1
|
Tammineni ER, Manno C, Oza G, Figueroa L. Skeletal muscle disorders as risk factors for type 2 diabetes. Mol Cell Endocrinol 2025; 599:112466. [PMID: 39848431 PMCID: PMC11886953 DOI: 10.1016/j.mce.2025.112466] [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: 11/12/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The incidence and prevalence of muscular disorders and of type 2 diabetes (T2D) is increasing and both represent highly significant healthcare problems, both economically and compromising quality of life. Interestingly, skeletal muscle dysfunction and T2D share some commonalities including dysregulated glucose homeostasis, increased oxidative stress, dyslipidemia, and cytokine alterations. Several lines of evidence have hinted to a relationship between skeletal muscle dysfunction and T2D. For instance, T2D affects skeletal muscle morphology, functionality, and overall health through altered protein metabolism, impaired mitochondrial function, and ultimately cell viability. Conversely, humans suffering from myopathies and their experimental models demonstrated increased incidence of T2D through altered muscle glucose disposal function due to abnormal calcium homeostasis, compromised mitochondrial function, dyslipidemia, increased inflammatory cytokines and fiber size alterations and disproportions. Lifestyle modifications are essential for improving and maintaining mobility and metabolic health in individuals suffering from myopathies along with T2D. In this review, we updated current literature evidence on clinical incidence of T2D in inflammatory, mitochondrial, metabolic myopathies, and muscular dystrophies and further discussed the molecular basis of these skeletal muscle disorders leading to T2D.
Collapse
Affiliation(s)
| | - Carlo Manno
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| | - Goldie Oza
- Centro de Investigación y Desarrollo Tecnológico en Electroquímica S. C., Queretaro, Mexico
| | - Lourdes Figueroa
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| |
Collapse
|
2
|
Dong M, Maturana AD. Effects of aging on calcium channels in skeletal muscle. Front Mol Biosci 2025; 12:1558456. [PMID: 40177518 PMCID: PMC11961898 DOI: 10.3389/fmolb.2025.1558456] [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: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
In skeletal muscle, calcium is not only essential to stimulate and sustain their contractions but also for muscle embryogenesis, regeneration, energy production in mitochondria, and fusion. Different ion channels contribute to achieving the various functions of calcium in skeletal muscles. Muscle contraction is initiated by releasing calcium from the sarcoplasmic reticulum through the ryanodine receptor channels gated mechanically by four dihydropyridine receptors of T-tubules. The calcium influx through store-operated calcium channels sustains the contraction and stimulates muscle regeneration. Mitochondrial calcium uniporter allows the calcium entry into mitochondria to stimulate oxidative phosphorylation. Aging alters the expression and activity of these different calcium channels, resulting in a reduction of skeletal muscle force generation and regeneration capacity. Regular physical training and bioactive molecules from nutrients can prevent the effects of aging on calcium channels. This review focuses on the current knowledge of the effects of aging on skeletal muscles' calcium channels.
Collapse
Affiliation(s)
| | - Andrés Daniel Maturana
- Department of Applied Biosciences, Graduate School of Bioagricultural Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
3
|
Schreiber J, Rotard L, Tourneur Y, Lafoux A, Berthier C, Allard B, Huchet C, Jacquemond V. Reduced voltage-activated Ca2+ release flux in muscle fibers from a rat model of Duchenne dystrophy. J Gen Physiol 2025; 157:e202413588. [PMID: 39718509 DOI: 10.1085/jgp.202413588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/14/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024] Open
Abstract
The potential pathogenic role of disturbed Ca2+ homeostasis in Duchenne muscular dystrophy (DMD) remains a complex, unsettled issue. We used muscle fibers isolated from 3-mo-old DMDmdx rats to further investigate the case. Most DMDmdx fibers exhibited no sign of trophic or morphology distinction as compared with WT fibers and mitochondria and t-tubule membrane networks also showed no stringent discrepancy. Under voltage clamp, values for holding current were similar in the two groups, whereas values for capacitance were larger in DMDmdx fibers, suggestive of enhanced amount of t-tubule membrane. The Ca2+ current density across the channel carried by the EC coupling voltage sensor (CaV1.1) was unchanged. The maximum rate of voltage-activated sarcoplasmic reticulum (SR) Ca2+ release was reduced by 25% in the DMDmdx fibers, with no change in voltage dependency. Imaging resting Ca2+ revealed rare spontaneous local SR Ca2+ release events with no sign of elevated activity in DMDmdx fibers. Under current clamp, DMDmdx fibers generated similar trains of action potentials as WT fibers. Results suggest that reduced peak amplitude of SR Ca2+ release is an inherent feature of this DMD model, likely contributing to muscle weakness. This occurs despite a preserved amount of releasable Ca2+ and with no change in excitability, CaV1.1 channel activity, and SR Ca2+ release at rest. Although we cannot exclude that fibers from the 3-mo-old animals do not yet display a fully developed disease phenotype, results provide limited support for pathomechanistic concepts frequently associated with DMD such as membrane fragility, excessive Ca2+ entry, or enhanced SR Ca2+ leak.
Collapse
Affiliation(s)
- Jonathan Schreiber
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Ludivine Rotard
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Yves Tourneur
- UFPE Department Nutrição, Cidade Universitária, Recife, Brazil
| | - Aude Lafoux
- Therassay Platform, CAPACITES, Nantes Université , Nantes, France
| | - Christine Berthier
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Bruno Allard
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Corinne Huchet
- Therassay Platform, CAPACITES, Nantes Université , Nantes, France
- Nantes Gene Therapy Laboratory, Nantes Université, INSERM UMR TARGET 1089, Nantes, France
| | - Vincent Jacquemond
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| |
Collapse
|
4
|
Stephenson KA, Barron A, Rae MG, O'Malley D. Inhibition of hippocampal interleukin-6 receptor-evoked signalling normalises long-term potentiation in dystrophin-deficient mdx mice. Brain Behav Immun Health 2025; 43:100935. [PMID: 39867844 PMCID: PMC11762146 DOI: 10.1016/j.bbih.2024.100935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/30/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked neuromuscular disorder, characterised by progressive immobility, chronic inflammation and premature death, is caused by the loss of the mechano-transducing signalling molecule, dystrophin. In non-contracting cells, such as neurons, dystrophin is likely to have a functional role in synaptic plasticity, anchoring post-synaptic receptors. Dystrophin-expressing hippocampal neurons are key to cognitive functions such as emotions, learning and the consolidation of memories. In the context of disease-induced chronic inflammation, we have explored the role of the pleiotropic cytokine, interleukin (IL)-6 in hippocampal dysfunction using immunofluorescence, electrophysiology and metabolic measurements in dystrophic mdx mice. Hippocampal long-term potentiation (LTP) of the Schaffer collateral-CA1 projections was suppressed in mdx slices. Given the importance of mitochondria-generated ATP in synaptic plasticity, reduced maximal respiration in the CA1 region may impact upon this process. Consistent with a role for IL-6 in this observation, early LTP was suppressed in dystrophin-expressing wildtype slices exposed to IL-6. In dystrophic mdx mice, exposure to IL-6 suppressed mitochondrial-mediated basal metabolism in CA1, CA3 and DG hippocampal regions. Furthermore, blocking IL-6-mediated signalling by administering neutralising monoclonal IL-6 receptor antibodies intrathecally, normalised LTP in mdx mice. The impact of dystrophin loss from the hippocampus was associated with modified cellular bioenergetics, which underpin energy-driven processes such as the induction of LTP. The additional challenge of pathophysiological levels of IL-6 resulted in altered cellular bioenergetics, which may be key to cognitive deficits associated with the loss of dystrophin.
Collapse
Affiliation(s)
- Kimberley A. Stephenson
- Department of Physiology, School of Medicine, University College Cork, Western Road, Cork, Ireland
| | - Aaron Barron
- Department of Anatomy and Neuroscience, University College Cork, Western Road, Cork, Ireland
| | - Mark G. Rae
- Department of Physiology, School of Medicine, University College Cork, Western Road, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, School of Medicine, University College Cork, Western Road, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Western Road, Cork, Ireland
| |
Collapse
|
5
|
Henzi BC, Putananickal N, Schmidt S, Nagy S, Rubino-Nacht D, Schaedelin S, Amthor H, Childs AM, Deconinck N, Horrocks I, Houwen-van Opstal S, Laugel V, Lobato ML, Osorio AN, Schara-Schmidt U, Spinty S, von Moers A, Lawrence F, Hafner P, Dorchies OM, Fischer D. Safety and efficacy of tamoxifen in non-ambulant patients with Duchenne muscular dystrophy: a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial (TAMDMD Group B). Neuromuscul Disord 2025; 47:105275. [PMID: 39879732 DOI: 10.1016/j.nmd.2025.105275] [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: 09/16/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
Most patients with Duchenne muscular dystrophy (DMD) are non-ambulant. Preserving proximal motor function is crucial, rarely studied. Tamoxifen, a selective oestrogen receptor modulator, reduced signs of muscular pathology in a DMD mouse model. Our objective was to assess the safety and efficacy of tamoxifen over 48 weeks in non-ambulant DMD patients. In this multicentre, randomised, double-blind, placebo-controlled, phase 3 trial at six European centres boys aged 10-16 years with genetically diagnosed DMD, non-ambulant and off corticosteroid treatment for ≥6 months, randomly assigned (1:1) to either 20 mg/day tamoxifen orally or placebo were included. The primary outcome was change in D2 motor function measure from baseline to week 48. Of 15 non-ambulant male patients with DMD screened, 14 were enrolled from January 24th, 2019, to January 6th, 2021. Eight patients were randomised to the treatment and six to the placebo group. The primary efficacy outcome did not differ significantly between tamoxifen and placebo (7.8 percentage points, 95 % CI, -26.82 to 11.22, p=0.359) with a trend not favouring tamoxifen. No deaths or life-threatening serious AEs occurred. Tamoxifen was safe but due to insufficient clinical evidence, it cannot be recommended as a treatment option for DMD. Trial registration: ClinicalTrials.gov (NCT03354039).
Collapse
Affiliation(s)
- Bettina C Henzi
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Niveditha Putananickal
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Simone Schmidt
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Sara Nagy
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Rubino-Nacht
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Sabine Schaedelin
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Helge Amthor
- Service de Neurologie et Réanimation Pédiatriques, APHP Paris Saclay, Hôpital Raymond Poincaré, 92380, Garches, France
| | | | - Nicolas Deconinck
- Department of Paediatric Neurology and Neuromuscular Reference Center, Hôpital Universitaire des Enfants Reine Fabiola (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Iain Horrocks
- Royal Hospital for Children, Glasgow, United Kingdom
| | - Saskia Houwen-van Opstal
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vincent Laugel
- Department of Pediatric Neurology, Strasbourg University Hospital, Strasbourg, France
| | - Mercedes Lopez Lobato
- Sección de Neurología Pediátrica, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Andrés Nascimento Osorio
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu and Center for Biomedical Research Network on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefan Spinty
- Alder Hey Children's Hospital, Liverpool, United Kingdom
| | - Arpad von Moers
- Department of Pediatrics, DRK Kliniken Berlin Westend, Berlin, Germany
| | | | - Patricia Hafner
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Olivier M Dorchies
- School of Pharmaceutical Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Dirk Fischer
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.
| |
Collapse
|
6
|
Butson C, Ntekim N, Acord S, Marks W. Genetic Panel Reveals Coexisting Neuromuscular Disorders in Patients With Duchenne Muscular Dystrophy. J Child Neurol 2025; 40:83-90. [PMID: 39429168 DOI: 10.1177/08830738241284683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Duchenne muscular dystrophy is a genetically based neuromuscular disorder characterized by progressive physical impairment and cardiomyopathy in children, leading to fatal cardiac or respiratory failure. Duchenne muscular dystrophy shares some overlapping clinical features with other disorders, complicating clinical differentiation. We hypothesized that some Duchenne muscular dystrophy patients may have a secondary neuromuscular disorders that could negatively skew data during pharmaceutical clinical trials and lead to incomplete treatment plans. Consecutive genetic panels on 353 patients were reviewed. Thirty-two (32; 9.1%) patients with Duchenne muscular dystrophy were identified. Three (3; 9.4%) were found to have at least 1 genetically confirmed secondary neuromuscular disorder. Overlooking these coexisting disorders could lead to unexpected treatment failures, potentially affecting medication efficacy in trials or commercial use. Secondary neuromuscular disorders should be considered in Duchenne muscular dystrophy patients before clinical trial enrollment or treatment planning, with expanded genetic testing, such as whole exome sequencing or whole genome sequencing, likely to reveal even more secondary disorders.
Collapse
Affiliation(s)
- Carter Butson
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - Nedeke Ntekim
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - Stephanie Acord
- Department of Neurology, Cook Children's Medical Center, Fort Worth, TX, USA
| | - Warren Marks
- Department of Neurology, Cook Children's Medical Center, Fort Worth, TX, USA
| |
Collapse
|
7
|
Dubinin MV, Stepanova AE, Igoshkina AD, Mikheeva IB, Talanov EY, Cherepanova AA, Belosludtsev KN. Effect of 2-Aminoethoxydiphenyl Borate on the State of Skeletal Muscles in Dystrophin-Deficient mdx Mice. FRONT BIOSCI-LANDMRK 2024; 29:428. [PMID: 39735998 DOI: 10.31083/j.fbl2912428] [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: 08/06/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 12/31/2024]
Abstract
OBJECTIVE Ca2+ overload of muscle fibers is one of the factors that secondarily aggravate the development of Duchenne muscular dystrophy (DMD). The purpose of this study is to evaluate the effects of the Ca2+ channel modulator 2-aminoethoxydiphenyl borate (APB) on skeletal muscle pathology in dystrophin-deficient mdx mice. METHODS Mice were randomly divided into six groups: wild type (WT), WT+3 mg/kg APB, WT+10 mg/kg APB, mdx, mdx+3 mg/kg APB, mdx+10 mg/kg APB. APB was administered intraperitoneally daily for 28 days. Finally, we assessed the grip strength and hanging time of mice, the histology and ultrastructure of the quadriceps, as well as the parameters reflecting quadricep mitochondrial function. RESULTS 3 mg/kg APB was shown to reduce creatine kinase activity in the serum, intensity of degeneration and the level of fibrosis in the quadriceps of mdx mice, and improved tissue ultrastructure. However, this effect of APB was not sufficient to improve grip strength and hanging time of mdx mice. The effect of 3 mg/kg APB may be due to improve Ca2+ homeostasis in skeletal muscles, as evidenced by a trend toward decreased Ca2+ overload of quadricep mitochondria. High dose of APB (10 mg/kg body weight) showed less pronounced effect on the pathological phenotype of mdx mice. Moreover, 10 mg/kg APB disrupted the ultrastructure of the quadriceps and caused a decrease in grip strength in WT mice. CONCLUSIONS APB is able to improve the phenotype in mdx mouse DMD model. However, the effect of APB is quite limited, which may be due to its multitargeting of Ca2+ channels in the membranes of muscle fibers and intracellular organelles, differentially expressed in DMD.
Collapse
Affiliation(s)
- Mikhail V Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, 424001 Yoshkar-Ola, Russia
| | - Anastasia E Stepanova
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, 424001 Yoshkar-Ola, Russia
| | - Anastasia D Igoshkina
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, 424001 Yoshkar-Ola, Russia
| | - Irina B Mikheeva
- Laboratory of Experimental Neurobiology, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Eugeny Yu Talanov
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Alena A Cherepanova
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, 424001 Yoshkar-Ola, Russia
| | - Konstantin N Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, 424001 Yoshkar-Ola, Russia
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| |
Collapse
|
8
|
Rajalekshmi R, Agrawal DK. Therapeutic Efficacy of Medicinal Plants with Allopathic Medicine in Musculoskeletal Diseases. INTERNATIONAL JOURNAL OF PLANT, ANIMAL AND ENVIRONMENTAL SCIENCES 2024; 14:104-129. [PMID: 39866300 PMCID: PMC11765655 DOI: 10.26502/ijpaes.4490170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Musculoskeletal diseases encompass a diverse array of disorders affecting the muscles, bones, joints, and connective tissues, leading to significant impairments in mobility, function, and quality of life. Affecting over 1.3 billion individuals globally, musculoskeletal diseases represent a major source of disability and economic burden. Conventional treatment modalities, including pharmacological interventions and surgical procedures, are frequently limited by adverse side effects, prolonged recovery periods, and patient dissatisfaction, particularly when focused solely on symptom management. In response, complementary and alternative medicine, particularly the use of medicinal plants, has garnered increasing interest to enhance the management of musculoskeletal diseases. Medicinal plants possess a wide spectrum of pharmacologically active compounds with anti-inflammatory, analgesic, and antioxidant properties, making them promising adjuncts to conventional therapies. This review critically evaluates the potential synergy between medicinal plants and allopathic medicine for the management of musculoskeletal diseases, with an emphasis on integrated therapy that combines both modalities. Specifically, a critical discussion is presented on how medicinal plants with scientifically supported pharmacological properties can augment the therapeutic efficacy of conventional medications, reduce their doses, and mitigate adverse effects. Furthermore, the challenges associated with incorporating herbal medicine into established healthcare systems are discussed, including the need for rigorous clinical validation, standardization, and regulatory frameworks. Overall, the article underscores the potential of integrated therapeutic approaches to improve clinical outcomes, enhance patient well-being, and establish a more sustainable model for the treatment of musculoskeletal diseases.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
9
|
Konieczny P. Systemic Treatment of Body-Wide Duchenne Muscular Dystrophy Symptoms. Clin Pharmacol Ther 2024; 116:1472-1484. [PMID: 38965715 DOI: 10.1002/cpt.3363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/09/2024] [Indexed: 07/06/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked disease that leads to premature death due to the loss of dystrophin. Current strategies predominantly focus on the therapeutic treatment of affected skeletal muscle tissue. However, certain results point to the fact that with successful treatment of skeletal muscle, DMD-exposed latent phenotypes in tissues, such as cardiac and smooth muscle, might lead to adverse effects and even death. Likewise, it is now clear that the absence of dystrophin affects the function of the nervous system, and that this phenotype is more pronounced when shorter dystrophins are absent, in addition to the full-length dystrophin that is present predominantly in the muscle. Here, I focus on the systemic aspects of DMD, highlighting the ubiquitous expression of the dystrophin gene in human tissues. Furthermore, I describe therapeutic strategies that have been tested in the clinic and point to unresolved questions regarding the function of distinct dystrophin isoforms, and the possibility of current therapeutic strategies to tackle phenotypes that relate to their absence.
Collapse
Affiliation(s)
- Patryk Konieczny
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
10
|
Jepsen WM, Fazenbaker A, Ramsey K, Bonfitto A, Naymik M, Turner B, Sloan J, Tiwari N, Bernes SM, Neilson DE, Sanchez-Castillo M, Huentelman MJ, Narayanan V. Duchenne Muscular Dystrophy in Two Half-Brothers Due to Inherited 306 Kb Inverted Insertion of 10p15.1 into Intron 44 of the Dp427m Transcript of the DMD Gene. Int J Mol Sci 2024; 25:11922. [PMID: 39595988 PMCID: PMC11593467 DOI: 10.3390/ijms252211922] [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: 10/08/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare genetic disorder caused by the absence of a fully functional dystrophin protein in myocytes. In skeletal muscle, the lack of dystrophin ultimately results in muscle wasting and the replacement of myocytes with fatty or fibrous tissues. In the heart, cardiomyocytes eventually fail and cause fatal cardiomyopathy. We present a case of a male patient and his younger brother with a maternally inherited inverted insertion of approximately 306 kb of chromosome 10 in the deep intronic region between exons 44 and 45 of the DMD gene, leading to Duchenne muscular dystrophy. Chromosomal microarray, comprehensive muscular dystrophy genetic testing, and whole exome sequencing were negative. Targeted transcriptome RNA sequencing at an external lab showed no aberrant splicing. Research whole genome sequencing identified the copy number gain and insertion. Subsequent reanalysis of the RNA sequencing data showed possible aberrant splicing involving DMD exons 44-45, and research RNA sequencing revealed a fusion between the DMD gene on the minus strand of chromosome X and the PFKFB3 gene on the plus strand of chromosome 10. We demonstrate that whole genome sequencing can be valuable for identifying intronic events in the DMD gene previously undetected or not reported by traditional clinical testing.
Collapse
Affiliation(s)
- Wayne M. Jepsen
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | | | - Keri Ramsey
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Anna Bonfitto
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Marcus Naymik
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Bryce Turner
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Jennifer Sloan
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | | | | | | | - Meredith Sanchez-Castillo
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Matt J. Huentelman
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| |
Collapse
|
11
|
Maiga AB, Pamanta I, Bamba S, Cissé L, Diarra S, Touré S, Yalcouyé A, Diallo S, Diallo S, Kané F, Diallo SH, Ba HO, Guinto CO, Fischbeck K, Landoure G, Cissé IA. A Novel Splice Site Variant in COL6A1 Causes Ullrich Congenital Muscular Dystrophy in a Consanguineous Malian Family. Mol Genet Genomic Med 2024; 12:e70032. [PMID: 39523858 PMCID: PMC11551527 DOI: 10.1002/mgg3.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Congenital muscular dystrophies (CMDs) are diverse early-onset conditions affecting skeletal muscle and connective tissue. This group includes collagen VI-related dystrophies such as Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM), caused by mutations in the COL6A1, COL6A2 and COL6A3 genes. We report a consanguineous Malian family with three siblings affected by UCMD due to a novel homozygous splice site variant in the COL6A1 gene. METHODS After obtaining consent, three affected siblings and their relatives underwent physical examinations by specialists and laboratory tests where possible. DNA was extracted from peripheral blood for genetic testing, including Whole Exome Sequencing (WES). Putative variants were confirmed through Sanger Sequencing and assessed for pathogenicity using in silico tools. RESULTS The three siblings and their healthy parents, from a consanguineous marriage, presented with early-onset progressive muscle weakness, walking difficulty, proximal motor deficits, severe muscle atrophy, hypotonia, skeletal deformities, joint hyperlaxity, ankyloses at the elbows and knees, keloid scars and dental crowding. No cardiac involvement was detected and creatine kinase (CK) levels were normal. All had low serum calcium levels, treated with oral supplements. Needle myography indicated myopathic patterns. WES identified a novel splice site variant in the first intron of COL6A1 (c.98-1G>C), which segregated with the disease within the family. This variant is predicted to cause exon 2 skipping in COL6A1, with a high CADD score of 33 and Splice AI predicting it as deleterious. CONCLUSION We identified a novel COL6A1 variant in a consanguineous family, highlighting the need for further studies in larger African cohorts to enhance genetic epidemiology and prepare for future therapeutic research.
Collapse
Affiliation(s)
- Alassane Baneye Maiga
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
| | - Ibrahim Pamanta
- Service de RhumatologieCentre Hospitalier Universitaire “Point G”BamakoMali
| | - Salia Bamba
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
- Yale University Pediatric Genomics Discovery ProgramNew HavenConnecticutUSA
| | - Lassana Cissé
- Service de MédecineHôpital Régional “Nianankoro Fomba”SégouMali
| | - Salimata Diarra
- Yale University Pediatric Genomics Discovery ProgramNew HavenConnecticutUSA
- Neurogenetic Branch, NINDS, NIHBethesdaMarylandUSA
| | - Sidi Touré
- Service de RhumatologieCentre Hospitalier Universitaire “Point G”BamakoMali
| | - Abdoulaye Yalcouyé
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
- McKusick‐Nathans Institute and Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Seydou Diallo
- Service de RhumatologieCentre Hospitalier Universitaire “Point G”BamakoMali
| | - Salimata Diallo
- Service de NeurologieCentre Hospitalier Universitaire “Gabriel Touré”BamakoMali
| | - Fousseyni Kané
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
| | | | - Hamidou Oumar Ba
- Service de CardiologieCentre Hospitalier Universitaire “Gabriel Touré”BamakoMali
| | - Cheick Oumar Guinto
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
- Service de NeurologieCentre Hospitalier Universitaire “Point G”BamakoMali
| | | | - Guida Landoure
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
- Service de NeurologieCentre Hospitalier Universitaire “Point G”BamakoMali
| | - Idrissa Ahmadou Cissé
- Faculté de Médecine et d'OdontostomatologieUniversité des Sciences, des Techniques et des Technologies de BamakoBamakoMali
- Service de RhumatologieCentre Hospitalier Universitaire “Point G”BamakoMali
| | | |
Collapse
|
12
|
Barrett P, Louie KW, Dupont JB, Mack DL, Maves L. Uncovering the Embryonic Origins of Duchenne Muscular Dystrophy. WIREs Mech Dis 2024; 16:e1653. [PMID: 39444092 PMCID: PMC11563919 DOI: 10.1002/wsbm.1653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/11/2024] [Accepted: 09/28/2024] [Indexed: 10/25/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a severe degenerative muscle disease caused by mutations in the DMD gene, which encodes dystrophin. Despite its initial description in the late 19th century by French neurologist Guillaume Duchenne de Boulogne, and identification of causal DMD genetic mutations in the 1980s, therapeutics remain challenging. The current standard of care is corticosteroid treatment, which delays the progression of muscle dysfunction but is associated with significant adverse effects. Emerging therapeutic approaches, including AAV-mediated gene transfer, CRISPR gene editing, and small molecule interventions, are under development but face considerable obstacles. Although DMD is viewed as a progressive muscle disease, muscle damage and abnormal molecular signatures are already evident during fetal myogenesis. This early onset of pathology suggests that the limited success of current therapies may partly be due to their administration after aberrant embryonic myogenesis has occurred in the absence of dystrophin. Consequently, identifying optimal therapeutic strategies and intervention windows for DMD may depend on a better understanding of the earliest DMD disease mechanisms. As newer techniques are applied, the field is gaining increasingly detailed insights into the early muscle developmental abnormalities in DMD. A comprehensive understanding of the initial events in DMD pathogenesis and progression will facilitate the generation and testing of effective therapeutic interventions.
Collapse
Affiliation(s)
- Philip Barrett
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA
| | - Ke'ale W Louie
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | | | - David L Mack
- Departments of Rehabilitation Medicine, Bioengineering and Neurobiology & Biophysics, Institute for Stem Cell and Regenerative Medicine, University of Washington Medicine, Seattle, Washington, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
13
|
Morales ED, Wang D, Burke MJ, Han J, Devine DD, Zhang K, Duan D. Transcriptional changes of genes encoding sarcoplasmic reticulum calcium binding and up-taking proteins in normal and Duchenne muscular dystrophy dogs. BMC Musculoskelet Disord 2024; 25:811. [PMID: 39402529 PMCID: PMC11472500 DOI: 10.1186/s12891-024-07927-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Cytosolic calcium overload contributes to muscle degradation in Duchenne muscular dystrophy (DMD). The sarcoplasmic reticulum (SR) is the primary calcium storage organelle in muscle. The sarco-endoplasmic reticulum ATPase (SERCA) pumps cytosolic calcium to the SR during muscle relaxation. Calcium is kept in the SR by calcium-binding proteins. METHODS Given the importance of the canine DMD model in translational studies, we examined transcriptional changes of SERCA (SERCA1 and SERCA2a), SERCA regulators (phospholamban, sarcolipin, myoregulin, and dwarf open reading frame), and SR calcium-binding proteins (calreticulin, calsequestrin 1, calsequestrin 2, and sarcalumenin) in skeletal muscle (diaphragm and extensor carpi ulnaris) and heart (left ventricle) in normal and affected male dogs by droplet digital PCR before the onset (≤ 2-m-old), at the active stage (8 to 16-m-old), and at the terminal stage (30 to 50-m-old) of the disease. Since many of these proteins are expressed in a fiber type-specific manner, we also evaluated fiber type composition in skeletal muscle. RESULTS In affected dog skeletal muscle, SERCA and its regulators were down-regulated at the active stage, but calcium-binding proteins (except for calsequestrin 1) were upregulated at the terminal stage. Surprisingly, nominal differences were detected in the heart. We also examined whether there exists sex-biased expression in 8 to 16-m-old dogs. Multiple transcripts were significantly downregulated in the heart and extensor carpi ulnaris muscle of female dogs. In fiber type analysis, we found significantly more type I fiber in the diaphragm of 8 to 16-m-old affected dogs, and significantly more type II fibers in the extensor carpi ulnaris of 30 to 50-m-old affected dogs. However, no difference was detected between male and female dogs. CONCLUSIONS Our study adds new knowledge to the understanding of muscle calcium regulation in normal and dystrophic canines.
Collapse
Affiliation(s)
- Emily D Morales
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Dongxin Wang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Matthew J Burke
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Jin Han
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Drake D Devine
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA.
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA.
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO, 65212, USA.
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
14
|
Siemionow M, Bocian K, Bozyk KT, Ziemiecka A, Siemionow K. Chimeric Cell Therapy Transfers Healthy Donor Mitochondria in Duchenne Muscular Dystrophy. Stem Cell Rev Rep 2024; 20:1819-1829. [PMID: 39017908 PMCID: PMC11445288 DOI: 10.1007/s12015-024-10756-w] [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] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a severe X-linked disorder characterized by dystrophin gene mutations and mitochondrial dysfunction, leading to progressive muscle weakness and premature death of DMD patients. We developed human Dystrophin Expressing Chimeric (DEC) cells, created by the fusion of myoblasts from normal donors and DMD patients, as a foundation for DT-DEC01 therapy for DMD. Our preclinical studies on mdx mouse models of DMD revealed enhanced dystrophin expression and functional improvements in cardiac, respiratory, and skeletal muscles after systemic intraosseous DEC administration. The current study explored the feasibility of mitochondrial transfer and fusion within the created DEC cells, which is crucial for developing new therapeutic strategies for DMD. Following mitochondrial staining with MitoTracker Deep Red and MitoTracker Green dyes, mitochondrial fusion and transfer was assessed by Flow cytometry (FACS) and confocal microscopy. The PEG-mediated fusion of myoblasts from normal healthy donors (MBN/MBN) and normal and DMD-affected donors (MBN/MBDMD), confirmed the feasibility of myoblast and mitochondrial fusion and transfer. The colocalization of the mitochondrial dyes MitoTracker Deep Red and MitoTracker Green confirmed the mitochondrial chimeric state and the creation of chimeric mitochondria, as well as the transfer of healthy donor mitochondria within the created DEC cells. These findings are unique and significant, introducing the potential of DT-DEC01 therapy to restore mitochondrial function in DMD patients and in other diseases where mitochondrial dysfunction plays a critical role.
Collapse
Affiliation(s)
- Maria Siemionow
- Chair and Department of Traumatology, Orthopedics and Surgery of the Hand, Poznan University of Medical Sciences, Poznan, 61‑545, Poland.
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland.
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Katarzyna Bocian
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, 02-096, Poland
- Polish Stem Cell Bank, FamiCord Group, Warsaw, 00-867, Poland
| | - Katarzyna T Bozyk
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland
| | - Anna Ziemiecka
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland
| | - Krzysztof Siemionow
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
15
|
Stirm M, Klymiuk N, Nagashima H, Kupatt C, Wolf E. Pig models for translational Duchenne muscular dystrophy research. Trends Mol Med 2024; 30:950-964. [PMID: 38749865 DOI: 10.1016/j.molmed.2024.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 10/12/2024]
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the X-linked DMD gene, resulting in the absence of dystrophin, progressive muscle degeneration, and heart failure. Genetically tailored pig models resembling human DMD mutations recapitulate the biochemical, clinical, and pathological hallmarks of DMD with an accelerated disease progression compared to human patients. DMD pigs have been used to evaluate therapeutic concepts such as gene editing to reframe a disrupted DMD reading frame or the delivery of artificial chromosome vectors carrying the complete DMD gene. Moreover, DMD pigs have been instrumental in validating new diagnostic modalities such as multispectral optoacoustic tomography (MSOT) for non-invasive monitoring of disease progression. DMD pigs may thus help to bridge the gap between proof-of-concept studies in cellular or rodent models and clinical studies in patients.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany
| | - Nikolai Klymiuk
- Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Large Animal Models in Cardiovascular Research, Internal Medical Department I, Technical University of Munich (TU Munich), 81675 Munich, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa 214-8571, Japan
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, 81675 Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
16
|
Slick RA, Sutton J, Haberman M, O'Brien BS, Tinklenberg JA, Mardikar A, Prom MJ, Beatka M, Gartz M, Vanden Avond MA, Siebers E, Mack DL, Gonzalez JP, Ebert AD, Nagaraju K, Lawlor MW. High mobility group box 1 (HMGB1) is a potential disease biomarker in cell and mouse models of Duchenne muscular dystrophy. Biol Open 2024; 13:bio060542. [PMID: 39158383 PMCID: PMC11391821 DOI: 10.1242/bio.060542] [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: 05/26/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder affecting 1:3500 male births and is associated with myofiber degeneration, regeneration, and inflammation. Glucocorticoid treatments have been the standard of care due to immunomodulatory/immunosuppressive properties but novel genetic approaches, including exon skipping and gene replacement therapy, are currently being developed. The identification of additional biomarkers to assess DMD-related inflammatory responses and the potential efficacy of these therapeutic approaches are thus of critical importance. The current study uses RNA sequencing of skeletal muscle from two mdx mouse models to identify high mobility group box 1 (HMGB1) as a candidate biomarker potentially contributing to DMD-related inflammation. HMGB1 protein content was increased in a human iPSC-derived skeletal myocyte model of DMD and microdystrophin treatment decreased HMGB1 back to control levels. In vivo, HMGB1 protein levels were increased in vehicle treated B10-mdx skeletal muscle compared to B10-WT and significantly decreased in B10-mdx animals treated with adeno-associated virus (AAV)-microdystrophin. However, HMGB1 protein levels were not increased in D2-mdx skeletal muscle compared to D2-WT, demonstrating a strain-specific difference in DMD-related immunopathology.
Collapse
Affiliation(s)
- Rebecca A. Slick
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jessica Sutton
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Margaret Haberman
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Benjamin S. O'Brien
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer A. Tinklenberg
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aashay Mardikar
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mariah J. Prom
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Margaret Beatka
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Melanie Gartz
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark A. Vanden Avond
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Emily Siebers
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - David L. Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98104, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98104, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98104, USA
| | | | - Allison D. Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kanneboyina Nagaraju
- AGADA BioSciences Inc., Halifax, Nova Scotia, B3H0A8, Canada
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY. Binghamton, NY 13902, USA
| | - Michael W. Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| |
Collapse
|
17
|
Dubinin MV, Stepanova AE, Mikheeva IB, Igoshkina AD, Cherepanova AA, Talanov EY, Khoroshavina EI, Belosludtsev KN. Reduction of Mitochondrial Calcium Overload via MKT077-Induced Inhibition of Glucose-Regulated Protein 75 Alleviates Skeletal Muscle Pathology in Dystrophin-Deficient mdx Mice. Int J Mol Sci 2024; 25:9892. [PMID: 39337383 PMCID: PMC11432509 DOI: 10.3390/ijms25189892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Duchenne muscular dystrophy is secondarily accompanied by Ca2+ excess in muscle fibers. Part of the Ca2+ accumulates in the mitochondria, contributing to the development of mitochondrial dysfunction and degeneration of muscles. In this work, we assessed the effect of intraperitoneal administration of rhodacyanine MKT077 (5 mg/kg/day), which is able to suppress glucose-regulated protein 75 (GRP75)-mediated Ca2+ transfer from the sarcoplasmic reticulum (SR) to mitochondria, on the Ca2+ overload of skeletal muscle mitochondria in dystrophin-deficient mdx mice and the concomitant mitochondrial dysfunction contributing to muscle pathology. MKT077 prevented Ca2+ overload of quadriceps mitochondria in mdx mice, reduced the intensity of oxidative stress, and improved mitochondrial ultrastructure, but had no effect on impaired oxidative phosphorylation. MKT077 eliminated quadriceps calcification and reduced the intensity of muscle fiber degeneration, fibrosis level, and normalized grip strength in mdx mice. However, we noted a negative effect of MKT077 on wild-type mice, expressed as a decrease in the efficiency of mitochondrial oxidative phosphorylation, SR stress development, ultrastructural disturbances in the quadriceps, and a reduction in animal endurance in the wire-hanging test. This paper discusses the impact of MKT077 modulation of mitochondrial dysfunction on the development of skeletal muscle pathology in mdx mice.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Calcium/metabolism
- Disease Models, Animal
- Dystrophin/metabolism
- Dystrophin/deficiency
- Dystrophin/genetics
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mitochondria/metabolism
- Mitochondria/drug effects
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/pathology
- Mitochondria, Muscle/ultrastructure
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Oxidative Phosphorylation/drug effects
- Oxidative Stress/drug effects
- Sarcoplasmic Reticulum/metabolism
- Sarcoplasmic Reticulum/drug effects
Collapse
Affiliation(s)
- Mikhail V Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Anastasia E Stepanova
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Irina B Mikheeva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
| | - Anastasia D Igoshkina
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Alena A Cherepanova
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Eugeny Yu Talanov
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
| | - Ekaterina I Khoroshavina
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Konstantin N Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
| |
Collapse
|
18
|
Nakamura M, Parkhurst SM. Calcium influx rapidly establishes distinct spatial recruitments of Annexins to cell wounds. Genetics 2024; 227:iyae101. [PMID: 38874345 PMCID: PMC11304956 DOI: 10.1093/genetics/iyae101] [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: 02/04/2024] [Revised: 02/04/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024] Open
Abstract
To survive daily damage, the formation of actomyosin ring at the wound edge is required to rapidly close cell wounds. Calcium influx is one of the start signals for these cell wound repair events. Here, we find that the rapid recruitment of all 3 Drosophila calcium-responding and phospholipid-binding Annexin proteins (AnxB9, AnxB10, and AnxB11) to distinct regions around the wound is regulated by the quantity of calcium influx rather than their binding to specific phospholipids. The distinct recruitment patterns of these Annexins regulate the subsequent recruitment of RhoGEF2 and RhoGEF3 through actin stabilization to form a robust actomyosin ring. Surprisingly, while the wound does not close in the absence of calcium influx, we find that reduced calcium influx can still initiate repair processes, albeit leading to severe repair phenotypes. Thus, our results suggest that, in addition to initiating repair events, the quantity of calcium influx is important for precise Annexin spatiotemporal protein recruitment to cell wounds and efficient wound repair.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Susan M Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
19
|
Subhan F, Zizzo MG, Serio R. Motor dysfunction of the gut in Duchenne muscular dystrophy: A review. Neurogastroenterol Motil 2024; 36:e14804. [PMID: 38651673 DOI: 10.1111/nmo.14804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Duchenne's muscular dystrophy (DMD) is a severe type of hereditary, neuromuscular disorder caused by a mutation in the dystrophin gene resulting in the absence or production of truncated dystrophin protein. Conventionally, clinical descriptions of the disorder focus principally on striated muscle defects; however, DMD manifestations involving gastrointestinal (GI) smooth muscle have been reported, even if not rigorously studied. PURPOSE The objective of the present review is to offer a comprehensive perspective on the existing knowledge concerning GI manifestations in DMD, focusing the attention on evidence in DMD patients and mdx mice. This includes an assessment of symptomatology, etiological pathways, and potential corrective approaches. This paper could provide helpful information about DMD gastrointestinal implications that could serve as a valuable orientation for prospective research endeavors in this field. This manuscript emphasizes the effectiveness of mdx mice, a DMD animal model, in unraveling mechanistic insights and exploring the pathological alterations in the GI tract. The gastrointestinal consequences evident in patients with DMD and the mdx mice models are a significant area of focus for researchers. The exploration of this area in depth could facilitate the development of more efficient therapeutic approaches and improve the well-being of individuals impacted by the condition.
Collapse
Affiliation(s)
- Fazal Subhan
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo, Italy
| | - Maria Grazia Zizzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo, Italy
- ATeN (Advanced Technologies Network) Center, Viale delle Scienze, University of Palermo, Palermo, Italy
| | - Rosa Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo, Italy
| |
Collapse
|
20
|
De Masi A, Zanou N, Strotjohann K, Lee D, Lima TI, Li X, Jeon J, Place N, Jung H, Auwerx J. Cyclo His-Pro Attenuates Muscle Degeneration in Murine Myopathy Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305927. [PMID: 38728626 PMCID: PMC11267275 DOI: 10.1002/advs.202305927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/11/2024] [Indexed: 05/12/2024]
Abstract
Among the inherited myopathies, a group of muscular disorders characterized by structural and metabolic impairments in skeletal muscle, Duchenne muscular dystrophy (DMD) stands out for its devastating progression. DMD pathogenesis is driven by the progressive degeneration of muscle fibers, resulting in inflammation and fibrosis that ultimately affect the overall muscle biomechanics. At the opposite end of the spectrum of muscle diseases, age-related sarcopenia is a common condition that affects an increasing proportion of the elderly. Although characterized by different pathological mechanisms, DMD and sarcopenia share the development of progressive muscle weakness and tissue inflammation. Here, the therapeutic effects of Cyclo Histidine-Proline (CHP) against DMD and sarcopenia are evaluated. In the mdx mouse model of DMD, it is shown that CHP restored muscle contractility and force production, accompanied by the reduction of fibrosis and inflammation in skeletal muscle. CHP furthermore prevented the development of cardiomyopathy and fibrosis in the diaphragm, the two leading causes of death for DMD patients. CHP also attenuated muscle atrophy and functional deterioration in a mouse model of age-related sarcopenia. These findings from two different models of muscle dysfunction hence warrant further investigation into the effects of CHP on muscle pathologies in animal models and eventually in patients.
Collapse
Affiliation(s)
- Alessia De Masi
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical SciencesFaculty of Biology‐MedicineUniversity of LausanneLausanne1015Switzerland
| | - Keno Strotjohann
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Dohyun Lee
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
| | - Tanes I. Lima
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Jongsu Jeon
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
| | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical SciencesFaculty of Biology‐MedicineUniversity of LausanneLausanne1015Switzerland
| | - Hoe‐Yune Jung
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
- School of Interdisciplinary Bioscience and BioengineeringPohang University of Science and Technology (POSTECH)Pohang37673South Korea
| | - Johan Auwerx
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| |
Collapse
|
21
|
Palestra F, Memoli G, Ventrici A, Trocchia M, Galdiero M, Varricchi G, Loffredo S. Ca 2+-Dependent Processes of Innate Immunity in IBD. Cells 2024; 13:1079. [PMID: 38994933 PMCID: PMC11240513 DOI: 10.3390/cells13131079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/15/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
IBD is an uncontrolled inflammatory condition of the gastrointestinal tract, which mainly manifests in two forms: ulcerative colitis (UC) and Crohn's disease (CD). The pathogenesis of IBD appears to be associated with an abnormal response of innate and adaptive immune cells. Innate immunity cells, such as macrophages, mast cells, and granulocytes, can produce proinflammatory (e.g., TNF-α) and oxidative stress (ROS) mediators promoting intestinal damage, and their abnormal responses can induce an imbalance in adaptive immunity, leading to the production of inflammatory cytokines that increase innate immune damage, abate intestinal barrier functions, and aggravate inflammation. Considering that Ca2+ signalling plays a key role in a plethora of cellular functions, this review has the purpose of deepening the potential Ca2+ involvement in IBD pathogenesis.
Collapse
Affiliation(s)
- Francesco Palestra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Gina Memoli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Annagioia Ventrici
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Marialuisa Trocchia
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Mariarosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| |
Collapse
|
22
|
Timpani CA, Debrincat D, Kourakis S, Boyer R, Formosa LE, Steele JR, Zhang H, Schittenhelm RB, Russell AP, Rybalka E, Lindsay A. Loss of endogenous estrogen alters mitochondrial metabolism and muscle clock-related protein Rbm20 in female mdx mice. FASEB J 2024; 38:e23718. [PMID: 38847487 DOI: 10.1096/fj.202400329r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 11/01/2024]
Abstract
Female carriers of a Duchenne muscular dystrophy (DMD) gene mutation manifest exercise intolerance and metabolic anomalies that may be exacerbated following menopause due to the loss of estrogen, a known regulator of skeletal muscle function and metabolism. Here, we studied the impact of estrogen depletion (via ovariectomy) on exercise tolerance and muscle mitochondrial metabolism in female mdx mice and the potential of estrogen replacement therapy (using estradiol) to protect against functional and metabolic perturbations. We also investigated the effect of estrogen depletion, and replacement, on the skeletal muscle proteome through an untargeted proteomic approach with TMT-labelling. Our study confirms that loss of estrogen in female mdx mice reduces exercise capacity, tricarboxylic acid cycle intermediates, and citrate synthase activity but that these deficits are offset through estrogen replacement therapy. Furthermore, ovariectomy downregulated protein expression of RNA-binding motif factor 20 (Rbm20), a critical regulator of sarcomeric and muscle homeostasis gene splicing, which impacted pathways involving ribosomal and mitochondrial translation. Estrogen replacement modulated Rbm20 protein expression and promoted metabolic processes and the upregulation of proteins involved in mitochondrial dynamics and metabolism. Our data suggest that estrogen mitigates dystrophinopathic features in female mdx mice and that estrogen replacement may be a potential therapy for post-menopausal DMD carriers.
Collapse
Affiliation(s)
- Cara A Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Didier Debrincat
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Stephanie Kourakis
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Rebecca Boyer
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Haijian Zhang
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
- Division of Neuropaediatrics and Developmental Medicine, University Children's Hospital of Basel (UKBB), Basel, Switzerland
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
23
|
Kodippili K, Hakim CH, Burke MJ, Yue Y, Teixeira JA, Zhang K, Yao G, Babu GJ, Herzog RW, Duan D. SERCA2a overexpression improves muscle function in a canine Duchenne muscular dystrophy model. Mol Ther Methods Clin Dev 2024; 32:101268. [PMID: 38911286 PMCID: PMC11190715 DOI: 10.1016/j.omtm.2024.101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/16/2024] [Indexed: 06/25/2024]
Abstract
Excessive cytosolic calcium accumulation contributes to muscle degeneration in Duchenne muscular dystrophy (DMD). Sarco/endoplasmic reticulum calcium ATPase (SERCA) is a sarcoplasmic reticulum (SR) calcium pump that actively transports calcium from the cytosol into the SR. We previously showed that adeno-associated virus (AAV)-mediated SERCA2a therapy reduced cytosolic calcium overload and improved muscle and heart function in the murine DMD model. Here, we tested whether AAV SERCA2a therapy could ameliorate muscle disease in the canine DMD model. 7.83 × 1013 vector genome particles of the AAV vector were injected into the extensor carpi ulnaris (ECU) muscles of four juvenile affected dogs. Contralateral ECU muscles received excipient. Three months later, we observed widespread transgene expression and significantly increased SERCA2a levels in the AAV-injected muscles. Treatment improved SR calcium uptake, significantly reduced calpain activity, significantly improved contractile kinetics, and significantly enhanced resistance to eccentric contraction-induced force loss. Nonetheless, muscle histology was not improved. To evaluate the safety of AAV SERCA2a therapy, we delivered the vector to the ECU muscle of adult normal dogs. We achieved strong transgene expression without altering muscle histology and function. Our results suggest that AAV SERCA2a therapy has the potential to improve muscle performance in a dystrophic large mammal.
Collapse
Affiliation(s)
- Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Chady H. Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Matthew J. Burke
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - James A. Teixeira
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Gang Yao
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212, USA
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212, USA
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
24
|
Muriel J, Lukyanenko V, Kwiatkowski TA, Li Y, Bhattacharya S, Banford KK, Garman D, Bulgart HR, Sutton RB, Weisleder N, Bloch RJ. Nanodysferlins support membrane repair and binding to TRIM72/MG53 but do not localize to t-tubules or stabilize Ca 2+ signaling. Mol Ther Methods Clin Dev 2024; 32:101257. [PMID: 38779337 PMCID: PMC11109471 DOI: 10.1016/j.omtm.2024.101257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Mutations in the DYSF gene, encoding the protein dysferlin, lead to several forms of muscular dystrophy. In healthy skeletal muscle, dysferlin concentrates in the transverse tubules and is involved in repairing the sarcolemma and stabilizing Ca2+ signaling after membrane disruption. The DYSF gene encodes 7-8 C2 domains, several Fer and Dysf domains, and a C-terminal transmembrane sequence. Because its coding sequence is too large to package in adeno-associated virus, the full-length sequence is not amenable to current gene delivery methods. Thus, we have examined smaller versions of dysferlin, termed "nanodysferlins," designed to eliminate several C2 domains, specifically C2 domains D, E, and F; B, D, and E; and B, D, E, and F. We also generated a variant by replacing eight amino acids in C2G in the nanodysferlin missing domains D through F. We electroporated dysferlin-null A/J mouse myofibers with Venus fusion constructs of these variants, or as untagged nanodysferlins together with GFP, to mark transfected fibers We found that, although these nanodysferlins failed to concentrate in transverse tubules, three of them supported membrane repair after laser wounding while all four bound the membrane repair protein, TRIM72/MG53, similar to WT dysferlin. By contrast, they failed to suppress Ca2+ waves after myofibers were injured by mild hypoosmotic shock. Our results suggest that the internal C2 domains of dysferlin are required for normal t-tubule localization and Ca2+ signaling and that membrane repair does not require these C2 domains.
Collapse
Affiliation(s)
- Joaquin Muriel
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Valeriy Lukyanenko
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas A. Kwiatkowski
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Yi Li
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sayak Bhattacharya
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Kassidy K. Banford
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Garman
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hannah R. Bulgart
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Roger B. Sutton
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Noah Weisleder
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Robert J. Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
25
|
Alnassar N, Hajto J, Rumney RMH, Verma S, Borczyk M, Saha C, Kanczler J, Butt AM, Occhipinti A, Pomeroy J, Angione C, Korostynski M, Górecki DC. Ablation of the dystrophin Dp71f alternative C-terminal variant increases sarcoma tumour cell aggressiveness. Hum Mol Genet 2024:ddae094. [PMID: 38850567 DOI: 10.1093/hmg/ddae094] [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: 12/29/2023] [Revised: 05/08/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024] Open
Abstract
Alterations in Dp71 expression, the most ubiquitous dystrophin isoform, have been associated with patient survival across tumours. Intriguingly, in certain malignancies, Dp71 acts as a tumour suppressor, while manifesting oncogenic properties in others. This diversity could be explained by the expression of two Dp71 splice variants encoding proteins with distinct C-termini, each with specific properties. Expression of these variants has impeded the exploration of their unique roles. Using CRISPR/Cas9, we ablated the Dp71f variant with the alternative C-terminus in a sarcoma cell line not expressing the canonical C-terminal variant, and conducted molecular (RNAseq) and functional characterisation of the knockout cells. Dp71f ablation induced major transcriptomic alterations, particularly affecting the expression of genes involved in calcium signalling and ECM-receptor interaction pathways. The genome-scale metabolic analysis identified significant downregulation of glucose transport via membrane vesicle reaction (GLCter) and downregulated glycolysis/gluconeogenesis pathway. Functionally, these molecular changes corresponded with, increased calcium responses, cell adhesion, proliferation, survival under serum starvation and chemotherapeutic resistance. Knockout cells showed reduced GLUT1 protein expression, survival without attachment and their migration and invasion in vitro and in vivo were unaltered, despite increased matrix metalloproteinases release. Our findings emphasise the importance of alternative splicing of dystrophin transcripts and underscore the role of the Dp71f variant, which appears to govern distinct cellular processes frequently dysregulated in tumour cells. The loss of this regulatory mechanism promotes sarcoma cell survival and treatment resistance. Thus, Dp71f is a target for future investigations exploring the intricate functions of specific DMD transcripts in physiology and across malignancies.
Collapse
Affiliation(s)
- Nancy Alnassar
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Jacek Hajto
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PAS, Smetna 12, Krakow 31155, Poland
| | - Robin M H Rumney
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, Tees Valley TS1 3BX, United Kingdom
| | - Malgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PAS, Smetna 12, Krakow 31155, Poland
| | - Chandrika Saha
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Janos Kanczler
- Bone & Joint Research Group, Department of Human Development and Health, University of Southampton, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Arthur M Butt
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, Tees Valley TS1 3BX, United Kingdom
| | - Joanna Pomeroy
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, Tees Valley TS1 3BX, United Kingdom
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PAS, Smetna 12, Krakow 31155, Poland
| | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| |
Collapse
|
26
|
Wu K, Shieh JS, Qin L, Guo JJ. Mitochondrial mechanisms in the pathogenesis of chronic inflammatory musculoskeletal disorders. Cell Biosci 2024; 14:76. [PMID: 38849951 PMCID: PMC11162051 DOI: 10.1186/s13578-024-01259-9] [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: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Chronic inflammatory musculoskeletal disorders characterized by prolonged muscle inflammation, resulting in enduring pain and diminished functionality, pose significant challenges for the patients. Emerging scientific evidence points to mitochondrial malfunction as a pivotal factor contributing to these ailments. Mitochondria play a critical role in powering skeletal muscle activity, but in the context of persistent inflammation, disruptions in their quantity, configuration, and performance have been well-documented. Various disturbances, encompassing alterations in mitochondrial dynamics (such as fission and fusion), calcium regulation, oxidative stress, biogenesis, and the process of mitophagy, are believed to play a central role in the progression of these disorders. Additionally, unfolded protein responses and the accumulation of fatty acids within muscle cells may adversely affect the internal milieu, impairing the equilibrium of mitochondrial functioning. The structural discrepancies between different mitochondrial subsets namely, intramyofibrillar and subsarcolemmal mitochondria likely impact their metabolic capabilities and susceptibility to inflammatory influences. The release of signals from damaged mitochondria is known to incite inflammatory responses. Intriguingly, migrasomes and extracellular vesicles serve as vehicles for intercellular transfer of mitochondria, aiding in the removal of impaired mitochondria and regulation of inflammation. Viral infections have been implicated in inducing stress on mitochondria. Prolonged dysfunction of these vital organelles sustains oxidative harm, metabolic irregularities, and heightened cytokine release, impeding the body's ability to repair tissues. This review provides a comprehensive analysis of advancements in understanding changes in the intracellular environment, mitochondrial architecture and distribution, biogenesis, dynamics, autophagy, oxidative stress, cytokines associated with mitochondria, vesicular structures, and associated membranes in the context of chronic inflammatory musculoskeletal disorders. Strategies targeting key elements regulating mitochondrial quality exhibit promise in the restoration of mitochondrial function, alleviation of inflammation, and enhancement of overall outcomes.
Collapse
Affiliation(s)
- Kailun Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of Soochow University/Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People's Republic of China
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Ju-Sheng Shieh
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 11490, Taiwan
| | - Ling Qin
- Musculoskeletal Research Laboratory of the Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Jiong Jiong Guo
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.
- MOE China-Europe Sports Medicine Belt and Road Joint Laboratory, Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Escobar-Huertas JF, Vaca-González JJ, Guevara JM, Ramirez-Martinez AM, Trabelsi O, Garzón-Alvarado DA. Duchenne and Becker muscular dystrophy: Cellular mechanisms, image analysis, and computational models: A review. Cytoskeleton (Hoboken) 2024; 81:269-286. [PMID: 38224155 DOI: 10.1002/cm.21826] [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: 05/24/2023] [Revised: 11/21/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
The muscle is the principal tissue that is capable to transform potential energy into kinetic energy. This process is due to the transformation of chemical energy into mechanical energy to enhance the movements and all the daily activities. However, muscular tissues can be affected by some pathologies associated with genetic alterations that affect the expression of proteins. As the muscle is a highly organized structure in which most of the signaling pathways and proteins are related to one another, pathologies may overlap. Duchenne muscular dystrophy (DMD) is one of the most severe muscle pathologies triggering degeneration and muscle necrosis. Several mathematical models have been developed to predict muscle response to different scenarios and pathologies. The aim of this review is to describe DMD and Becker muscular dystrophy in terms of cellular behavior and molecular disorders and to present an overview of the computational models implemented to understand muscle behavior with the aim of improving regenerative therapy.
Collapse
Affiliation(s)
- J F Escobar-Huertas
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Juan Jairo Vaca-González
- Escuela de pregrado, Dirección Académica, Vicerrectoría de Sede, Universidad Nacional de Colombia, Sede la Paz, Cesar, Colombia
| | - Johana María Guevara
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Olfa Trabelsi
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - D A Garzón-Alvarado
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
28
|
Mostosi D, Molinaro M, Saccone S, Torrente Y, Villa C, Farini A. Exploring the Gut Microbiota-Muscle Axis in Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:5589. [PMID: 38891777 PMCID: PMC11171690 DOI: 10.3390/ijms25115589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
The gut microbiota plays a pivotal role in maintaining the dynamic balance of intestinal epithelial and immune cells, crucial for overall organ homeostasis. Dysfunctions in these intricate relationships can lead to inflammation and contribute to the pathogenesis of various diseases. Recent findings uncovered the existence of a gut-muscle axis, revealing how alterations in the gut microbiota can disrupt regulatory mechanisms in muscular and adipose tissues, triggering immune-mediated inflammation. In the context of Duchenne muscular dystrophy (DMD), alterations in intestinal permeability stand as a potential origin of molecules that could trigger muscle degeneration via various pathways. Metabolites produced by gut bacteria, or fragments of bacteria themselves, may have the ability to migrate from the gut into the bloodstream and ultimately infiltrate distant muscle tissues, exacerbating localized pathologies. These insights highlight alternative pathological pathways in DMD beyond the musculoskeletal system, paving the way for nutraceutical supplementation as a potential adjuvant therapy. Understanding the complex interplay between the gut microbiota, immune system, and muscular health offers new perspectives for therapeutic interventions beyond conventional approaches to efficiently counteract the multifaceted nature of DMD.
Collapse
Affiliation(s)
- Debora Mostosi
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (D.M.); (Y.T.); (C.V.)
| | - Monica Molinaro
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.M.); (S.S.)
| | - Sabrina Saccone
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.M.); (S.S.)
| | - Yvan Torrente
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (D.M.); (Y.T.); (C.V.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.M.); (S.S.)
| | - Chiara Villa
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy; (D.M.); (Y.T.); (C.V.)
| | - Andrea Farini
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.M.); (S.S.)
| |
Collapse
|
29
|
Mareedu S, Fefelova N, Galindo CL, Prakash G, Mukai R, Sadoshima J, Xie LH, Babu GJ. Improved mitochondrial function in the hearts of sarcolipin-deficient dystrophin and utrophin double-knockout mice. JCI Insight 2024; 9:e170185. [PMID: 38564291 PMCID: PMC11141945 DOI: 10.1172/jci.insight.170185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease associated with cardiomyopathy. DMD cardiomyopathy is characterized by abnormal intracellular Ca2+ homeostasis and mitochondrial dysfunction. We used dystrophin and utrophin double-knockout (mdx:utrn-/-) mice in a sarcolipin (SLN) heterozygous-knockout (sln+/-) background to examine the effect of SLN reduction on mitochondrial function in the dystrophic myocardium. Germline reduction of SLN expression in mdx:utrn-/- mice improved cardiac sarco/endoplasmic reticulum (SR) Ca2+ cycling, reduced cardiac fibrosis, and improved cardiac function. At the cellular level, reducing SLN expression prevented mitochondrial Ca2+ overload, reduced mitochondrial membrane potential loss, and improved mitochondrial function. Transmission electron microscopy of myocardial tissues and proteomic analysis of mitochondria-associated membranes showed that reducing SLN expression improved mitochondrial structure and SR-mitochondria interactions in dystrophic cardiomyocytes. These findings indicate that SLN upregulation plays a substantial role in the pathogenesis of cardiomyopathy and that reducing SLN expression has clinical implications in the treatment of DMD cardiomyopathy.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Calcium/metabolism
- Cardiomyopathies/metabolism
- Cardiomyopathies/genetics
- Cardiomyopathies/pathology
- Disease Models, Animal
- Dystrophin/genetics
- Dystrophin/metabolism
- Mice, Inbred mdx
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/ultrastructure
- Mitochondria, Heart/genetics
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proteolipids/metabolism
- Proteolipids/genetics
- Utrophin/genetics
- Utrophin/metabolism
Collapse
Affiliation(s)
- Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Cristi L. Galindo
- Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Goutham Prakash
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
30
|
Robertson R, Li S, Filippelli RL, Chang NC. Muscle stem cell dysfunction in rhabdomyosarcoma and muscular dystrophy. Curr Top Dev Biol 2024; 158:83-121. [PMID: 38670717 DOI: 10.1016/bs.ctdb.2024.01.019] [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: 04/28/2024]
Abstract
Muscle stem cells (MuSCs) are crucial to the repair and homeostasis of mature skeletal muscle. MuSC dysfunction and dysregulation of the myogenic program can contribute to the development of pathology ranging from cancers like rhabdomyosarcoma (RMS) or muscle degenerative diseases such as Duchenne muscular dystrophy (DMD). Both diseases exhibit dysregulation at nearly all steps of myogenesis. For instance, MuSC self-renewal processes are altered. In RMS, this leads to the creation of tumor propagating cells. In DMD, impaired asymmetric stem cell division creates a bias towards producing self-renewing stem cells instead of committing to differentiation. Hyperproliferation of these cells contribute to tumorigenesis in RMS and symmetric expansion of the self-renewing MuSC population in DMD. Both diseases also exhibit a repression of factors involved in terminal differentiation, halting RMS cells in the proliferative stage and thus driving tumor growth. Conversely, the MuSCs in DMD exhibit impaired differentiation and fuse prematurely, affecting myonuclei maturation and the integrity of the dystrophic muscle fiber. Finally, both disease states cause alterations to the MuSC niche. Various elements of the niche such as inflammatory and migratory signaling that impact MuSC behavior are dysregulated. Here we show how these seemingly distantly related diseases indeed have similarities in MuSC dysfunction, underlying the importance of considering MuSCs when studying the pathophysiology of muscle diseases.
Collapse
Affiliation(s)
- Rebecca Robertson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Shulei Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Romina L Filippelli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Natasha C Chang
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada.
| |
Collapse
|
31
|
Nakamura M, Parkhurst SM. Calcium influx rapidly establishes distinct spatial recruitments of Annexins to cell wounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569799. [PMID: 38105960 PMCID: PMC10723296 DOI: 10.1101/2023.12.03.569799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
To survive daily damage, the formation of actomyosin ring at the wound periphery is required to rapidly close cell wounds. Calcium influx is one of the start signals for these cell wound repair events. Here, we find that rapid recruitment of all three Drosophila calcium responding and phospholipid binding Annexin proteins (AnxB9, AnxB10, AnxB11) to distinct regions around the wound are regulated by the quantity of calcium influx rather than their binding to specific phospholipids. The distinct recruitment patterns of these Annexins regulate the subsequent recruitment of RhoGEF2 and RhoGEF3 through actin stabilization to form a robust actomyosin ring. Surprisingly, we find that reduced extracellular calcium and depletion of intracellular calcium affect cell wound repair differently, despite these two conditions exhibiting similar GCaMP signals. Thus, our results suggest that, in addition to initiating repair events, both the quantity and sources of calcium influx are important for precise Annexin spatiotemporal protein recruitment to cell wounds and efficient wound repair.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| |
Collapse
|
32
|
da Silva HNM, Mizobuti DS, Pereira VA, da Rocha GL, da Cruz MV, de Oliveira AG, Silveira LR, Minatel E. LED therapy plus idebenone treatment targeting calcium and mitochondrial signaling pathways in dystrophic muscle cells. Cell Stress Chaperones 2023; 28:773-785. [PMID: 37578579 PMCID: PMC10746663 DOI: 10.1007/s12192-023-01369-2] [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: 05/17/2023] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023] Open
Abstract
Intracellular calcium dysregulation, oxidative stress, and mitochondrial dysfunction are some of the main pathway contributors towards disease progression in Duchenne muscular dystrophy (DMD). This study is aimed at investigating the effects of light emitting diode therapy (LEDT) and idebenone antioxidant treatment, applied alone or together in dystrophic primary muscle cells from mdx mice, the experimental model of DMD. Mdx primary muscle cells were submitted to LEDT and idebenone treatment and evaluated for cytotoxic effects and calcium and mitochondrial signaling pathways. LEDT and idebenone treatment showed no cytotoxic effects on the dystrophic muscle cells. Regarding the calcium pathways, after LEDT and idebenone treatment, a significant reduction in intracellular calcium content, calpain-1, calsequestrin, and sarcolipin levels, was observed. In addition, a significant reduction in oxidative stress level markers, such as H2O2, and 4-HNE levels, was observed. Regarding mitochondrial signaling pathways, a significant increase in oxidative capacity (by OCR and OXPHOS levels) was observed. In addition, the PGC-1α, SIRT-1, and PPARδ levels were significantly higher in the LEDT plus idebenone treated-dystrophic muscle cells. Together, the findings suggest that LEDT and idebenone treatment, alone or in conjunction, can modulate the calcium and mitochondrial signaling pathways, such as SLN, SERCA 1, and PGC-1α, contributing towards the improvement of the dystrophic phenotype in mdx muscle cells. In addition, data from the LEDT plus idebenone treatment showed slightly better results than those of each separate treatment in terms of SLN, OXPHOS, and SIRT-1.
Collapse
Affiliation(s)
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Valéria Andrade Pereira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Marcos Vinícius da Cruz
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), Campinas, Brazil
| | - André Gustavo de Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), Campinas, Brazil
| | - Leonardo Reis Silveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), Campinas, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil.
| |
Collapse
|
33
|
Haffner V, Nourian Z, Boerman EM, Lambert MD, Hanft LM, Krenz M, Baines CP, Duan D, McDonald KS, Domeier TL. Calcium handling dysfunction and cardiac damage following acute ventricular preload challenge in the dystrophin-deficient mouse heart. Am J Physiol Heart Circ Physiol 2023; 325:H1168-H1177. [PMID: 37737731 PMCID: PMC10907071 DOI: 10.1152/ajpheart.00265.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy and is caused by mutations in the dystrophin gene. Dystrophin deficiency is associated with structural and functional changes of the muscle cell sarcolemma and/or stretch-induced ion channel activation. In this investigation, we use mice with transgenic cardiomyocyte-specific expression of the GCaMP6f Ca2+ indicator to test the hypothesis that dystrophin deficiency leads to cardiomyocyte Ca2+ handling abnormalities following preload challenge. α-MHC-MerCreMer-GCaMP6f transgenic mice were developed on both a wild-type (WT) or dystrophic (Dmdmdx-4Cv) background. Isolated hearts of 3-7-mo male mice were perfused in unloaded Langendorff mode (0 mmHg) and working heart mode (preload = 20 mmHg). Following a 30-min preload challenge, hearts were perfused in unloaded Langendorff mode with 40 μM blebbistatin, and GCaMP6f was imaged using confocal fluorescence microscopy. Incidence of premature ventricular complexes (PVCs) was monitored before and following preload elevation at 20 mmHg. Hearts of both wild-type and dystrophic mice exhibited similar left ventricular contractile function. Following preload challenge, dystrophic hearts exhibited a reduction in GCaMP6f-positive cardiomyocytes and an increase in number of cardiomyocytes exhibiting Ca2+ waves/overload. Incidence of cardiac arrhythmias was low in both wild-type and dystrophic hearts during unloaded Langendorff mode. However, after preload elevation to 20-mmHg hearts of dystrophic mice exhibited an increased incidence of PVCs compared with hearts of wild-type mice. In conclusion, these data indicate susceptibility to preload-induced Ca2+ overload, ventricular damage, and ventricular dysfunction in male Dmdmdx-4Cv hearts. Our data support the hypothesis that cardiomyocyte Ca2+ overload underlies cardiac dysfunction in muscular dystrophy.NEW & NOTEWORTHY The mechanisms of cardiac disease progression in muscular dystrophy are complex and poorly understood. Using a transgenic mouse model with cardiomyocyte-specific expression of the GCaMP6f Ca2+ indicator, the present study provides further support for the Ca2+-overload hypothesis of disease progression and ventricular arrhythmogenesis in muscular dystrophy.
Collapse
Affiliation(s)
- Vivian Haffner
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Zahra Nourian
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Michelle D Lambert
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Christopher P Baines
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, School of Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
34
|
Rawls A, Diviak BK, Smith CI, Severson GW, Acosta SA, Wilson-Rawls J. Pharmacotherapeutic Approaches to Treatment of Muscular Dystrophies. Biomolecules 2023; 13:1536. [PMID: 37892218 PMCID: PMC10605463 DOI: 10.3390/biom13101536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic muscle-wasting disorders that are subdivided based on the region of the body impacted by muscle weakness as well as the functional activity of the underlying genetic mutations. A common feature of the pathophysiology of muscular dystrophies is chronic inflammation associated with the replacement of muscle mass with fibrotic scarring. With the progression of these disorders, many patients suffer cardiomyopathies with fibrosis of the cardiac tissue. Anti-inflammatory glucocorticoids represent the standard of care for Duchenne muscular dystrophy, the most common muscular dystrophy worldwide; however, long-term exposure to glucocorticoids results in highly adverse side effects, limiting their use. Thus, it is important to develop new pharmacotherapeutic approaches to limit inflammation and fibrosis to reduce muscle damage and promote repair. Here, we examine the pathophysiology, genetic background, and emerging therapeutic strategies for muscular dystrophies.
Collapse
Affiliation(s)
- Alan Rawls
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
| | - Bridget K. Diviak
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Cameron I. Smith
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Grant W. Severson
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Sofia A. Acosta
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
| |
Collapse
|
35
|
Henzi BC, Schmidt S, Nagy S, Rubino-Nacht D, Schaedelin S, Putananickal N, Stimpson G, Amthor H, Childs AM, Deconinck N, de Groot I, Horrocks I, Houwen-van Opstal S, Laugel V, Lopez Lobato M, Madruga Garrido M, Nascimento Osorio A, Schara-Schmidt U, Spinty S, von Moers A, Lawrence F, Hafner P, Dorchies OM, Fischer D. Safety and efficacy of tamoxifen in boys with Duchenne muscular dystrophy (TAMDMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol 2023; 22:890-899. [PMID: 37739572 DOI: 10.1016/s1474-4422(23)00285-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/28/2023] [Accepted: 07/20/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Drug repurposing could provide novel treatment options for Duchenne muscular dystrophy. Because tamoxifen-an oestrogen receptor regulator-reduced signs of muscular pathology in a Duchenne muscular dystrophy mouse model, we aimed to assess the safety and efficacy of tamoxifen in humans as an adjunct to corticosteroid therapy over a period of 48 weeks. METHODS We did a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial at 12 study centres in seven European countries. We enrolled ambulant boys aged 6·5-12·0 years with a genetically confirmed diagnosis of Duchenne muscular dystrophy and who were on stable corticosteroid treatment for more than 6 months. Exclusion criteria included ophthalmological disorders, including cataracts, and haematological disorders. We randomly assigned (1:1) participants using an online randomisation tool to either 20 mg tamoxifen orally per day or matched placebo, stratified by centre and corticosteroid intake. Participants, caregivers, and clinical investigators were masked to treatment assignments. Tamoxifen was taken in addition to standard care with corticosteroids, and participants attended study visits for examinations every 12 weeks. The primary efficacy outcome was the change from baseline to week 48 in scores on the D1 domain of the Motor Function Measure in the intention-to-treat population (defined as all patients who fulfilled the inclusion criteria and began treatment). This study is registered with ClinicalTrials.gov (NCT03354039) and is completed. FINDINGS Between May 24, 2018, and Oct 14, 2020, 95 boys were screened for inclusion, and 82 met inclusion criteria and were initially enrolled into the study. Three boys were excluded after initial screening due to cataract diagnosis or revoked consent directly after screening, but before randomisation. A further boy assigned to the placebo group did not begin treatment. Therefore, 40 individuals assigned tamoxifen and 38 allocated placebo were included in the intention-to-treat population. The primary efficacy outcome did not differ significantly between tamoxifen (-3·05%, 95% CI -7·02 to 0·91) and placebo (-6·15%, -9·19 to -3·11; 2·90% difference, -3·02 to 8·82, p=0·33). Severe adverse events occurred in two participants: one participant who received tamoxifen had a fall, and one who received placebo suffered a panic attack. No deaths or life-threatening serious adverse events occurred. Viral infections were the most common adverse events. INTERPRETATION Tamoxifen was safe and well tolerated, but no difference between groups was reported for the primary efficacy endpoint. Slower disease progression, defined by loss of motor function over time, was indicated in the tamoxifen group compared with the placebo group, but differences in outcome measures were neither clinically nor statistically significant. Currently, we cannot recommend the use of tamoxifen in daily clinical practice as a treatment option for boys with Duchenne muscular dystrophy due to insufficient clinical evidence. FUNDING Thomi Hopf Foundation, ERA-Net, Swiss National Science Foundation, Duchenne UK, Joining Jack, Duchenne Parent Project, Duchenne Parent Project Spain, Fondation Suisse de Recherche sur les Maladies Musculaires, Association Monegasque contre les Myopathies.
Collapse
Affiliation(s)
- Bettina C Henzi
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland; Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Simone Schmidt
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Sara Nagy
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Rubino-Nacht
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Schaedelin
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Niveditha Putananickal
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Georgia Stimpson
- Developmental Neuroscience Research and Teaching Department, Faculty of Population Health Sciences, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Helge Amthor
- Service de Neurologie et Réanimation Pédiatriques, APHP Paris Saclay, Hôpital Raymond Poincaré, Garches, France
| | | | - Nicolas Deconinck
- Department of Paediatric Neurology and Neuromuscular Reference Center, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Imelda de Groot
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Saskia Houwen-van Opstal
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Vincent Laugel
- Department of Pediatric Neurology, Strasbourg University Hospital, Strasbourg, France
| | - Mercedes Lopez Lobato
- Sección de Neurología Pediátrica, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Marcos Madruga Garrido
- Sección de Neurología Pediátrica, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Andrés Nascimento Osorio
- Neuromuscular Unit, Department of Neurology, Hospital Sant Joan de Déu and Center for Biomedical Research Network on Rare Diseases, ISCIII, Barcelona, Spain
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Arpad von Moers
- Department of Pediatrics, DRK Kliniken Berlin Westend, Berlin, Germany
| | | | - Patricia Hafner
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Olivier M Dorchies
- School of Pharmaceutical Sciences and Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Dirk Fischer
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
36
|
Sharlo KA, Lvova ID, Tyganov SA, Zaripova KA, Belova SP, Kostrominova TY, Shenkman BS, Nemirovskaya TL. The Effect of SERCA Activation on Functional Characteristics and Signaling of Rat Soleus Muscle upon 7 Days of Unloading. Biomolecules 2023; 13:1354. [PMID: 37759754 PMCID: PMC10526198 DOI: 10.3390/biom13091354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Skeletal muscle abnormalities and atrophy during unloading are accompanied by the accumulation of excess calcium in the sarcoplasm. We hypothesized that calcium accumulation may occur, among other mechanisms, due to the inhibition of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. Consequently, the use of the SERCA activator will reduce the level of calcium in the sarcoplasm and prevent the negative consequences of muscle unloading. Wistar rats were randomly assigned into one of three groups (eight rats per group): control rats with placebo (C), 7 days of unloading/hindlimb suspension with placebo (7HS), and 7 days of unloading treated with SERCA activator CDN1163 (7HSC). After seven days of unloading the soleus muscle, the 7HS group displayed increased fatigue in the ex vivo test, a significant increase in the level of calcium-dependent CaMK II phosphorylation and the level of tropomyosin oxidation, as well as a decrease in the content of mitochondrial DNA and protein, slow-type myosin mRNA, and the percentage of slow-type muscle fibers. All of these changes were prevented in the 7HSC group. Moreover, treatment with CDN1163 blocked a decrease in the phosphorylation of p70S6k, an increase in eEF2 phosphorylation, and an increase in MuRF-1 mRNA expression. Nevertheless, there were no differences in the degree of fast and slow muscle fiber atrophy between the 7HS and 7HSC groups. Conclusion: SERCA activation during 7 days of unloading prevented an increase in soleus fatigue, the decrease of slow-type myosin, mitochondrial markers, and markers of calcium homeostasis but had no effect on muscle atrophy.
Collapse
Affiliation(s)
- Kristina A. Sharlo
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Irina D. Lvova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Sergey A. Tyganov
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Ksenia A. Zaripova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Svetlana P. Belova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana Y. Kostrominova
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine-Northwest, Gary, IN 46202, USA;
| | - Boris S. Shenkman
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana L. Nemirovskaya
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| |
Collapse
|
37
|
Rahman NIA, Lam CL, Sulaiman N, Abdullah NAH, Nordin F, Ariffin SHZ, Yazid MD. PAX7, a Key for Myogenesis Modulation in Muscular Dystrophies through Multiple Signaling Pathways: A Systematic Review. Int J Mol Sci 2023; 24:13051. [PMID: 37685856 PMCID: PMC10487808 DOI: 10.3390/ijms241713051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
Muscular dystrophy is a heterogenous group of hereditary muscle disorders caused by mutations in the genes responsible for muscle development, and is generally defined by a disastrous progression of muscle wasting and massive loss in muscle regeneration. Pax7 is closely associated with myogenesis, which is governed by various signaling pathways throughout a lifetime and is frequently used as an indicator in muscle research. In this review, an extensive literature search adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines was performed to identify research that examined signaling pathways in living models, while quantifying Pax7 expression in myogenesis. A total of 247 articles were retrieved from the Web of Science (WoS), PubMed and Scopus databases and were thoroughly examined and evaluated, resulting in 19 articles which met the inclusion criteria. Admittedly, we were only able to discuss the quantification of Pax7 carried out in research affecting various type of genes and signaling pathways, rather than the expression of Pax7 itself, due to the massive differences in approach, factor molecules and signaling pathways analyzed across the research. However, we highlighted the thorough evidence for the alteration of the muscle stem cell precursor Pax7 in multiple signaling pathways described in different living models, with an emphasis on the novel approach that could be taken in manipulating Pax7 expression itself in dystrophic muscle, towards the discovery of an effective treatment for muscular dystrophy. Therefore, we believe that this could be applied to the potential gap in muscle research that could be filled by tuning the well-established marker expression to improve dystrophic muscle.
Collapse
Affiliation(s)
- Nor Idayu A. Rahman
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Chung Liang Lam
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Nur Atiqah Haizum Abdullah
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Fazlina Nordin
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Shahrul Hisham Zainal Ariffin
- Centre of Biotechnology & Functional Food, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| |
Collapse
|
38
|
Dalla Barba F, Soardi M, Mouhib L, Risato G, Akyürek EE, Lucon-Xiccato T, Scano M, Benetollo A, Sacchetto R, Richard I, Argenton F, Bertolucci C, Carotti M, Sandonà D. Modeling Sarcoglycanopathy in Danio rerio. Int J Mol Sci 2023; 24:12707. [PMID: 37628888 PMCID: PMC10454440 DOI: 10.3390/ijms241612707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sarcoglycanopathies, also known as limb girdle muscular dystrophy 3-6, are rare muscular dystrophies characterized, although heterogeneous, by high disability, with patients often wheelchair-bound by late adolescence and frequently developing respiratory and cardiac problems. These diseases are currently incurable, emphasizing the importance of effective treatment strategies and the necessity of animal models for drug screening and therapeutic verification. Using the CRISPR/Cas9 genome editing technique, we generated and characterized δ-sarcoglycan and β-sarcoglycan knockout zebrafish lines, which presented a progressive disease phenotype that worsened from a mild larval stage to distinct myopathic features in adulthood. By subjecting the knockout larvae to a viscous swimming medium, we were able to anticipate disease onset. The δ-SG knockout line was further exploited to demonstrate that a δ-SG missense mutant is a substrate for endoplasmic reticulum-associated degradation (ERAD), indicating premature degradation due to protein folding defects. In conclusion, our study underscores the utility of zebrafish in modeling sarcoglycanopathies through either gene knockout or future knock-in techniques. These novel zebrafish lines will not only enhance our understanding of the disease's pathogenic mechanisms, but will also serve as powerful tools for phenotype-based drug screening, ultimately contributing to the development of a cure for sarcoglycanopathies.
Collapse
Affiliation(s)
- Francesco Dalla Barba
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Michela Soardi
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Leila Mouhib
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
- Randall Center for Cell and Molecular Biophysics, King’s College London, London WC2R 2LS, UK
| | - Giovanni Risato
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Via Giustiniani, 2, 35128 Padova, Italy
| | - Eylem Emek Akyürek
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, Legnaro, 35020 Padova, Italy
| | - Tyrone Lucon-Xiccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Martina Scano
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Alberto Benetollo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, Legnaro, 35020 Padova, Italy
| | - Isabelle Richard
- Genethon, F-91002 Evry, France
- INSERM, U951, INTEGRARE Research Unit, F-91002 Evry, France
| | - Francesco Argenton
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy
| | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Marcello Carotti
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| | - Dorianna Sandonà
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35131 Padova, Italy; (F.D.B.)
| |
Collapse
|
39
|
Shah MNA, Yokota T. Cardiac therapies for Duchenne muscular dystrophy. Ther Adv Neurol Disord 2023; 16:17562864231182934. [PMID: 37425427 PMCID: PMC10328182 DOI: 10.1177/17562864231182934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/02/2023] [Indexed: 07/11/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease that results in life-limiting complications such as loss of skeletal muscle function as well as respiratory and cardiac complications. Advanced therapeutics in pulmonary care have significantly reduced respiratory complication-related mortality, making cardiomyopathy the main determinant factor of survival. While there are multiple therapies such as the use of anti-inflammatory drugs, physical therapy, and ventilatory assistance targeted toward delaying the disease progression in DMD, a cure remains elusive. In the last decade, several therapeutic approaches have been developed to improve patient survival. These include small molecule-based therapy, micro-dystrophin gene delivery, CRISPR-mediated gene editing, nonsense readthrough, exon skipping, and cardiosphere-derived cell therapy. Associated with the specific benefits of each of these approaches are their individual risks and limitations. The variability in the genetic aberrations leading to DMD also limits the widespread use of these therapies. While numerous approaches have been explored to treat DMD pathophysiology, only a handful have successfully advanced through the preclinical stages. In this review, we summarize the currently approved as well as the most promising therapeutics undergoing clinical trials aimed toward treating DMD with a focus on its cardiac manifestations.
Collapse
Affiliation(s)
- Md Nur Ahad Shah
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
40
|
Bez Batti Angulski A, Hosny N, Cohen H, Martin AA, Hahn D, Bauer J, Metzger JM. Duchenne muscular dystrophy: disease mechanism and therapeutic strategies. Front Physiol 2023; 14:1183101. [PMID: 37435300 PMCID: PMC10330733 DOI: 10.3389/fphys.2023.1183101] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/13/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and ultimately fatal disease of skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. The identification of the dystrophin gene as central to DMD pathogenesis has led to the understanding of the muscle membrane and the proteins involved in membrane stability as the focal point of the disease. The lessons learned from decades of research in human genetics, biochemistry, and physiology have culminated in establishing the myriad functionalities of dystrophin in striated muscle biology. Here, we review the pathophysiological basis of DMD and discuss recent progress toward the development of therapeutic strategies for DMD that are currently close to or are in human clinical trials. The first section of the review focuses on DMD and the mechanisms contributing to membrane instability, inflammation, and fibrosis. The second section discusses therapeutic strategies currently used to treat DMD. This includes a focus on outlining the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, repair, and/or a range of dystrophin-independent approaches. The final section highlights the different therapeutic strategies for DMD currently in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
41
|
Marcadet L, Juracic ES, Khan N, Bouredji Z, Yagita H, Ward LM, Tupling AR, Argaw A, Frenette J. RANKL Inhibition Reduces Cardiac Hypertrophy in mdx Mice and Possibly in Children with Duchenne Muscular Dystrophy. Cells 2023; 12:1538. [PMID: 37296659 PMCID: PMC10253225 DOI: 10.3390/cells12111538] [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: 05/02/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Cardiomyopathy has become one of the leading causes of death in patients with Duchenne muscular dystrophy (DMD). We recently reported that the inhibition of the interaction between the receptor activator of nuclear factor κB ligand (RANKL) and receptor activator of nuclear factor κB (RANK) significantly improves muscle and bone functions in dystrophin-deficient mdx mice. RANKL and RANK are also expressed in cardiac muscle. Here, we investigate whether anti-RANKL treatment prevents cardiac hypertrophy and dysfunction in dystrophic mdx mice. Anti-RANKL treatment significantly reduced LV hypertrophy and heart mass, and maintained cardiac function in mdx mice. Anti-RANKL treatment also inhibited NFκB and PI3K, two mediators implicated in cardiac hypertrophy. Furthermore, anti-RANKL treatment increased SERCA activity and the expression of RyR, FKBP12, and SERCA2a, leading possibly to an improved Ca2+ homeostasis in dystrophic hearts. Interestingly, preliminary post hoc analyses suggest that denosumab, a human anti-RANKL, reduced left ventricular hypertrophy in two patients with DMD. Taken together, our results indicate that anti-RANKL treatment prevents the worsening of cardiac hypertrophy in mdx mice and could potentially maintain cardiac function in teenage or adult patients with DMD.
Collapse
Affiliation(s)
- Laetitia Marcadet
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
| | - Emma Sara Juracic
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (E.S.J.); (A.R.T.)
| | - Nasrin Khan
- The Ottawa Pediatric Bone Health Research Group, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; (N.K.); (L.M.W.)
| | - Zineb Bouredji
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
| | - Hideo Yagita
- Department of Immunology, School of Medicine, Juntendo University, Tokyo 113-8421, Japan;
| | - Leanne M. Ward
- The Ottawa Pediatric Bone Health Research Group, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; (N.K.); (L.M.W.)
- The Department of Pediatrics, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - A. Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON N2L 3G1, Canada; (E.S.J.); (A.R.T.)
| | - Anteneh Argaw
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada; (L.M.); (Z.B.); (A.A.)
- Department of Rehabilitation, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
42
|
Hahn D, Quick JD, Thompson BR, Crabtree A, Hackel BJ, Bates FS, Metzger JM. Rapid restitution of contractile dysfunction by synthetic copolymers in dystrophin-deficient single live skeletal muscle fibers. Skelet Muscle 2023; 13:9. [PMID: 37208786 PMCID: PMC10197332 DOI: 10.1186/s13395-023-00318-y] [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: 12/08/2022] [Accepted: 05/05/2023] [Indexed: 05/21/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by the lack of dystrophin, a cytoskeletal protein essential for the preservation of the structural integrity of the muscle cell membrane. DMD patients develop severe skeletal muscle weakness, degeneration, and early death. We tested here amphiphilic synthetic membrane stabilizers in mdx skeletal muscle fibers (flexor digitorum brevis; FDB) to determine their effectiveness in restoring contractile function in dystrophin-deficient live skeletal muscle fibers. After isolating FDB fibers via enzymatic digestion and trituration from thirty-three adult male mice (9 C57BL10, 24 mdx), these were plated on a laminin-coated coverslip and treated with poloxamer 188 (P188; PEO75-PPO30-PEO75; 8400 g/mol), architecturally inverted triblock (PPO15-PEO200-PPO15, 10,700 g/mol), and diblock (PEO75-PPO16-C4, 4200 g/mol) copolymers. We assessed the twitch kinetics of sarcomere length (SL) and intracellular Ca2+ transient by Fura-2AM by field stimulation (25 V, 0.2 Hz, 25 °C). Twitch contraction peak SL shortening of mdx FDB fibers was markedly depressed to 30% of the dystrophin-replete control FDB fibers from C57BL10 (P < 0.001). Compared to vehicle-treated mdx FDB fibers, copolymer treatment robustly and rapidly restored the twitch peak SL shortening (all P < 0.05) by P188 (15 μM = + 110%, 150 μM = + 220%), diblock (15 μM = + 50%, 150 μM = + 50%), and inverted triblock copolymer (15 μM = + 180%, 150 μM = + 90%). Twitch peak Ca2+ transient from mdx FDB fibers was also depressed compared to C57BL10 FDB fibers (P < 0.001). P188 and inverted triblock copolymer treatment of mdx FDB fibers increased the twitch peak Ca2+ transient (P < 0.001). This study shows synthetic block copolymers with varied architectures can rapidly and highly effectively enhance contractile function in live dystrophin-deficient skeletal muscle fibers.
Collapse
Affiliation(s)
- Dongwoo Hahn
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Joseph D Quick
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Brian R Thompson
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Adelyn Crabtree
- Chemical Engineering & Materials Science, University of Minnesota, 151 Amundson Hall, 421 Washington Avenue SE, Minneapolis, MN, 55455, USA
| | - Benjamin J Hackel
- Chemical Engineering & Materials Science, University of Minnesota, 151 Amundson Hall, 421 Washington Avenue SE, Minneapolis, MN, 55455, USA
| | - Frank S Bates
- Chemical Engineering & Materials Science, University of Minnesota, 151 Amundson Hall, 421 Washington Avenue SE, Minneapolis, MN, 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology & Physiology, Medical School, University of Minnesota, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
43
|
Acin‐Perez R, Benincá C, Fernandez del Rio L, Shu C, Baghdasarian S, Zanette V, Gerle C, Jiko C, Khairallah R, Khan S, Rincon Fernandez Pacheco D, Shabane B, Erion K, Masand R, Dugar S, Ghenoiu C, Schreiner G, Stiles L, Liesa M, Shirihai OS. Inhibition of ATP synthase reverse activity restores energy homeostasis in mitochondrial pathologies. EMBO J 2023; 42:e111699. [PMID: 36912136 PMCID: PMC10183817 DOI: 10.15252/embj.2022111699] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 03/14/2023] Open
Abstract
The maintenance of cellular function relies on the close regulation of adenosine triphosphate (ATP) synthesis and hydrolysis. ATP hydrolysis by mitochondrial ATP Synthase (CV) is induced by loss of proton motive force and inhibited by the mitochondrial protein ATPase inhibitor (ATPIF1). The extent of CV hydrolytic activity and its impact on cellular energetics remains unknown due to the lack of selective hydrolysis inhibitors of CV. We find that CV hydrolytic activity takes place in coupled intact mitochondria and is increased by respiratory chain defects. We identified (+)-Epicatechin as a selective inhibitor of ATP hydrolysis that binds CV while preventing the binding of ATPIF1. In cells with Complex-III deficiency, we show that inhibition of CV hydrolytic activity by (+)-Epichatechin is sufficient to restore ATP content without restoring respiratory function. Inhibition of CV-ATP hydrolysis in a mouse model of Duchenne Muscular Dystrophy is sufficient to improve muscle force without any increase in mitochondrial content. We conclude that the impact of compromised mitochondrial respiration can be lessened using hydrolysis-selective inhibitors of CV.
Collapse
Affiliation(s)
- Rebeca Acin‐Perez
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Cristiane Benincá
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Lucia Fernandez del Rio
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Cynthia Shu
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Siyouneh Baghdasarian
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Vanessa Zanette
- Department of BioinformaticsUniversity Federal of ParanaCuritibaBrazil
| | - Christoph Gerle
- Institute for Protein ResearchOsaka UniversitySuitaJapan
- RIKEN SPring‐8 CenterSayo‐gunJapan
| | - Chimari Jiko
- Institute for Integrated Radiation and Nuclear ScienceKyoto UniversityKyotoJapan
| | | | | | | | - Byourak Shabane
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | | | | | | | | | | | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Marc Liesa
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular Cellular Integrative PhysiologyUniversity of CaliforniaLos AngelesCAUSA
- Institut de Biologia Molecular de Barcelona, IBMB, CSICBarcelonaCataloniaSpain
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular Cellular Integrative PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
44
|
Cahill T, Chan S, Overton IM, Hardiman G. Transcriptome Profiling Reveals Enhanced Mitochondrial Activity as a Cold Adaptive Strategy to Hypothermia in Zebrafish Muscle. Cells 2023; 12:1366. [PMID: 37408201 PMCID: PMC10216211 DOI: 10.3390/cells12101366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/01/2023] [Accepted: 05/07/2023] [Indexed: 07/07/2023] Open
Abstract
The utilisation of synthetic torpor for interplanetary travel once seemed farfetched. However, mounting evidence points to torpor-induced protective benefits from the main hazards of space travel, namely, exposure to radiation and microgravity. To determine the radio-protective effects of an induced torpor-like state we exploited the ectothermic nature of the Danio rerio (zebrafish) in reducing their body temperatures to replicate the hypothermic states seen during natural torpor. We also administered melatonin as a sedative to reduce physical activity. Zebrafish were then exposed to low-dose radiation (0.3 Gy) to simulate radiation exposure on long-term space missions. Transcriptomic analysis found that radiation exposure led to an upregulation of inflammatory and immune signatures and a differentiation and regeneration phenotype driven by STAT3 and MYOD1 transcription factors. In addition, DNA repair processes were downregulated in the muscle two days' post-irradiation. The effects of hypothermia led to an increase in mitochondrial translation including genes involved in oxidative phosphorylation and a downregulation of extracellular matrix and developmental genes. Upon radiation exposure, increases in endoplasmic reticulum stress genes were observed in a torpor+radiation group with downregulation of immune-related and ECM genes. Exposing hypothermic zebrafish to radiation also resulted in a downregulation of ECM and developmental genes however, immune/inflammatory related pathways were downregulated in contrast to that observed in the radiation only group. A cross-species comparison was performed with the muscle of hibernating Ursus arctos horribilis (brown bear) to define shared mechanisms of cold tolerance. Shared responses show an upregulation of protein translation and metabolism of amino acids, as well as a hypoxia response with the shared downregulation of glycolysis, ECM, and developmental genes.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK;
| | - Sherine Chan
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
- JLABS at the Children’s National Research and Innovation Campus, Washington, DC 20012, USA
| | - Ian M. Overton
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK;
| | - Gary Hardiman
- School of Biological Sciences, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK;
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
45
|
Wang K, Smith SH, Iijima H, Hettinger ZR, Mallepally A, Shroff SG, Ambrosio F. Bioengineered 3D Skeletal Muscle Model Reveals Complement 4b as a Cell-Autonomous Mechanism of Impaired Regeneration with Aging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207443. [PMID: 36650030 DOI: 10.1002/adma.202207443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/11/2022] [Indexed: 05/17/2023]
Abstract
A mechanistic understanding of cell-autonomous skeletal muscle changes after injury can lead to novel interventions to improve functional recovery in an aged population. However, major knowledge gaps persist owing to limitations of traditional biological aging models. 2D cell culture represents an artificial environment, while aging mammalian models are contaminated by influences from non-muscle cells and other organs. Here, a 3D muscle aging system is created to overcome the limitations of these traditional platforms. It is shown that old muscle constructs (OMC) manifest a sarcopenic phenotype, as evidenced by hypotrophic myotubes, reduced contractile function, and decreased regenerative capacity compared to young muscle constructs. OMC also phenocopy the regenerative responses of aged muscle to two interventions, pharmacological and biological. Interrogation of muscle cell-specific mechanisms that contribute to impaired regeneration over time further reveals that an aging-induced increase of complement component 4b (C4b) delays muscle progenitor cell amplification and impairs functional recovery. However, administration of complement factor I, a C4b inactivator, improves muscle regeneration in vitro and in vivo, indicating that C4b inhibition may be a novel approach to enhance aged muscle repair. Collectively, the model herein exhibits capabilities to study cell-autonomous changes in skeletal muscle during aging, regeneration, and intervention.
Collapse
Affiliation(s)
- Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephen H Smith
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Zachary R Hettinger
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Medicine, Division of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Adarsh Mallepally
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sanjeev G Shroff
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
46
|
Sarcoplasmic Reticulum Ca 2+ Buffer Proteins: A Focus on the Yet-To-Be-Explored Role of Sarcalumenin in Skeletal Muscle Health and Disease. Cells 2023; 12:cells12050715. [PMID: 36899851 PMCID: PMC10000884 DOI: 10.3390/cells12050715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Sarcalumenin (SAR) is a luminal Ca2+ buffer protein with high capacity but low affinity for calcium binding found predominantly in the longitudinal sarcoplasmic reticulum (SR) of fast- and slow-twitch skeletal muscles and the heart. Together with other luminal Ca2+ buffer proteins, SAR plays a critical role in modulation of Ca2+ uptake and Ca2+ release during excitation-contraction coupling in muscle fibers. SAR appears to be important in a wide range of other physiological functions, such as Sarco-Endoplasmic Reticulum Calcium ATPase (SERCA) stabilization, Store-Operated-Calcium-Entry (SOCE) mechanisms, muscle fatigue resistance and muscle development. The function and structural features of SAR are very similar to those of calsequestrin (CSQ), the most abundant and well-characterized Ca2+ buffer protein of junctional SR. Despite the structural and functional similarity, very few targeted studies are available in the literature. The present review provides an overview of the role of SAR in skeletal muscle physiology, as well as of its possible involvement and dysfunction in muscle wasting disorders, in order to summarize the current knowledge on SAR and drive attention to this important but still underinvestigated/neglected protein.
Collapse
|
47
|
Ishii MN, Nakashima M, Kamiguchi H, Zach N, Kuboki R, Baba R, Hirakawa T, Suzuki K, Quinton M. Urine titin as a novel biomarker for Duchenne muscular dystrophy. Neuromuscul Disord 2023; 33:302-308. [PMID: 36871413 DOI: 10.1016/j.nmd.2023.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/10/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is the most severe form of muscular dystrophy that is caused by lack of dystrophin, a critical structural protein in skeletal muscle. DMD treatments, and quantitative biomarkers to assess the efficacy of potential treatments, are urgently needed. Previous evidence has shown that titin, a muscle cell protein, is increased in the urine of patients with DMD, suggesting its usefulness as a DMD biomarker. Here, we demonstrated that the elevated titin in urine is directly associated with the lack of dystrophin and urine titin responses to drug treatment. We performed a drug intervention study using mdx mice, a DMD mouse model. We showed that mdx mice, which lack dystrophin due to a mutation in exon 23 of the Dmd gene, have elevated urine titin. Treatment with an exon skipper that targets exon 23 rescued muscle dystrophin level and dramatically decreased urine titin in mdx mice and correlates with dystrophin expression. We also demonstrated that titin levels were significantly increased in the urine of patients with DMD. This suggests that elevated urine titin level might be a hallmark of DMD and a useful pharmacodynamic marker for therapies designed to restore dystrophin levels.
Collapse
Affiliation(s)
- Misawa Niki Ishii
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan.
| | - Masato Nakashima
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan
| | - Hidenori Kamiguchi
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan
| | - Neta Zach
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, US, 350 Massachusetts Ave Cambridge, MA 02139, United Kingdom
| | - Ryosuke Kuboki
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan
| | - Rina Baba
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan
| | - Takeshi Hirakawa
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan
| | - Kazunori Suzuki
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Tokyo, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Japan
| | - Maria Quinton
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, US, 350 Massachusetts Ave Cambridge, MA 02139, United Kingdom
| |
Collapse
|
48
|
Morales ED, Yue Y, Watkins TB, Han J, Pan X, Gibson AM, Hu B, Brito‐Estrada O, Yao G, Makarewich CA, Babu GJ, Duan D. Dwarf Open Reading Frame (DWORF) Gene Therapy Ameliorated Duchenne Muscular Dystrophy Cardiomyopathy in Aged mdx Mice. J Am Heart Assoc 2023; 12:e027480. [PMID: 36695318 PMCID: PMC9973626 DOI: 10.1161/jaha.122.027480] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/21/2022] [Indexed: 01/26/2023]
Abstract
Background Cardiomyopathy is a leading health threat in Duchenne muscular dystrophy (DMD). Cytosolic calcium upregulation is implicated in DMD cardiomyopathy. Calcium is primarily removed from the cytosol by the sarcoendoplasmic reticulum calcium ATPase (SERCA). SERCA activity is reduced in DMD. Improving SERCA function may treat DMD cardiomyopathy. Dwarf open reading frame (DWORF) is a recently discovered positive regulator for SERCA, hence, a potential therapeutic target. Methods and Results To study DWORF's involvement in DMD cardiomyopathy, we quantified DWORF expression in the heart of wild-type mice and the mdx model of DMD. To test DWORF gene therapy, we engineered and characterized an adeno-associated virus serotype 9-DWORF vector. To determine if this vector can mitigate DMD cardiomyopathy, we delivered it to 6-week-old mdx mice (6×1012 vector genome particles/mouse) via the tail vein. Exercise capacity, heart histology, and cardiac function were examined at 18 months of age. We found DWORF expression was significantly reduced at the transcript and protein levels in mdx mice. Adeno-associated virus serotype 9-DWORF vector significantly enhanced SERCA activity. Systemic adeno-associated virus serotype 9-DWORF therapy reduced myocardial fibrosis and improved treadmill running, electrocardiography, and heart hemodynamics. Conclusions Our data suggest that DWORF deficiency contributes to SERCA dysfunction in mdx mice and that DWORF gene therapy holds promise to treat DMD cardiomyopathy.
Collapse
Affiliation(s)
- Emily D. Morales
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Thais B. Watkins
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Jin Han
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Aaron M. Gibson
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical CenterThe Heart InstituteCincinnatiOH
| | - Bryan Hu
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Omar Brito‐Estrada
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical CenterThe Heart InstituteCincinnatiOH
| | - Gang Yao
- Department of Biomedical, Biological & Chemical Engineering, College of EngineeringThe University of MissouriColumbiaMO
| | - Catherine A. Makarewich
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical CenterThe Heart InstituteCincinnatiOH
- Department of PediatricsThe University of Cincinnati College of MedicineCincinnatiOH
| | - Gopal J. Babu
- Department of Cell Biology and Molecular MedicineRutgers, New Jersey Medical SchoolNewarkNJ
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
- Department of Biomedical, Biological & Chemical Engineering, College of EngineeringThe University of MissouriColumbiaMO
- Department of Neurology, School of MedicineThe University of MissouriColumbiaMO
- Department of Biomedical Sciences, College of Veterinary MedicineThe University of MissouriColumbiaMO
| |
Collapse
|
49
|
Hunter KD, Crozier RWE, Braun JL, Fajardo VA, MacNeil AJ. Acute activation of SERCA with CDN1163 attenuates IgE-mediated mast cell activation through selective impairment of ROS and p38 signaling. FASEB J 2023; 37:e22748. [PMID: 36624659 DOI: 10.1096/fj.202201272r] [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: 08/05/2022] [Revised: 11/13/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
Mast cells are granulocytic immune sentinels present in vascularized tissues that drive chronic inflammatory mechanisms characteristic of allergic pathologies. IgE-mediated mast cell activation leads to a rapid mobilization of Ca2+ from intracellular stores, which is essential for the release of preformed mediators via degranulation and de novo synthesized proinflammatory cytokines and chemokines. Given its potent signaling capacity, the dynamics of Ca2+ localization are highly regulated by various pumps and channels controlling cytosolic Ca2+ concentrations. Among these is sarco/endoplasmic reticulum Ca2+ -ATPase (SERCA), which functions to maintain low cytosolic Ca2+ concentrations by actively transporting cytosolic Ca2+ ions into the endoplasmic reticulum. In this study, we characterized the role of SERCA in allergen-activated mast cells using IgE-sensitized bone marrow-derived mast cells (BMMCs) treated with the SERCA activating compound, CDN1163, and simultaneously stimulated with allergen through FcεRI under stem cell factor (SCF) potentiation. Acute treatment with CDN1163 was found to attenuate early phase mast cell degranulation along with reactive oxygen species (ROS) production. Additionally, treatment with CDN1163 significantly reduced secretion of IL-6, IL-13, and CCL3, suggesting a role for SERCA in the late phase mast cell response. The protective effects of SERCA activation via CDN1163 treatment on the early and late phase mast cell response may be driven by the selective suppression of p38 MAPK signaling. Together, these findings implicate SERCA as an important regulator of the mast cell response to allergen and suggest SERCA activity may offer therapeutic potential targeting allergic pathologies, warranting further investigation.
Collapse
Affiliation(s)
- Katie D Hunter
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Robert W E Crozier
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Jessica L Braun
- Department of Kinesiology, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| |
Collapse
|
50
|
Valduga AH, Mizobuti DS, Moraes FDSR, Mâncio RD, Moraes LHR, Hermes TDA, Macedo AB, Minatel E. Protection of dystrophic muscle cells using Idebenone correlates with the interplay between calcium, oxidative stress and inflammation. Int J Exp Pathol 2023; 104:4-12. [PMID: 36565155 PMCID: PMC9845605 DOI: 10.1111/iep.12463] [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: 08/25/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/25/2022] Open
Abstract
There is strong cross-talk between abnormal intracellular calcium concentration, high levels of reactive oxygen species (ROS) and an exacerbated inflammatory process in the dystrophic muscles of mdx mice, the experimental model of Duchenne muscular dystrophy (DMD). In this study, we investigated effects of Idebenone, a potent anti-oxidant, on oxidative stress markers, the anti-oxidant defence system, intracellular calcium concentrations and the inflammatory process in primary dystrophic muscle cells from mdx mice. Dystrophic muscle cells were treated with Idebenone (0.05 μM) for 24 h. The untreated mdx muscle cells were used as controls. The MTT assay showed that Idebenone did not have a cytotoxic effect on the dystrophic muscle cells. The Idebenone treatment was able to reduce the levels of oxidative stress markers, such as H2 O2 and 4-HNE, as well as decreasing intracellular calcium influx in the dystrophic muscle cells. Regarding Idebenone effects on the anti-oxidant defence system, an up-regulation of catalase levels, glutathione reductase (GR), glutathione peroxidase (GPx) and superoxide dismutase (SOD) activity was observed in the dystrophic muscle cells. In addition, the Idebenone treatment was also associated with reduction in inflammatory molecules, such as nuclear factor kappa-B (NF-κB) and tumour necrosis factor (TNF) in mdx muscle cells. These outcomes supported the use of Idebenone as a protective agent against oxidative stress and related signalling mechanisms involved in dystrophinopathies, such as DMD.
Collapse
Affiliation(s)
- Amanda Harduim Valduga
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Daniela Sayuri Mizobuti
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Fernanda dos Santos Rapucci Moraes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Rafael Dias Mâncio
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Luis Henrique Rapucci Moraes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Túlio de Almeida Hermes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Aline Barbosa Macedo
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Elaine Minatel
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| |
Collapse
|