity, and Gradients: Learning from Sarcoglycanopathies. Int J Mol Sci 2021; 22:ijms22052502. [PMID: 33801487 PMCID: PMC7958856 DOI: 10.3390/ijms22052502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle, the most abundant tissue in the body, is heterogeneous. This heterogeneity forms the basis of muscle diversity, which is reflected in the specialized functions of muscles in different parts of the body. However, these different parts are not always clearly delimitated, and this often gives rise to gradients within the same muscle and even across the body. During the last decade, several studies on muscular disorders both in mice and in humans have observed particular distribution patterns of muscle weakness during disease, indicating that the same mutation can affect muscles differently. Moreover, these phenotypical differences reveal gradients of severity, existing alongside other architectural gradients. These two factors are especially prominent in sarcoglycanopathies. Nevertheless, very little is known about the mechanism(s) driving the phenotypic diversity of the muscles affected by these diseases. Here, we will review the available literature on sarcoglycanopathies, focusing on phenotypic differences among affected muscles and gradients, characterization techniques, molecular signatures, and cell population heterogeneity, highlighting the possibilities opened up by new technologies. This review aims to revive research interest in the diverse disease phenotype affecting different muscles, in order to pave the way for new therapeutic interventions.
Collapse
|
34
|
Griffin DA, Pozsgai ER, Heller KN, Potter RA, Peterson EL, Rodino-Klapac LR. Preclinical Systemic Delivery of Adeno-Associated α-Sarcoglycan Gene Transfer for Limb-Girdle Muscular Dystrophy. Hum Gene Ther 2021; 32:390-404. [PMID: 33349138 PMCID: PMC8066346 DOI: 10.1089/hum.2019.199] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2D/R3 (LGMD2D/R3) is a progressive muscular dystrophy that manifests with muscle weakness, respiratory abnormalities, and in rare cases cardiomyopathy. LGMD2D/R3 is caused by mutations in the SGCA gene resulting in loss of protein and concomitant loss of some or all components of the dystrophin-associated glycoprotein complex. The sgca-null (sgca−/−) mouse recapitulates the clinical phenotype of patients with LGMD2D/R3, including dystrophic features such as muscle necrosis and fibrosis, elevated serum creatine kinase (CK), and reduction in the generation of absolute muscle force and locomotor activity. Thus, sgca−/− mice provide a relevant model to test the safety and efficacy of gene transfer. We designed a self-complementary AAVrh74 vector containing a codon-optimized full-length human SGCA (hSGCA) transgene driven by a muscle-specific promoter, shortened muscle creatine kinase (tMCK). In this report, we test the efficacy and safety of scAAVrh74.tMCK.hSGCA in sgca−/− mice using a dose-escalation design to evaluate a single systemic injection of 1.0 × 1012, 3.0 × 1012, and 6.0 × 1012 vg total dose compared with vehicle-treatment and wild-type mice. In sgca−/− mice, treatment with scAAVrh74.tMCK.hSGCA resulted in robust expression of α-sarcoglycan protein at the sarcolemma membrane in skeletal muscle at all doses tested. In addition, scAAVrh74.tMCK.hSGCA was effective in improving the histopathology of limb and diaphragm muscle of sgca−/− mice, as indicated by reductions in fibrosis, central nucleation, and normalization of myofiber size. These molecular changes were concomitant with significant increases in specific force generation in the diaphragm and tibialis anterior muscle, protection against eccentric force loss, and reduction in serum CK. Locomotor activity was improved at all doses of vector-treated compared with vehicle-treated sgca−/− mice. Lastly, vector toxicity was not detected in a serum chemistry panel and by gross necropsy. Collectively, these findings provide support for a systemic delivery of scAAVrh74.tMCK.hSGCA in a clinical setting for the treatment of LGMD2D/R3.
Collapse
Affiliation(s)
- Danielle A Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Eric R Pozsgai
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Rachael A Potter
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Ellyn L Peterson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | | |
Collapse
|
35
|
Barlow J, Sfyri PP, Mitchell R, Verpoorten S, Scully D, Andreou C, Papadopoulos P, Patel K, Matsakas A. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106:700-713. [PMID: 33450106 DOI: 10.1113/ep088937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the impact of obesity-independent hyperlipidaemia on skeletal muscle stem cell function of ApoE-deficient (ApoE-/- ) mice? What is the main finding and its importance? Compromised muscle stem cell function accounts for the impaired muscle regeneration in hyperlipidaemic ApoE-/- mice. Importantly, impaired muscle regeneration is normalised by administration of platelet releasate. ABSTRACT Muscle satellite cells are important stem cells for skeletal muscle regeneration and repair after injury. ApoE-deficient mice, an established mouse model of hyperlipidaemia and atherosclerosis, show evidence of oxidative stress-induced lesions and fat infiltration in skeletal muscle followed by impaired repair after injury. However, the mechanisms underpinning attenuated muscle regeneration remain to be fully defined. Key to addressing the latter is to understand the properties of muscle stem cells from ApoE-deficient mice and their myogenic potential. Muscle stem cells from ApoE-deficient mice were cultured both ex vivo (on single fibres) and in vitro (primary myoblasts) and their myogenic capacity was determined. Skeletal muscle regeneration was studied on days 5 and 10 after cardiotoxin injury. ApoE-deficient muscle stem cells showed delayed activation and differentiation on single muscle fibres ex vivo. Impaired proliferation and differentiation profiles were also evident on isolated primary muscle stem cells in culture. ApoE-deficient mice displayed impaired skeletal muscle regeneration after acute injury in vivo. Administration of platelet releasate in ApoE-deficient mice reversed the deficits of muscle regeneration after acute injury to wild-type levels. These findings indicate that muscle stem cell myogenic potential is perturbed in skeletal muscle of a mouse model of hyperlipidaemia. We propose that platelet releasate could be a therapeutic intervention for conditions with associated myopathy such as peripheral arterial disease.
Collapse
Affiliation(s)
- Joseph Barlow
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Rob Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - David Scully
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Charalampos Andreou
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Petros Papadopoulos
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| |
Collapse
|
36
|
Gibbs EM, McCourt JL, Shin KM, Hammond KG, Marshall JL, Crosbie RH. Loss of sarcospan exacerbates pathology in mdx mice, but does not affect utrophin amelioration of disease. Hum Mol Genet 2021; 30:149-159. [PMID: 33432327 PMCID: PMC8091037 DOI: 10.1093/hmg/ddaa264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/26/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023] Open
Abstract
The dystrophin-glycoprotein complex (DGC) is a membrane adhesion complex that provides structural stability at the sarcolemma by linking the myocyte's internal cytoskeleton and external extracellular matrix. In Duchenne muscular dystrophy (DMD), the absence of dystrophin leads to the loss of the DGC at the sarcolemma, resulting in sarcolemmal instability and progressive muscle damage. Utrophin (UTRN), an autosomal homolog of dystrophin, is upregulated in dystrophic muscle and partially compensates for the loss of dystrophin in muscle from patients with DMD. Here, we examine the interaction between Utr and sarcospan (SSPN), a small transmembrane protein that is a core component of both UTRN-glycoprotein complex (UGC) and DGC. We show that additional loss of SSPN causes an earlier onset of disease in dystrophin-deficient mdx mice by reducing the expression of the UGC at the sarcolemma. In order to further evaluate the role of SSPN in maintaining therapeutic levels of Utr at the sarcolemma, we tested the effect of Utr transgenic overexpression in mdx mice lacking SSPN (mdx:SSPN -/-:Utr-Tg). We found that overexpression of Utr restored SSPN to the sarcolemma in mdx muscle but that the ablation of SSPN in mdx muscle reduced Utr at the membrane. Nevertheless, Utr overexpression reduced central nucleation and improved grip strength in both lines. These findings demonstrate that high levels of Utr transgenic overexpression ameliorate the mdx phenotype independently of SSPN expression but that loss of SSPN may impair Utr-based mechanisms that rely on lower levels of Utr protein.
Collapse
Affiliation(s)
- Elizabeth M Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Jackie L McCourt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Kara M Shin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Katherine G Hammond
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Jamie L Marshall
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
37
|
Bawa S, Gameros S, Baumann K, Brooks DS, Kollhoff JA, Zolkiewski M, Re Cecconi AD, Panini N, Russo M, Piccirillo R, Johnson DK, Kashipathy MM, Battaile KP, Lovell S, Bouyain SEA, Kawakami J, Geisbrecht ER. Costameric integrin and sarcoglycan protein levels are altered in a Drosophila model for Limb-girdle muscular dystrophy type 2H. Mol Biol Cell 2020; 32:260-273. [PMID: 33296226 PMCID: PMC8098830 DOI: 10.1091/mbc.e20-07-0453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mutations in two different domains of the ubiquitously expressed TRIM32 protein give rise to two clinically separate diseases, one of which is Limb-girdle muscular dystrophy type 2H (LGMD2H). Uncovering the muscle-specific role of TRIM32 in LGMD2H pathogenesis has proven difficult, as neurogenic phenotypes, independent of LGMD2H pathology, are present in TRIM32 KO mice. We previously established a platform to study LGMD2H pathogenesis using Drosophila melanogaster as a model. Here we show that LGMD2H disease-causing mutations in the NHL domain are molecularly and structurally conserved between fly and human TRIM32. Furthermore, transgenic expression of a subset of myopathic alleles (R394H, D487N, and 520fs) induce myofibril abnormalities, altered nuclear morphology, and reduced TRIM32 protein levels, mimicking phenotypes in patients afflicted with LGMD2H. Intriguingly, we also report for the first time that the protein levels of βPS integrin and sarcoglycan δ, both core components of costameres, are elevated in TRIM32 disease-causing alleles. Similarly, murine myoblasts overexpressing a catalytically inactive TRIM32 mutant aberrantly accumulate α- and β-dystroglycan and α-sarcoglycan. We speculate that the stoichiometric loss of costamere components disrupts costamere complexes to promote muscle degeneration.
Collapse
Affiliation(s)
- Simranjot Bawa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506
| | - Samantha Gameros
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506
| | - Kenny Baumann
- School of Biological Sciences, University of Missouri-Kansas City, MO 64110
| | - David S Brooks
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506
| | - Joseph A Kollhoff
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506
| | - Michal Zolkiewski
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506
| | | | - Nicolò Panini
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Massimo Russo
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | | | - David K Johnson
- Molecular Graphics and Modeling Laboratory, Computational Chemical Biology Core, University of Kansas, Lawrence, KS 66047
| | | | | | - Scott Lovell
- Protein Structure Laboratory, University of Kansas, Lawrence, KS 66047
| | - Samuel E A Bouyain
- School of Biological Sciences, University of Missouri-Kansas City, MO 64110
| | - Jessica Kawakami
- School of Biological Sciences, University of Missouri-Kansas City, MO 64110
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506.,School of Biological Sciences, University of Missouri-Kansas City, MO 64110
| |
Collapse
|
38
|
Bronisz-Budzyńska I, Kozakowska M, Podkalicka P, Kachamakova-Trojanowska N, Łoboda A, Dulak J. The role of Nrf2 in acute and chronic muscle injury. Skelet Muscle 2020; 10:35. [PMID: 33287890 PMCID: PMC7722332 DOI: 10.1186/s13395-020-00255-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) is considered as a master cytoprotective factor regulating the expression of genes encoding anti-oxidant, anti-inflammatory, and detoxifying proteins. The role of Nrf2 in the pathophysiology of skeletal muscles has been evaluated in different experimental models, however, due to inconsistent data, we aimed to investigate how Nrf2 transcriptional deficiency (Nrf2tKO) affects muscle functions both in an acute and chronic injury. The acute muscle damage was induced in mice of two genotypes-WT and Nrf2tKO mice by cardiotoxin (CTX) injection. To investigate the role of Nrf2 in chronic muscle pathology, mdx mice that share genetic, biochemical, and histopathological features with Duchenne muscular dystrophy (DMD) were crossed with mice lacking transcriptionally active Nrf2 and double knockouts (mdx/Nrf2tKO) were generated. To worsen the dystrophic phenotype, the analysis of disease pathology was also performed in aggravated conditions, by applying a long-term treadmill test. We have observed slightly increased muscle damage in Nrf2tKO mice after CTX injection. Nevertheless, transcriptional ablation of Nrf2 in mdx mice did not significantly aggravate the most deleterious, pathological hallmarks of DMD related to degeneration, inflammation, fibrotic scar formation, angiogenesis, and the number and proliferation of satellite cells in non-exercised conditions. On the other hand, upon chronic exercises, the degeneration and inflammatory infiltration of the gastrocnemius muscle, but not the diaphragm, turned to be increased in Nrf2tKOmdx in comparison to mdx mice. In conclusion, the lack of transcriptionally active Nrf2 influences moderately muscle pathology in acute CTX-induced muscle injury and chronic DMD mouse model, without affecting muscle functionality. Hence, in general, we demonstrated that the deficiency of Nrf2 transcriptional activity has no profound impact on muscle pathology in various models of muscle injury.
Collapse
Affiliation(s)
- Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | | | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
39
|
Smith LR, Kok HJ, Zhang B, Chung D, Spradlin RA, Rakoczy KD, Lei H, Boesze-Battaglia K, Barton ER. Matrix Metalloproteinase 13 from Satellite Cells is Required for Efficient Muscle Growth and Regeneration. Cell Physiol Biochem 2020; 54:333-353. [PMID: 32275813 DOI: 10.33594/000000223] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIMS Cell migration and extracellular matrix remodeling underlie normal mammalian development and growth as well as pathologic tumor invasion. Skeletal muscle is no exception, where satellite cell migration replenishes nuclear content in damaged tissue and extracellular matrix reforms during regeneration. A key set of enzymes that regulate these processes are matrix metalloproteinases (MMP)s. The collagenase MMP-13 is transiently upregulated during muscle regeneration, but its contribution to damage resolution is unknown. The purpose of this work was to examine the importance of MMP-13 in muscle regeneration and growth in vivo and to delineate a satellite cell specific role for this collagenase. METHODS Mice with total and satellite cell specific Mmp13 deletion were utilized to determine the importance of MMP-13 for postnatal growth, regeneration after acute injury, and in chronic injury from a genetic cross with dystrophic (mdx) mice. We also evaluated insulin-like growth factor 1 (IGF-1) mediated hypertrophy in the presence and absence of MMP-13. We employed live-cell imaging and 3D migration measurements on primary myoblasts obtained from these animals. Outcome measures included muscle morphology and function. RESULTS Under basal conditions, Mmp13-/- mice did not exhibit histological or functional deficits in muscle. However, following acute injury, regeneration was impaired at 11 and 14 days post injury. Muscle hypertrophy caused by increased IGF-1 was blunted with minimal satellite cell incorporation in the absence of MMP-13. Mmp13-/- primary myoblasts displayed reduced migratory capacity in 2D and 3D, while maintaining normal proliferation and differentiation. Satellite cell specific deletion of MMP-13 recapitulated the effects of global MMP-13 ablation on muscle regeneration, growth and myoblast movement. CONCLUSION These results show that satellite cells provide an essential autocrine source of MMP-13, which not only regulates their migration, but also supports postnatal growth and resolution of acute damage.
Collapse
Affiliation(s)
- Lucas R Smith
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Neurobiology, Physiology & Behavior, Physical Medicine & Rehabilitation, University of California, Davis, CA, USA
| | - Hui Jean Kok
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Boshi Zhang
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Du Chung
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ray A Spradlin
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Kyla D Rakoczy
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Hanqin Lei
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | | | - Elisabeth R Barton
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA, .,Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| |
Collapse
|
40
|
Tomazoni SS, Casalechi HL, Ferreira CDSB, Serra AJ, Dellê H, Brito RBDO, de Melo BL, Vanin AA, Ribeiro NF, Pereira AL, Monteiro KKDS, Marcos RL, de Carvalho PDTC, Frigo L, Leal-Junior ECP. Can photobiomodulation therapy be an alternative to pharmacological therapies in decreasing the progression of skeletal muscle impairments of mdx mice? PLoS One 2020; 15:e0236689. [PMID: 32785240 PMCID: PMC7423120 DOI: 10.1371/journal.pone.0236689] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/29/2020] [Indexed: 01/16/2023] Open
Abstract
Objective To compare the effects of photobiomodulation therapy (PBMT) and pharmacological therapy (glucocorticoids and non-steroidal anti-inflammatory drugs) applied alone and in different combinations in mdx mice. Methods The animals were randomized and divided into seven experimental groups treated with placebo, PBMT, prednisone, non-steroidal anti-inflammatory drug (NSAIDs), PBMT plus prednisone and PBMT plus NSAID. Wild type animals were used as control. All treatments were performed during 14 consecutive weeks. Muscular morphology, protein expression of dystrophin and functional performance were assessed at the end of the last treatment. Results Both treatments with prednisone and PBMT applied alone or combined, were effective in preserving muscular morphology. In addition, the treatments with PBMT (p = 0.0005), PBMT plus prednisone (p = 0.0048) and PBMT plus NSAID (p = 0.0021) increased dystrophin gene expression compared to placebo-control group. However, in the functional performance the PBMT presented better results compared to glucocorticoids (p<0.0001). In contrast, the use of NSAIDs did not appear to add benefits to skeletal muscle tissue in mdx mice. Conclusion We believe that the promising and optimistic results about the PBMT in skeletal muscle of mdx mice may in the future contribute to this therapy to be considered a safe alternative for patients with Duchenne Muscular Dystrophy (DMD) in a washout period (between treatment periods with glucocorticoids), allowing them to remain receiving effective and safe treatment in this period, avoiding at this way periods without administration of any treatment.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Combined Modality Therapy
- Disease Models, Animal
- Disease Progression
- Dystrophin/genetics
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/radiation effects
- Glucocorticoids/pharmacology
- Humans
- Low-Level Light Therapy
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/radiation effects
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/therapy
- Prednisone/pharmacology
Collapse
Affiliation(s)
- Shaiane Silva Tomazoni
- Department of Global Public Health and Primary Care, Physiotherapy Research Group, University of Bergen, Bergen, Norway
| | - Heliodora Leão Casalechi
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Cheila de Sousa Bacelar Ferreira
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Andrey Jorge Serra
- Postgraduate Program in Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Humberto Dellê
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | | | - Brunno Lemes de Melo
- Postgraduate Program in Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Adriane Aver Vanin
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Neide Firmo Ribeiro
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Amanda Lima Pereira
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Kadma Karênina Damasceno Soares Monteiro
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Rodrigo Labat Marcos
- Postgraduate Program in Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | | | - Lucio Frigo
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Department of Periodontology, Dental Research Division, Universidade de Guarulhos (UnG), Guarulhos, São Paulo, Brazil
| | - Ernesto Cesar Pinto Leal-Junior
- Department of Global Public Health and Primary Care, Physiotherapy Research Group, University of Bergen, Bergen, Norway
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
41
|
Fabian L, Dowling JJ. Zebrafish Models of LAMA2-Related Congenital Muscular Dystrophy (MDC1A). Front Mol Neurosci 2020; 13:122. [PMID: 32742259 PMCID: PMC7364686 DOI: 10.3389/fnmol.2020.00122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 01/28/2023] Open
Abstract
LAMA2-related congenital muscular dystrophy (CMD; LAMA2-MD), also referred to as merosin deficient CMD (MDC1A), is a severe neonatal onset muscle disease caused by recessive mutations in the LAMA2 gene. LAMA2 encodes laminin α2, a subunit of the extracellular matrix (ECM) oligomer laminin 211. There are currently no treatments for MDC1A, and there is an incomplete understanding of disease pathogenesis. Zebrafish, due to their high degree of genetic conservation with humans, large clutch sizes, rapid development, and optical clarity, have emerged as an excellent model system for studying rare Mendelian diseases. They are particularly suitable as a model for muscular dystrophy because they contain at least one orthologue to all major human MD genes, have muscle that is similar to human muscle in structure and function, and manifest obvious and easily measured MD related phenotypes. In this review article, we present the existing zebrafish models of MDC1A, and discuss their contribution to the understanding of MDC1A pathomechanisms and therapy development.
Collapse
Affiliation(s)
- Lacramioara Fabian
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurology, Hospital for Sick Children, Toronto, ON, Canada.,Departments of Pediatrics and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
42
|
Al-Khalili Szigyarto C. Duchenne Muscular Dystrophy: recent advances in protein biomarkers and the clinical application. Expert Rev Proteomics 2020; 17:365-375. [PMID: 32713262 DOI: 10.1080/14789450.2020.1773806] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Early biomarker discovery studies have praised the value of their emerging results, predicting an unprecedented impact on health care. Biomarkers are expected to provide tests with increased specificity and sensitivity compared to existing measures, improve the decision-making process, and accelerate the development of therapies. For rare disorders, like Duchenne Muscular Dystrophy (DMD) such biomarkers can assist the development of therapies, therefore also helping to find a cure for the disease. AREA COVERED State-of-the-art technologies have been used to identify blood biomarkers for DMD and efforts have been coordinated to develop and promote translation of biomarkers for clinical practice. Biomarker translation to clinical practice is however, adjoined by challenges related to the complexity of the disease, involving numerous biological processes, and the limited sample resources. This review highlights the current progress on the development of biomarkers, describing the proteomics technologies used, the most promising findings and the challenges encountered. EXPERT OPINION Strategies for effective use of samples combined with orthogonal proteomics methods for protein quantification are essential for translating biomarkers to the patient's bed side. Progress is achieved only if strong evidence is provided that the biomarker constitutes a reliable indicator of the patient's health status for a specific context of use.
Collapse
Affiliation(s)
- Cristina Al-Khalili Szigyarto
- Science for Life Laboratory, KTH - Royal Institute of Technology , Solna, Sweden.,School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology , Stockholm, Sweden
| |
Collapse
|
43
|
Podkalicka P, Mucha O, Bronisz-Budzyńska I, Kozakowska M, Pietraszek-Gremplewicz K, Cetnarowska A, Głowniak-Kwitek U, Bukowska-Strakova K, Cieśla M, Kulecka M, Ostrowski J, Mikuła M, Potulska-Chromik A, Kostera-Pruszczyk A, Józkowicz A, Łoboda A, Dulak J. Lack of miR-378 attenuates muscular dystrophy in mdx mice. JCI Insight 2020; 5:135576. [PMID: 32493839 PMCID: PMC7308053 DOI: 10.1172/jci.insight.135576] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/29/2020] [Indexed: 01/09/2023] Open
Abstract
The severity of Duchenne muscular dystrophy (DMD), an incurable disease caused by the lack of dystrophin, might be modulated by different factors, including miRNAs. Among them, miR-378 is considered of high importance for muscle biology, but intriguingly, its role in DMD and its murine model (mdx mice) has not been thoroughly addressed so far. Here, we demonstrate that dystrophic mice additionally globally lacking miR-378 (double-KO [dKO] animals) exhibited better physical performance and improved absolute muscle force compared with mdx mice. Accordingly, markers of muscle damage in serum were significantly decreased in dKO mice, accompanied by diminished inflammation, fibrosis, and reduced abundance of regenerating fibers within muscles. The lack of miR-378 also normalized the aggravated fusion of dystrophin-deficient muscle satellite cells (mSCs). RNA sequencing of gastrocnemius muscle transcriptome revealed fibroblast growth factor 1 (Fgf1) as one of the most significantly downregulated genes in mice devoid of miR-378, indicating FGF1 as one of the mediators of changes driven by the lack of miR-378. In conclusion, we suggest that targeting miR-378 has the potential to ameliorate DMD pathology.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | | | - Anna Cetnarowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Urszula Głowniak-Kwitek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and.,Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Medical College, Jagiellonian University, Krakow, Poland
| | - Maciej Cieśla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland.,Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Michał Mikuła
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | | | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, and
| |
Collapse
|
44
|
Forcina L, Cosentino M, Musarò A. Mechanisms Regulating Muscle Regeneration: Insights into the Interrelated and Time-Dependent Phases of Tissue Healing. Cells 2020; 9:E1297. [PMID: 32456017 PMCID: PMC7290814 DOI: 10.3390/cells9051297] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a massive body of knowledge which has been produced related to the mechanisms guiding muscle regeneration, great interest still moves the scientific community toward the study of different aspects of skeletal muscle homeostasis, plasticity, and regeneration. Indeed, the lack of effective therapies for several physiopathologic conditions suggests that a comprehensive knowledge of the different aspects of cellular behavior and molecular pathways, regulating each regenerative stage, has to be still devised. Hence, it is important to perform even more focused studies, taking the advantage of robust markers, reliable techniques, and reproducible protocols. Here, we provide an overview about the general aspects of muscle regeneration and discuss the different approaches to study the interrelated and time-dependent phases of muscle healing.
Collapse
Affiliation(s)
| | | | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy; (L.F.); (M.C.)
| |
Collapse
|
45
|
Porcari P, Hall MG, Clark CA, Greally E, Straub V, Blamire AM. Time-dependent diffusion MRI as a probe of microstructural changes in a mouse model of Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2020; 33:e4276. [PMID: 32101354 DOI: 10.1002/nbm.4276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/05/2020] [Accepted: 01/30/2020] [Indexed: 06/10/2023]
Abstract
Dystrophic muscles show a high variability of fibre sizes and altered sarcolemmal integrity, which are typically assessed by histology. Time-dependent diffusion MRI is sensitive to tissue microstructure and its investigation through age-related changes in dystrophic and healthy muscles may help the understanding of the onset and progression of Duchenne muscular dystrophy (DMD). We investigated the capability of time-dependent diffusion MRI to quantify age and disease-related changes in hind-limb muscle microstructure between dystrophic (mdx) and wild-type (WT) mice of three age groups (7.5, 22 and 44 weeks). Diffusion time-dependent apparent diffusion coefficients (ADCs) of the gastrocnemius and tibialis anterior muscles were determined versus age and diffusion-gradient orientation at six diffusion times (Δ; range: 25-350 ms). Mean muscle ADCs were compared between groups and ages, and correlated with T2 , using Student's t test, one-way analysis of variance and Pearson correlation, respectively. Muscle fibre sizes and sarcolemmal integrity were evaluated by histology and compared with diffusion measurements. Hind-limb muscle ADC showed characteristic restricted diffusion behaviour in both mdx and WT animals with decreasing ADC values at longer Δ. Significant differences in ADC were observed at long Δ values (≥ 250 ms; p < 0.05, comparison between groups; p < 0.01, comparison between ages) with ADC increased by 5-15% in dystrophic muscles, indicative of reduced diffusion restriction. No significant correlation was found between T2 and ADC. Additionally, muscle fibre size distributions showed higher variability and lower mean fibre size in mdx than WT animals (p < 0.001). The extensive Evans Blue Dye uptake shown in dystrophic muscles revealed substantial sarcolemmal damage, suggesting diffusion measurements as more consistent with altered permeability rather than changes in muscle fibre sizes. This study shows the potential of diffusion MRI to non-invasively discriminate between dystrophic and healthy muscles with enhanced sensitivity when using long Δ.
Collapse
Affiliation(s)
- Paola Porcari
- Institute of Genetic Medicine and Centre for In Vivo Imaging, Newcastle University, Newcastle upon Tyne, UK
| | - Matt G Hall
- Developmental Imaging and Biophysics Section, UCL GOS Institute of Child Health, London, UK
| | - Chris A Clark
- Developmental Imaging and Biophysics Section, UCL GOS Institute of Child Health, London, UK
| | - Elizabeth Greally
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew M Blamire
- Institute of Cellular Medicine and Centre for In Vivo Imaging, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
46
|
Grounds MD, Terrill JR, Al-Mshhdani BA, Duong MN, Radley-Crabb HG, Arthur PG. Biomarkers for Duchenne muscular dystrophy: myonecrosis, inflammation and oxidative stress. Dis Model Mech 2020; 13:13/2/dmm043638. [PMID: 32224496 PMCID: PMC7063669 DOI: 10.1242/dmm.043638] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked disease that causes severe loss of muscle mass and function in young children. Promising therapies for DMD are being developed, but the long lead times required when using clinical outcome measures are hindering progress. This progress would be facilitated by robust molecular biomarkers in biofluids, such as blood and urine, which could be used to monitor disease progression and severity, as well as to determine optimal drug dosing before a full clinical trial. Many candidate DMD biomarkers have been identified, but there have been few follow-up studies to validate them. This Review describes the promising biomarkers for dystrophic muscle that have been identified in muscle, mainly using animal models. We strongly focus on myonecrosis and the associated inflammation and oxidative stress in DMD muscle, as the lack of dystrophin causes repeated bouts of myonecrosis, which are the key events that initiate the resultant severe dystropathology. We discuss the early events of intrinsic myonecrosis, along with early regeneration in the context of histological and other measures that are used to quantify its incidence. Molecular biomarkers linked to the closely associated events of inflammation and oxidative damage are discussed, with a focus on research related to protein thiol oxidation and to neutrophils. We summarise data linked to myonecrosis in muscle, blood and urine of dystrophic animal species, and discuss the challenge of translating such biomarkers to the clinic for DMD patients, especially to enhance the success of clinical trials. Summary: This Review discusses biomarkers in blood and urine linked to myonecrosis, inflammation and oxidative stress, to enhance development of therapies for DMD, and the challenges to be overcome for clinical translation.
Collapse
Affiliation(s)
- Miranda D Grounds
- School of Human Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Jessica R Terrill
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Basma A Al-Mshhdani
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Marisa N Duong
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Hannah G Radley-Crabb
- School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Peter G Arthur
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
47
|
Kanamori H, Naruse G, Yoshida A, Minatoguchi S, Watanabe T, Kawaguchi T, Yamada Y, Mikami A, Kawasaki M, Takemura G, Minatoguchi S. Metformin Enhances Autophagy and Provides Cardioprotection in δ-Sarcoglycan Deficiency-Induced Dilated Cardiomyopathy. Circ Heart Fail 2020; 12:e005418. [PMID: 30922066 DOI: 10.1161/circheartfailure.118.005418] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Metformin is a popular antidiabetic agent that is also used to treat heart failure patients with type 2 diabetes mellitus. Several reports suggest that metformin may also have cardioprotective effects in patients without diabetes mellitus. In the present study, we investigated the possible therapeutic effect of metformin in heart failure and its underlying molecular mechanisms using a δ-sarcoglycan-deficient mouse model of dilated cardiomyopathy. METHODS AND RESULTS Thirty-two-week-old δ-sarcoglycan-deficient mice exhibiting established cardiomyopathy with extensive left ventricular dilatation and dysfunction were administered saline or metformin (200 mg/kg per day) for 4 weeks using osmotic mini-pumps. Metformin partially reversed the left ventricular dilatation (reverse remodeling) and significantly improved cardiac function. The hearts of metformin-treated mice showed less fibrosis, less cardiomyocyte hypertrophy, and fewer degenerative subcellular changes than saline-treated mice. These effects were accompanied by restored expression of the sarcomeric proteins myosin heavy chain and troponin I, and their transcription factor, GATA-4. Autophagy was enhanced in the hearts from metformin-treated mice, as indicated by increase of myocardial microtubule-associated protein-1 LC-3 (light chain 3)-II levels and LC3-II/-I ratios as well as levels of cathepsin D and ATP. In addition, increased numbers of autophagic vacuoles and lysosomes were accompanied increased AMP-activated protein kinase activity and suppression of mammalian target of rapamycin phosphorylation. Finally, autophagic flux assays using short-term chloroquine treatment revealed that autophagy was activated in δ-sarcoglycan-deficient hearts and was further augmented by metformin treatment. CONCLUSIONS Metformin is a beneficial pharmacological tool that mitigates heart failure caused by δ-sarcoglycan deficiency in association with enhanced autophagy.
Collapse
Affiliation(s)
- Hiromitsu Kanamori
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Genki Naruse
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Akihiro Yoshida
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Shingo Minatoguchi
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Takatomo Watanabe
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Tomonori Kawaguchi
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Yoshihisa Yamada
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Atsushi Mikami
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Masanori Kawasaki
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Genzou Takemura
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan.,Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.)
| | - Shinya Minatoguchi
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| |
Collapse
|
48
|
Accorsi A, Cramer ML, Girgenrath M. Fibrogenesis in LAMA2-Related Muscular Dystrophy Is a Central Tenet of Disease Etiology. Front Mol Neurosci 2020; 13:3. [PMID: 32116541 PMCID: PMC7010923 DOI: 10.3389/fnmol.2020.00003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
LAMA2-related congenital muscular dystrophy, also known as MDC1A, is caused by loss-of-function mutations in the alpha2 chain of Laminin-211. Loss of this protein interrupts the connection between the muscle cell and its extracellular environment and results in an aggressive, congenital-onset muscular dystrophy characterized by severe hypotonia, lack of independent ambulation, and early mortality driven by respiratory complications and/or failure to thrive. Of the pathomechanisms of MDC1A, the earliest and most prominent is widespread and rampant fibrosis. Here, we will discuss some of the key drivers of fibrosis including TGF-beta and renin–angiotensin system signaling and consequences of these pathways including myofibroblast transdifferentiation and matrix remodeling. We will also highlight some of the differences in fibrogenesis in congenital muscular dystrophy (CMD) with that seen in Duchenne muscular dystrophy (DMD). Finally, we will connect the key signaling pathways in the pathogenesis of MDC1A to the current status of the therapeutic approaches that have been tested in the preclinical models of MDC1A to treat fibrosis.
Collapse
Affiliation(s)
| | - Megan L Cramer
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, United States
| | | |
Collapse
|
49
|
Lindsay A, Baumann CW, Rebbeck RT, Yuen SL, Southern WM, Hodges JS, Cornea RL, Thomas DD, Ervasti JM, Lowe DA. Mechanical factors tune the sensitivity of mdx muscle to eccentric strength loss and its protection by antioxidant and calcium modulators. Skelet Muscle 2020; 10:3. [PMID: 32007101 PMCID: PMC6995146 DOI: 10.1186/s13395-020-0221-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
Background Dystrophin deficiency sensitizes skeletal muscle of mice to eccentric contraction (ECC)-induced strength loss. ECC protocols distinguish dystrophin-deficient from healthy, wild type muscle, and test the efficacy of therapeutics for Duchenne muscular dystrophy (DMD). However, given the large lab-to-lab variability in ECC-induced strength loss of dystrophin-deficient mouse skeletal muscle (10–95%), mechanical factors of the contraction likely impact the degree of loss. Therefore, the purpose of this study was to evaluate the extent to which mechanical variables impact sensitivity of dystrophin-deficient mouse skeletal muscle to ECC. Methods We completed ex vivo and in vivo muscle preparations of the dystrophin-deficient mdx mouse and designed ECC protocols within physiological ranges of contractile parameters (length change, velocity, contraction duration, and stimulation frequencies). To determine whether these contractile parameters affected known factors associated with ECC-induced strength loss, we measured sarcolemmal damage after ECC as well as strength loss in the presence of the antioxidant N-acetylcysteine (NAC) and small molecule calcium modulators that increase SERCA activity (DS-11966966 and CDN1163) or lower calcium leak from the ryanodine receptor (Chloroxine and Myricetin). Results The magnitude of length change, work, and stimulation duration ex vivo and in vivo of an ECC were the most important determinants of strength loss in mdx muscle. Passive lengthening and submaximal stimulations did not induce strength loss. We further showed that sarcolemmal permeability was associated with muscle length change, but it only accounted for a minimal fraction (21%) of the total strength loss (70%). The magnitude of length change also significantly influenced the degree to which NAC and small molecule calcium modulators protected against ECC-induced strength loss. Conclusions These results indicate that ECC-induced strength loss of mdx skeletal muscle is dependent on the mechanical properties of the contraction and that mdx muscle is insensitive to ECC at submaximal stimulation frequencies. Rigorous design of ECC protocols is critical for effective use of strength loss as a readout in evaluating potential therapeutics for muscular dystrophy.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, MMC 388, 420 Delaware Street SE, Minneapolis, 55455, USA.,Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA.,Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, 3220, Australia
| | - Cory W Baumann
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, MMC 388, 420 Delaware Street SE, Minneapolis, 55455, USA
| | - Robyn T Rebbeck
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - Samantha L Yuen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - William M Southern
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - James S Hodges
- Division of Biostatistics, University of Minnesota, A460 Mayo Building, 420 Delaware Street SE, Minneapolis, 55455, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - Dawn A Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, MMC 388, 420 Delaware Street SE, Minneapolis, 55455, USA.
| |
Collapse
|
50
|
Yu L, Zhang X, Yang Y, Li D, Tang K, Zhao Z, He W, Wang C, Sahoo N, Converso-Baran K, Davis CS, Brooks SV, Bigot A, Calvo R, Martinez NJ, Southall N, Hu X, Marugan J, Ferrer M, Xu H. Small-molecule activation of lysosomal TRP channels ameliorates Duchenne muscular dystrophy in mouse models. SCIENCE ADVANCES 2020; 6:eaaz2736. [PMID: 32128386 PMCID: PMC7032923 DOI: 10.1126/sciadv.aaz2736] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/22/2019] [Indexed: 05/12/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease caused by mutations in dystrophin that compromise sarcolemma integrity. Currently, there is no treatment for DMD. Mutations in transient receptor potential mucolipin 1 (ML1), a lysosomal Ca2+ channel required for lysosomal exocytosis, produce a DMD-like phenotype. Here, we show that transgenic overexpression or pharmacological activation of ML1 in vivo facilitates sarcolemma repair and alleviates the dystrophic phenotypes in both skeletal and cardiac muscles of mdx mice (a mouse model of DMD). Hallmark dystrophic features of DMD, including myofiber necrosis, central nucleation, fibrosis, elevated serum creatine kinase levels, reduced muscle force, impaired motor ability, and dilated cardiomyopathies, were all ameliorated by increasing ML1 activity. ML1-dependent activation of transcription factor EB (TFEB) corrects lysosomal insufficiency to diminish muscle damage. Hence, targeting lysosomal Ca2+ channels may represent a promising approach to treat DMD and related muscle diseases.
Collapse
MESH Headings
- Animals
- Biomarkers
- Biopsy
- Disease Models, Animal
- Dystrophin/genetics
- Fluorescent Antibody Technique
- Gene Expression
- Lysosomes/drug effects
- Lysosomes/metabolism
- Mice
- Mice, Inbred mdx
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Transient Receptor Potential Channels/agonists
Collapse
Affiliation(s)
- Lu Yu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
| | - Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
| | - Yexin Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Kaiyuan Tang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
| | - Zifan Zhao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
| | - Wanwan He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ce Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
| | - Nirakar Sahoo
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
- Department of Biology, The University of Texas Rio Grande Valley, 1201 W University Dr., Edinburg, TX 78539, USA
| | - Kimber Converso-Baran
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carol S. Davis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Susan V. Brooks
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anne Bigot
- Sorbonne Université, INSERM, AIM, Center for Research in Myology, UMRS974, GH Pitié-Salpétrière, 75651 Paris Cedex 13, France
| | - Raul Calvo
- NIH/NCATS/NCGC, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | | | - Noel Southall
- NIH/NCATS/NCGC, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Xin Hu
- NIH/NCATS/NCGC, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Juan Marugan
- NIH/NCATS/NCGC, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Marc Ferrer
- NIH/NCATS/NCGC, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 4114 Biological Sciences Building, 1105 North University, Ann Arbor, MI 48109, USA
- Corresponding author.
| |
Collapse
|