1
|
Gerhalter T, Marty B, Gast L, Roemer F, Baudin P, Trollmann R, Uder M, Carlier P, Nagel A. Longitudinal Follow-Up of Patients With Duchenne Muscular Dystrophy Using Quantitative 23Na and 1H MRI. J Cachexia Sarcopenia Muscle 2025; 16:e13812. [PMID: 40254293 PMCID: PMC12009636 DOI: 10.1002/jcsm.13812] [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: 07/22/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Quantitative muscle MRI commonly evaluates disease activity and muscle wasting in Duchenne muscular dystrophy (DMD). Disturbances in ion homeostasis contribute to DMD pathophysiology, but their relationships with disease progression is unclear. 23Na MRI may provide insights into the disease course and treatment response. This longitudinal study assessed whether sodium levels are elevated in DMD patients regardless of fat fraction (FF) and whether baseline sodium levels influence FF changes over time. Additionally, we quantified the effect of slice selection on measured sodium values. METHODS Thirteen DMD boys (age 7.8 ± 2.4 years) underwent MRI of lower leg muscles at 3T at three visits, spaced 6 months apart. We assessed FF for disease progression and water T2, pH, apparent tissue sodium concentration (aTSC), and intracellular-weighted 23Na signal (ICwS) for disease activity. Fourteen healthy boys (age 9.5 ± 1.7 years) underwent the same MRI protocol once. Linear regression and mixed-effect modelling were used to examine sodium level increases and their impact on FF changes. RESULTS In DMD, muscles with FF < 10% exhibited significantly elevated aTSC (24.8 ± 4.6 mM vs. 14.5 ± 2.1 mM in controls, p < 0.001) and higher ICwS (23.6 ± 2.5 a.u. vs. 14.1 ± 2.1 a.u., p < 0.001). At Visit 1, FF values showed a significant negative association with aTSC (β = -17.30, p = 0.016) and ICwS (β = -21.02, p < 0.001). The first mixed-effect model, which assessed aTSC alone, showed no significant effect on FF progression but indicated a weak trend (p = 0.098). The second, more comprehensive model-incorporating also ICwS and water T2-revealed that FF changes were positively associated with aTSC (p = 0.0023) and negatively associated with ICwS and wT2 (p < 0.001 and p = 0.025, respectively), with ICwS showing a significant interaction with time (p = 0.0033). Varying slice positioning and slice number demonstrated minimal impact on aTSC and ICwS, with low CV (2%-4%) in the mid-belly region. CONCLUSIONS The study demonstrates significant MRI-based changes related to dystrophic alterations in DMD. We identified early alterations in sodium homeostasis, independent of FF. Our findings suggest that the relationship between sodium levels and FF progression is complex and may not be fully explained by total sodium measurements alone. Given the small sample size, further validation in larger cohorts is needed. Combined 1H and 23Na-MRI may offer deeper insights into how metabolic and ionic changes interact with FF progression and overall disease activity.
Collapse
Affiliation(s)
- Teresa Gerhalter
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Benjamin Marty
- NMR LaboratoryNeuromuscular Investigation Center, Institute of MyologyParisFrance
| | - Lena V. Gast
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Frank Roemer
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Pierre‐Yves Baudin
- NMR LaboratoryNeuromuscular Investigation Center, Institute of MyologyParisFrance
| | - Regina Trollmann
- Department of Pediatrics, Division NeuropediatricsFAUErlangenGermany
| | - Michael Uder
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Pierre G. Carlier
- Saint‐Luc University HospitalBrusselsBelgium
- Erasme University HospitalBrusselsBelgium
- Liège State UniversityLiègeBelgium
| | - Armin M. Nagel
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
- Division of Medical Physics in RadiologyDKFZHeidelbergGermany
| |
Collapse
|
2
|
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
|
3
|
Auger M, Sorroza-Martinez L, Brahiti N, Huppé CA, Faucher-Giguère L, Arbi I, Hervault M, Cheng X, Gaillet B, Couture F, Guay D, Soultan AH. Enhancing peptide and PMO delivery to mouse airway epithelia by chemical conjugation with the amphiphilic peptide S10. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102290. [PMID: 39233851 PMCID: PMC11372590 DOI: 10.1016/j.omtn.2024.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/26/2024] [Indexed: 09/06/2024]
Abstract
Delivery of antisense oligonucleotides (ASOs) to airway epithelial cells is arduous due to the physiological barriers that protect the lungs and the endosomal entrapment phenomenon, which prevents ASOs from reaching their intracellular targets. Various delivery strategies involving peptide-, lipid-, and polymer-based carriers are being investigated, yet the challenge remains. S10 is a peptide-based delivery agent that enables the intracellular delivery of biomolecules such as GFP, CRISPR-associated nuclease ribonucleoprotein (RNP), base editor RNP, and a fluorescent peptide into lung cells after intranasal or intratracheal administrations to mice, ferrets, and rhesus monkeys. Herein, we demonstrate that covalently attaching S10 to a fluorescently labeled peptide or a functional splice-switching phosphorodiamidate morpholino oligomer improves their intracellular delivery to airway epithelia in mice after a single intranasal instillation. Data reveal a homogeneous delivery from the trachea to the distal region of the lungs, specifically into the cells lining the airway. Quantitative measurements further highlight that conjugation via a disulfide bond through a pegylated (PEG) linker was the most beneficial strategy compared with direct conjugation (without the PEG linker) or conjugation via a permanent thiol-maleimide bond. We believe that S10-based conjugation provides a great strategy to achieve intracellular delivery of peptides and ASOs with therapeutic properties in lungs.
Collapse
Affiliation(s)
- Maud Auger
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
- Département de génie chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot 1065, av. de la Médecine, Bureau 3550, Québec, QC G1V 0A6, Canada
| | - Luis Sorroza-Martinez
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
- Département de génie chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot 1065, av. de la Médecine, Bureau 3550, Québec, QC G1V 0A6, Canada
| | - Nadine Brahiti
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
| | - Carole-Ann Huppé
- Centre Collégial de Transfert de Technologie en Biotechnologies TransBIOTech, 201 Rue Monseigneur-Bourget, Lévis, QC G6V 6Z3, Canada
| | | | - Imen Arbi
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
| | - Maxime Hervault
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
| | - Xue Cheng
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
| | - Bruno Gaillet
- Département de génie chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot 1065, av. de la Médecine, Bureau 3550, Québec, QC G1V 0A6, Canada
| | - Frédéric Couture
- Centre Collégial de Transfert de Technologie en Biotechnologies TransBIOTech, 201 Rue Monseigneur-Bourget, Lévis, QC G6V 6Z3, Canada
| | - David Guay
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
- Département de génie chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot 1065, av. de la Médecine, Bureau 3550, Québec, QC G1V 0A6, Canada
| | - Al-Halifa Soultan
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC G1P 4S6, Canada
| |
Collapse
|
4
|
Guedira G, Petermann O, Scapozza L, Ismail HM. Diapocynin treatment induces functional and structural improvements in an advanced disease state in the mdx 5Cv mice. Biomed Pharmacother 2024; 177:116957. [PMID: 38908198 DOI: 10.1016/j.biopha.2024.116957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/30/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular disorder affecting children. It affects nearly 1 male birth over 5000. Oxidative stress is a pervasive feature in the pathogenesis of DMD. Recent work shows that the main generators of ROS are NADPH oxidases (NOX), suggesting that they are an early and promising target in DMD. In addition, skeletal muscles of mdx mice, a murine model of DMD, overexpress NOXes. We investigated the impact of diapocynin, a dimer of the NOX inhibitor apocynin, on the chronic disease phase of mdx5Cv mice. Treatment of these mice with diapocynin from 7 to 10 months of age resulted in decreased hypertrophy of several muscles, prevented force loss induced by tetanic and eccentric contractions, improved muscle and respiratory functions, decreased fibrosis of the diaphragm and positively regulated the expression of disease modifiers. These encouraging results ensure the potential role of diapocynin in future treatment strategies.
Collapse
Affiliation(s)
- Ghali Guedira
- Pharmaceutical Biochemistry/Chemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Olivier Petermann
- Pharmaceutical Biochemistry/Chemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry/Chemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| | - Hesham M Ismail
- Pharmaceutical Biochemistry/Chemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
5
|
Aldharee H. Duchenne muscular dystrophy in Saudi Arabia: a review of the current literature. Front Neurol 2024; 15:1392274. [PMID: 39087004 PMCID: PMC11288836 DOI: 10.3389/fneur.2024.1392274] [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: 02/27/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
In the past three decades, significant improvements have occurred in the study of Duchenne muscular dystrophy (DMD). DMD is a rare, severe neuromuscular disease that causes death due to cardiovascular and respiratory complications among affected boys. Since the 1980s, ongoing preclinical and clinical studies have been conducted to explore the disease in depth and discover potential therapeutic strategies. In Saudi Arabia, it is unclear whether health services and research efforts are keeping pace with global achievements. Therefore, this review aims to explore the diagnostic and management strategies and research efforts in Saudi Arabia over the past three decades. I searched the PubMed/Medline, Scopus, and Web of Science databases and included all published articles on the epidemiology, genetics, diagnosis, and management of DMD/BMD in this review. The findings suggest a lack of local standardized diagnostic strategies, a poor understanding of epidemiology and common pathogenic variants, and a critical need for preclinical and clinical research. At the time of writing, no such comprehensive review has been published. Challenges, limitations, and future perspectives are also discussed in this article.
Collapse
Affiliation(s)
- Hitham Aldharee
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|
6
|
Gandhi S, Sweeney HL, Hart CC, Han R, Perry CGR. Cardiomyopathy in Duchenne Muscular Dystrophy and the Potential for Mitochondrial Therapeutics to Improve Treatment Response. Cells 2024; 13:1168. [PMID: 39056750 PMCID: PMC11274633 DOI: 10.3390/cells13141168] [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: 01/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by mutations to the dystrophin gene, resulting in deficiency of dystrophin protein, loss of myofiber integrity in skeletal and cardiac muscle, and eventual cell death and replacement with fibrotic tissue. Pathologic cardiac manifestations occur in nearly every DMD patient, with the development of cardiomyopathy-the leading cause of death-inevitable by adulthood. As early cardiac abnormalities are difficult to detect, timely diagnosis and appropriate treatment modalities remain a challenge. There is no cure for DMD; treatment is aimed at delaying disease progression and alleviating symptoms. A comprehensive understanding of the pathophysiological mechanisms is crucial to the development of targeted treatments. While established hypotheses of underlying mechanisms include sarcolemmal weakening, upregulation of pro-inflammatory cytokines, and perturbed ion homeostasis, mitochondrial dysfunction is thought to be a potential key contributor. Several experimental compounds targeting the skeletal muscle pathology of DMD are in development, but the effects of such agents on cardiac function remain unclear. The synergistic integration of small molecule- and gene-target-based drugs with metabolic-, immune-, or ion balance-enhancing compounds into a combinatorial therapy offers potential for treating dystrophin deficiency-induced cardiomyopathy, making it crucial to understand the underlying mechanisms driving the disorder.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Cora C. Hart
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
7
|
Ó Murchú SC, O'Halloran KD. BREATHE DMD: boosting respiratory efficacy after therapeutic hypoxic episodes in Duchenne muscular dystrophy. J Physiol 2024; 602:3255-3272. [PMID: 38837229 DOI: 10.1113/jp280280] [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: 03/08/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal genetic neuromuscular disorder, characterised by progressive decline in skeletal muscle function due to the secondary consequences of dystrophin deficiency. Weakness extends to the respiratory musculature, and cardiorespiratory failure is the leading cause of death in men with DMD. Intermittent hypoxia has emerged as a potential therapy to counteract ventilatory insufficiency by eliciting long-term facilitation of breathing. Mechanisms of sensory and motor facilitation of breathing have been well delineated in animal models. Various paradigms of intermittent hypoxia have been designed and implemented in human trials culminating in clinical trials in people with spinal cord injury and amyotrophic lateral sclerosis. Application of therapeutic intermittent hypoxia to DMD is considered together with discussion of the potential barriers to progression owing to the complexity of this devastating disease. Notwithstanding the considerable challenges and potential pitfalls of intermittent hypoxia-based therapies for DMD, we suggest it is incumbent on the research community to explore the potential benefits in pre-clinical models. Intermittent hypoxia paradigms should be implemented to explore the proclivity to express respiratory plasticity with the longer-term aim of preserving and potentiating ventilation in pre-clinical models and people with DMD.
Collapse
Affiliation(s)
- Seán C Ó Murchú
- Department of Physiology, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
8
|
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
|
9
|
Terrill JR, Bautista APR, Tsioutsias I, Grounds MD, Arthur PG. Oxidised Albumin Levels in Plasma and Skeletal Muscle as Biomarkers of Disease Progression and Treatment Efficacy in Dystrophic mdx Mice. Antioxidants (Basel) 2024; 13:720. [PMID: 38929159 PMCID: PMC11201235 DOI: 10.3390/antiox13060720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Redox modifications to the plasma protein albumin have the potential to be used as biomarkers of disease progression and treatment efficacy in pathologies associated with inflammation and oxidative stress. One such pathology is Duchenne muscular dystrophy (DMD), a fatal childhood disease characterised by severe muscle wasting. We have previously shown in the mdx mouse model of DMD that plasma albumin thiol oxidation is increased; therefore, the first aim of this paper was to establish that albumin thiol oxidation in plasma reflects levels within mdx muscle tissue. We therefore developed a method to measure tissue albumin thiol oxidation. We show that albumin thiol oxidation was increased in both mdx muscle and plasma, with levels correlated with measures of dystropathology. In dystrophic muscle, albumin content was associated with areas of myonecrosis. The second aim was to test the ability of plasma thiol oxidation to track acute changes in dystropathology: we therefore subjected mdx mice to a single treadmill exercise session (known to increase myonecrosis) and took serial blood samples. This acute exercise caused a transient increase in total plasma albumin oxidation and measures of dystropathology. Together, these data support the use of plasma albumin thiol oxidation as a biomarker to track active myonecrosis in DMD.
Collapse
Affiliation(s)
- Jessica R. Terrill
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.R.T.); (A.P.R.B.); (I.T.)
| | - Angelo Patrick R. Bautista
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.R.T.); (A.P.R.B.); (I.T.)
| | - Irene Tsioutsias
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.R.T.); (A.P.R.B.); (I.T.)
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia;
| | - Miranda D. Grounds
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia;
| | - Peter G. Arthur
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.R.T.); (A.P.R.B.); (I.T.)
| |
Collapse
|
10
|
Gasparella F, Nogara L, Germinario E, Tibaudo L, Ciciliot S, Piccoli G, Venegas FC, Fontana F, Sales G, Sabbatini D, Foot J, Jarolimek W, Blaauw B, Canton M, Vitiello L. A Novel MAO-B/SSAO Inhibitor Improves Multiple Aspects of Dystrophic Phenotype in mdx Mice. Antioxidants (Basel) 2024; 13:622. [PMID: 38929061 PMCID: PMC11201281 DOI: 10.3390/antiox13060622] [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: 01/31/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is one of the most frequent and severe childhood muscle diseases. Its pathophysiology is multifaceted and still incompletely understood, but we and others have previously shown that oxidative stress plays an important role. In particular, we have demonstrated that inhibition of mitochondrial monoamine oxidases could improve some functional and biohumoral markers of the pathology. In the present study we report the use of dystrophic mdx mice to evaluate the efficacy of a dual monoamine oxidase B (MAO-B)/semicarbazide-sensitive amine oxidase (SSAO) inhibitor, PXS-5131, in reducing inflammation and fibrosis and improving muscle function. We found that a one-month treatment starting at three months of age was able to decrease reactive oxygen species (ROS) production, fibrosis, and inflammatory infiltrate in the tibialis anterior (TA) and diaphragm muscles. Importantly, we also observed a marked improvement in the capacity of the gastrocnemius muscle to maintain its force when challenged with eccentric contractions. Upon performing a bulk RNA-seq analysis, PXS-5131 treatment affected the expression of genes involved in inflammatory processes and tissue remodeling. We also studied the effect of prolonged treatment in older dystrophic mice, and found that a three-month administration of PXS-5131 was able to greatly reduce the progression of fibrosis not only in the diaphragm but also in the heart. Taken together, these results suggest that PXS-5131 is an effective inhibitor of fibrosis and inflammation in dystrophic muscles, a finding that could open a new therapeutic avenue for DMD patients.
Collapse
Affiliation(s)
- Francesca Gasparella
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.G.); (F.F.); (G.S.)
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy;
- Department of Pharmaceutical Sciences, University of Padova, 35131 Padova, Italy
| | - Elena Germinario
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
| | - Lucia Tibaudo
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.G.); (F.F.); (G.S.)
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
| | - Stefano Ciciliot
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy;
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Giorgia Piccoli
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy;
| | - Francisca Carolina Venegas
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy
| | - Francesca Fontana
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.G.); (F.F.); (G.S.)
| | - Gabriele Sales
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.G.); (F.F.); (G.S.)
| | - Daniele Sabbatini
- Department of Neurosciences, University of Padova, 35128 Padova, Italy;
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35131 Padova, Italy
| | - Jonathan Foot
- Syntara Ltd., Sydney, NSW 2086, Australia; (J.F.); (W.J.)
| | | | - Bert Blaauw
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy;
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (L.N.); (E.G.); (G.P.); (F.C.V.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy
| | - Libero Vitiello
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.G.); (F.F.); (G.S.)
| |
Collapse
|
11
|
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
|
12
|
Rose N, Estrada Chavez B, Sonam S, Nguyen T, Grenci G, Bigot A, Muchir A, Ladoux B, Cadot B, Le Grand F, Trichet L. Bioengineering a miniaturized in vitro 3D myotube contraction monitoring chip to model muscular dystrophies. Biomaterials 2023; 293:121935. [PMID: 36584444 DOI: 10.1016/j.biomaterials.2022.121935] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/17/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022]
Abstract
Quantification of skeletal muscle functional contraction is essential to assess the outcomes of therapeutic procedures for neuromuscular disorders. Muscle three-dimensional "Organ-on-chip" models usually require a substantial amount of biological material, which rarely can be obtained from patient biopsies. Here, we developed a miniaturized 3D myotube culture chip with contraction monitoring capacity at the single cell level. Optimized micropatterned substrate design enabled to obtain high culture yields in tightly controlled microenvironments, with myotubes derived from primary human myoblasts displaying spontaneous contractions. Analysis of nuclear morphology confirmed similar myonuclei structure between obtained myotubes and in vivo myofibers, as compared to 2D monolayers. LMNA-related Congenital Muscular Dystrophy (L-CMD) was modeled with successful development of diseased 3D myotubes displaying reduced contraction. The miniaturized myotube technology can thus be used to study contraction characteristics and evaluate how diseases affect muscle organization and force generation. Importantly, it requires significantly fewer starting materials than current systems, which should substantially improve drug screening capability.
Collapse
Affiliation(s)
- Nicolas Rose
- Sorbonne Université, Inserm UMRS 974, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Berenice Estrada Chavez
- Sorbonne Université, Inserm UMRS 974, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Surabhi Sonam
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France.
| | - Thao Nguyen
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France.
| | - Gianluca Grenci
- Mechanobiology Institute, National University of Singapore, 117411, Singapore.
| | - Anne Bigot
- Sorbonne Université, Inserm UMRS 974, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Antoine Muchir
- Sorbonne Université, Inserm UMRS 974, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Benoît Ladoux
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France.
| | - Bruno Cadot
- Sorbonne Université, Inserm UMRS 974, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Fabien Le Grand
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, 69008, Lyon, France.
| | - Léa Trichet
- Sorbonne Université, CNRS UMR 7574, Laboratoire de Chimie de La Matière Condensée de Paris, 75005, Paris, France.
| |
Collapse
|
13
|
Khairullin AE, Grishin SN, Ziganshin AU. P2 Receptor Signaling in Motor Units in Muscular Dystrophy. Int J Mol Sci 2023; 24:1587. [PMID: 36675094 PMCID: PMC9865441 DOI: 10.3390/ijms24021587] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The purine signaling system is represented by purine and pyrimidine nucleotides and nucleosides that exert their effects through the adenosine, P2X and P2Y receptor families. It is known that, under physiological conditions, P2 receptors play only a minor role in modulating the functions of cells and systems; however, their role significantly increases under some pathophysiological conditions, such as stress, ischemia or hypothermia, when they can play a dominant role as a signaling molecule. The diversity of P2 receptors and their wide distribution in the body make them very attractive as a target for the pharmacological action of drugs with a new mechanism of action. The review is devoted to the involvement of P2 signaling in the development of pathologies associated with a loss of muscle mass. The contribution of adenosine triphosphate (ATP) as a signal molecule in the pathogenesis of a number of muscular dystrophies (Duchenne, Becker and limb girdle muscular dystrophy 2B) is considered. To understand the processes involving the purinergic system, the role of the ATP and P2 receptors in several models associated with skeletal muscle degradation is also discussed.
Collapse
Affiliation(s)
- Adel E. Khairullin
- Department of Biochemistry, Kazan State Medical University, 420012 Kazan, Russia
- Research Laboratory of Mechanobiology, Kazan Federal University, 420008 Kazan, Russia
| | - Sergey N. Grishin
- Department of Medicinal Physics, Kazan State Medical University, 420012 Kazan, Russia
| | - Ayrat U. Ziganshin
- Department of Pharmacology, Kazan State Medical University, 420012 Kazan, Russia
| |
Collapse
|
14
|
Dowling P, Gargan S, Swandulla D, Ohlendieck K. Proteomic profiling of impaired excitation-contraction coupling and abnormal calcium handling in muscular dystrophy. Proteomics 2022; 22:e2200003. [PMID: 35902360 PMCID: PMC10078611 DOI: 10.1002/pmic.202200003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/06/2022]
Abstract
The X-linked inherited neuromuscular disorder Duchenne muscular dystrophy is characterised by primary abnormalities in the membrane cytoskeletal component dystrophin. The almost complete absence of the Dp427-M isoform of dystrophin in skeletal muscles renders contractile fibres more susceptible to progressive degeneration and a leaky sarcolemma membrane. This in turn results in abnormal calcium homeostasis, enhanced proteolysis and impaired excitation-contraction coupling. Biochemical and mass spectrometry-based proteomic studies of both patient biopsy specimens and genetic animal models of dystrophinopathy have demonstrated significant changes in the concentration and/or physiological function of essential calcium-regulatory proteins in dystrophin-lacking voluntary muscles. Abnormalities include dystrophinopathy-associated changes in voltage sensing receptors, calcium release channels, calcium pumps and calcium binding proteins. This review article provides an overview of the importance of the sarcolemmal dystrophin-glycoprotein complex and the wider dystrophin complexome in skeletal muscle and its linkage to depolarisation-induced calcium-release mechanisms and the excitation-contraction-relaxation cycle. Besides chronic inflammation, fat substitution and reactive myofibrosis, a major pathobiochemical hallmark of X-linked muscular dystrophy is represented by the chronic influx of calcium ions through the damaged plasmalemma in conjunction with abnormal intracellular calcium fluxes and buffering. Impaired calcium handling proteins should therefore be included in an improved biomarker signature of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
15
|
Kocabaş M, Kocabaş FK, Aksu Ö, Benzer F, Erişir M, Sahilli YÇ. Aroclor 1254 impairs sperm quality, fertilization ability, and embryo development of rainbow trout (Oncorhynchus mykiss). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:84581-84589. [PMID: 35781661 DOI: 10.1007/s11356-022-21789-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 06/28/2022] [Indexed: 06/15/2023]
Abstract
The polychlorinated biphenyls (PCBs) in aquatic environment adversely affect non-target organisms, including fish. Especially, the male reproduction and next generation can be damaged through high exposure to these pollutants. Hence, the sperm cells were exposed to sublethal concentrations of Aroclor 1254 (0, 1, 5, 10, or 25 mg/l) for 4 h. The sperm quality parameters were analyzed by SCA (Sperm Class Analyzer). The fertility, eyeing, and hatching rates were determined as gamete markers. Lipid peroxidation (malondialdehyde-MDA), glutathione (GSH), and antioxidant enzymes [superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT)] were measured for determination of oxidative stress. Our results showed that Aroclor 1254 negatively affected the motility rate and duration, fertilization rate, embryogenesis, and hatching and also triggered antioxidant defense mechanisms at the highest concentration (25 mg L-1). Furthermore, linear speed (VSL), linearity index (LIN), and amplitude lateral head (ALH) were significantly changed after exposure to 25 mg L-1, and the lowest concentrations (1 and 10 mg L-1) did not significantly affect the motility and fertilizing capacity. The embryogenesis and hatching were significantly affected by sperm exposure to 1, 10, and 25 mg L-1 of Aroclor 1254. Consequently, Aroclor 1254 causes potential hazards in male germ cells, and the exposure of sperm cells to pollutants can adversely affect next generation of wild populations.
Collapse
Affiliation(s)
- Mehmet Kocabaş
- Department of Wildlife Ecology and Management, Faculty of Forestry, Karadeniz Technical University, 61080, Trabzon, Turkey
| | | | - Önder Aksu
- Fisheries Faculty, Munzur University, 62000, Tunceli, Turkey
| | - Fulya Benzer
- Department of Food Engineering, Faculty of Engineering, Munzur University, Tunceli, Turkey
| | - Mine Erişir
- Department of Basic Sciences, Faculty of Veterinary, Fırat University, Elazığ, Turkey
| | - Yeliz Çakir Sahilli
- Department of Chemistry and Chemical Processing Technologies, Tunceli Vocational School, Munzur University, 62000, Tunceli, Turkey
| |
Collapse
|
16
|
Singh S, Singh T, Kunja C, Dhoat NS, Dhania NK. Gene-editing, immunological and iPSCs based therapeutics for muscular dystrophy. Eur J Pharmacol 2021; 912:174568. [PMID: 34656607 DOI: 10.1016/j.ejphar.2021.174568] [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: 06/24/2021] [Revised: 09/25/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
Muscular dystrophy is a well-known genetically heterogeneous group of rare muscle disorders. This progressive disease causes the breakdown of skeletal muscles over time and leads to grave weakness. This breakdown is caused by a diverse pattern of mutations in dystrophin and dystrophin associated protein complex. These mutations lead to the production of altered proteins in response to which, the body stimulates production of various cytokines and immune cells, particularly reactive oxygen species and NFκB. Immune cells display/exhibit a dual role by inducing muscle damage and muscle repair. Various anti-oxidants, anti-inflammatory and glucocorticoid drugs serve as potent therapeutics for muscular dystrophy. Along with the above mentioned therapeutics, induced pluripotent stem cells also serve as a novel approach paving a way for personalized treatment. These pluripotent stem cells allow regeneration of large numbers of regenerative myogenic progenitors that can be administered in muscular dystrophy patients which assist in the recovery of lost muscle fibers. In this review, we have summarized gene-editing, immunological and induced pluripotent stem cell based therapeutics for muscular dystrophy treatment.
Collapse
Affiliation(s)
- Shagun Singh
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Tejpal Singh
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Chaitanya Kunja
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Navdeep S Dhoat
- Department of Pediatrics Surgery, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Narender K Dhania
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India.
| |
Collapse
|
17
|
Cho S, Jang J. Recent Trends in Biofabrication Technologies for Studying Skeletal Muscle Tissue-Related Diseases. Front Bioeng Biotechnol 2021; 9:782333. [PMID: 34778240 PMCID: PMC8578921 DOI: 10.3389/fbioe.2021.782333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
In native skeletal muscle, densely packed myofibers exist in close contact with surrounding motor neurons and blood vessels, which are embedded in the fibrous connective tissue. In comparison to conventional two-dimensional (2D) cultures, the three-dimensional (3D) engineered skeletal muscle models allow structural and mechanical resemblance with native skeletal muscle tissue by providing geometric confinement and physiological matrix stiffness to the cells. In addition, various external stimuli applied to these models enhance muscle maturation along with cell-cell and cell-extracellular matrix interaction. Therefore, 3D in vitro muscle models can adequately recapitulate the pathophysiologic events occurring in tissue-tissue interfaces inside the native skeletal muscle such as neuromuscular junction. Moreover, 3D muscle models can induce pathological phenotype of human muscle dystrophies such as Duchenne muscular dystrophy by incorporating patient-derived induced pluripotent stem cells and human primary cells. In this review, we discuss the current biofabrication technologies for modeling various skeletal muscle tissue-related diseases (i.e., muscle diseases) including muscular dystrophies and inflammatory muscle diseases. In particular, these approaches would enable the discovery of novel phenotypic markers and the mechanism study of human muscle diseases with genetic mutations.
Collapse
Affiliation(s)
- Seungyeun Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea
| |
Collapse
|
18
|
Zabłocka B, Górecki DC, Zabłocki K. Disrupted Calcium Homeostasis in Duchenne Muscular Dystrophy: A Common Mechanism behind Diverse Consequences. Int J Mol Sci 2021; 22:11040. [PMID: 34681707 PMCID: PMC8537421 DOI: 10.3390/ijms222011040] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) leads to disability and death in young men. This disease is caused by mutations in the DMD gene encoding diverse isoforms of dystrophin. Loss of full-length dystrophins is both necessary and sufficient for causing degeneration and wasting of striated muscles, neuropsychological impairment, and bone deformities. Among this spectrum of defects, abnormalities of calcium homeostasis are the common dystrophic feature. Given the fundamental role of Ca2+ in all cells, this biochemical alteration might be underlying all the DMD abnormalities. However, its mechanism is not completely understood. While abnormally elevated resting cytosolic Ca2+ concentration is found in all dystrophic cells, the aberrant mechanisms leading to that outcome have cell-specific components. We probe the diverse aspects of calcium response in various affected tissues. In skeletal muscles, cardiomyocytes, and neurons, dystrophin appears to serve as a scaffold for proteins engaged in calcium homeostasis, while its interactions with actin cytoskeleton influence endoplasmic reticulum organisation and motility. However, in myoblasts, lymphocytes, endotheliocytes, and mesenchymal and myogenic cells, calcium abnormalities cannot be clearly attributed to the loss of interaction between dystrophin and the calcium toolbox proteins. Nevertheless, DMD gene mutations in these cells lead to significant defects and the calcium anomalies are a symptom of the early developmental phase of this pathology. As the impaired calcium homeostasis appears to underpin multiple DMD abnormalities, understanding this alteration may lead to the development of new therapies. In fact, it appears possible to mitigate the impact of the abnormal calcium homeostasis and the dystrophic phenotype in the total absence of dystrophin. This opens new treatment avenues for this incurable disease.
Collapse
Affiliation(s)
- Barbara Zabłocka
- Molecular Biology Unit, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Dariusz C. Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael’s Building, White Swan Road, Portsmouth PO1 2DT, UK
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
| | - Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland
| |
Collapse
|
19
|
Kiriaev L, Kueh S, Morley JW, Houweling PJ, Chan S, North KN, Head SI. Dystrophin-negative slow-twitch soleus muscles are not susceptible to eccentric contraction induced injury over the lifespan of the mdx mouse. Am J Physiol Cell Physiol 2021; 321:C704-C720. [PMID: 34432537 DOI: 10.1152/ajpcell.00122.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the second most common fatal genetic disease in humans and is characterized by the absence of a functional copy of the protein dystrophin from skeletal muscle. In dystrophin-negative humans and rodents, regenerated skeletal muscle fibers show abnormal branching. The number of fibers with branches and the complexity of branching increases with each cycle of degeneration/regeneration. Previously, using the mdx mouse model of DMD, we have proposed that once the number and complexity of branched fibers present in dystrophic fast-twitch EDL muscle surpasses a stable level, we term the "tipping point," the branches, in and of themselves, mechanically weaken the muscle by rupturing when subjected to high forces during eccentric contractions. Here, we use the slow-twitch soleus muscle from the dystrophic mdx mouse to study prediseased "periambulatory" dystrophy at 2-3 wk, the peak regenerative "adult" phase at 6-9 wk, and "old" at 58-112 wk. Using isolated mdx soleus muscles, we examined contractile function and response to eccentric contraction correlated with the amount and complexity of regenerated branched fibers. The intact muscle was enzymatically dispersed into individual fibers in order to count fiber branching and some muscles were optically cleared to allow laser scanning confocal microscopy. We demonstrate throughout the lifespan of the mdx mouse that dystrophic slow-twitch soleus muscle is no more susceptible to eccentric contraction-induced injury than age-matched littermate controls and that this is correlated with a reduction in the number and complexity of branched fibers compared with fast-twitch dystrophic EDL muscles.
Collapse
MESH Headings
- Age Factors
- Animals
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Kinetics
- Male
- Mice, Inbred mdx
- Muscle Contraction
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/pathology
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle Fibers, Slow-Twitch/pathology
- Muscle Strength
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Mutation
- Mice
Collapse
Affiliation(s)
- Leonit Kiriaev
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Sindy Kueh
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Peter J Houweling
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Stephen Chan
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Stewart I Head
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
20
|
Sun Z, Xu D, Zhao L, Li X, Li S, Huang X, Li C, Sun L, Liu B, Jiang Z, Zhang L. A new therapeutic effect of fenofibrate in Duchenne muscular dystrophy: The promotion of myostatin degradation. Br J Pharmacol 2021; 179:1237-1250. [PMID: 34553378 DOI: 10.1111/bph.15678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Duchenne muscular dystrophy (DMD) is a degenerative muscle disease with no effective drug treatment. This study investigated the positive effects of fenofibrate on dystrophic muscles. EXPERIMENTAL APPROACH Myostatin expression in serum and muscle tissue of DMD patients and mdx mice were tested. Primary myoblasts isolated from mdx mice were challenged with an inflammatory stimulus and treated with fenofibrate. In animal experiments, 6-week-old male mdx mice were treated with fenofibrate (100 mg/kg) administered orally once per day for 6 weeks. Tests of muscle function plus histology and biochemical analyses of serum were conducted to evaluate the effects of fenofibrate. The expressions of myostatin, MuRF1, and atrogin-1 in skeletal muscle were evaluated by Western blotting and real-time PCR. Total and oxidative myosin heavy chain (MHC) were assessed via immunofluorescence. KEY RESULTS Increased expression of myostatin protein was found in dystrophic muscle of DMD patients and mdx mice. Fenofibrate enhanced myofibre differentiation by downregulating the expression of myostatin protein but not mRNA in primary myoblasts of mdx mice. Fenofibrate significantly improved muscle function while ameliorating muscle damage in mdx mice. These benefits are accompanied by an anti-inflammatory effect. Fenofibrate treatment returned myofibre function by inhibiting the expressions of myostatin, MuRF1, and atrogin-1 protein in the gastrocnemius muscle and diaphragm, while leaving the mRNA level of myostatin unaffected. CONCLUSIONS AND IMPLICATIONS Fenofibrate substantially slows muscle dystrophy by promoting the degradation of myostatin protein, which may indicate a new therapeutic focus for DMD patients.
Collapse
Affiliation(s)
- Zeren Sun
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Lei Zhao
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Xihua Li
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Sijia Li
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Xiaofei Huang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Chunjie Li
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Bing Liu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China.,Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
21
|
Ebrahimi M, Lad H, Fusto A, Tiper Y, Datye A, Nguyen CT, Jacques E, Moyle LA, Nguyen T, Musgrave B, Chávez-Madero C, Bigot A, Chen C, Turner S, Stewart BA, Pegoraro E, Vitiello L, Gilbert PM. De novo revertant fiber formation and therapy testing in a 3D culture model of Duchenne muscular dystrophy skeletal muscle. Acta Biomater 2021; 132:227-244. [PMID: 34048976 DOI: 10.1016/j.actbio.2021.05.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
The biological basis of Duchenne muscular dystrophy (DMD) pathology is only partially characterized and there are still few disease-modifying therapies available, therein underlying the value of strategies to model and study DMD. Dystrophin, the causative gene of DMD, is responsible for linking the cytoskeleton of muscle fibers to the extracellular matrix beyond the sarcolemma. We posited that disease-associated phenotypes not yet captured by two-dimensional culture methods would arise by generating multinucleated muscle cells within a three-dimensional (3D) extracellular matrix environment. Herein we report methods to produce 3D human skeletal muscle microtissues (hMMTs) using clonal, immortalized myoblast lines established from healthy and DMD donors. We also established protocols to evaluate immortalized hMMT self-organization and myotube maturation, as well as calcium handling, force generation, membrane stability (i.e., creatine kinase activity and Evans blue dye permeability) and contractile apparatus organization following electrical-stimulation. In examining hMMTs generated with a cell line wherein the dystrophin gene possessed a duplication of exon 2, we observed rare dystrophin-positive myotubes, which were not seen in 2D cultures. Further, we show that treating DMD hMMTs with a β1-integrin activating antibody, improves contractile apparatus maturation and stability. Hence, immortalized myoblast-derived DMD hMMTs offer a pre-clinical system with which to investigate the potential of duplicated exon skipping strategies and those that protect muscle cells from contraction-induced injury. STATEMENT OF SIGNIFICANCE: Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disorder that is caused by mutation of the dystrophin gene. The biological basis of DMD pathology is only partially characterized and there is no cure for this fatal disease. Here we report a method to produce 3D human skeletal muscle microtissues (hMMTs) using immortalized human DMD and healthy myoblasts. Morphological and functional assessment revealed DMD-associated pathophysiology including impaired calcium handling and de novo formation of dystrophin-positive revertant muscle cells in immortalized DMD hMMTs harbouring an exon 2 duplication, a feature of many DMD patients that has not been recapitulated in culture prior to this report. We further demonstrate that this "DMD in a dish" system can be used as a pre-clinical assay to test a putative DMD therapeutic and study the mechanism of action.
Collapse
Affiliation(s)
- Majid Ebrahimi
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Heta Lad
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Aurora Fusto
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Yekaterina Tiper
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Asiman Datye
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Christine T Nguyen
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Erik Jacques
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Louise A Moyle
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Thy Nguyen
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Brennen Musgrave
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Carolina Chávez-Madero
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Anne Bigot
- Sorbonne Universite, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris UMRS974, France
| | - Chun Chen
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Scott Turner
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Bryan A Stewart
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Elena Pegoraro
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Libero Vitiello
- Department of Biology, University of Padua, Padua 35131, Italy; Interuniversity Institute of Myology (IIM), Italy
| | - Penney M Gilbert
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada.
| |
Collapse
|
22
|
Animal models for researching approaches to therapy of Duchenne muscular dystrophy. Transgenic Res 2021; 30:709-725. [PMID: 34409525 DOI: 10.1007/s11248-021-00278-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/11/2021] [Indexed: 01/17/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a relatively widespread genetic disease which develops as a result of a mutation in the gene DMD encoding dystrophin. In this review, animal models of DMD are described. These models are used in preclinical studies to elucidate the pathogenesis of the disease or to develop effective treatments; each animal model has its own advantages and disadvantages. For instance, Caenorhabditis elegans, Drosophila melanogaster, and zebrafish (sapje) are suitable for large-scale chemical screening of large numbers of small molecules, but their disease phenotype differs from that of mammals. The use of larger animals is important for understanding of the potential efficacy of various treatments for DMD. While mdx mice have their advantages, they exhibit a milder disease phenotype compared to humans or dogs, making it difficult to evaluate the efficacy of new treatment for DMD. The disease in dogs and pigs is more severe and progresses faster than in mice, but it is more difficult to breed and obtain sufficient numbers of specimens in order to achieve statistically significant results. Moreover, working with large animals is also more labor-intensive. Therefore, when choosing the optimal animal model for research, it is worth considering all the goals and objectives.
Collapse
|
23
|
Mareedu S, Million ED, Duan D, Babu GJ. Abnormal Calcium Handling in Duchenne Muscular Dystrophy: Mechanisms and Potential Therapies. Front Physiol 2021; 12:647010. [PMID: 33897454 PMCID: PMC8063049 DOI: 10.3389/fphys.2021.647010] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked muscle-wasting disease caused by the loss of dystrophin. DMD is associated with muscle degeneration, necrosis, inflammation, fatty replacement, and fibrosis, resulting in muscle weakness, respiratory and cardiac failure, and premature death. There is no curative treatment. Investigations on disease-causing mechanisms offer an opportunity to identify new therapeutic targets to treat DMD. An abnormal elevation of the intracellular calcium (Cai2+) concentration in the dystrophin-deficient muscle is a major secondary event, which contributes to disease progression in DMD. Emerging studies have suggested that targeting Ca2+-handling proteins and/or mechanisms could be a promising therapeutic strategy for DMD. Here, we provide an updated overview of the mechanistic roles the sarcolemma, sarcoplasmic/endoplasmic reticulum, and mitochondria play in the abnormal and sustained elevation of Cai2+ levels and their involvement in DMD pathogenesis. We also discuss current approaches aimed at restoring Ca2+ homeostasis as potential therapies for DMD.
Collapse
Affiliation(s)
- Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Emily D Million
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, United States.,Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, United States
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
24
|
Plasma membrane integrity in health and disease: significance and therapeutic potential. Cell Discov 2021; 7:4. [PMID: 33462191 PMCID: PMC7813858 DOI: 10.1038/s41421-020-00233-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Maintenance of plasma membrane integrity is essential for normal cell viability and function. Thus, robust membrane repair mechanisms have evolved to counteract the eminent threat of a torn plasma membrane. Different repair mechanisms and the bio-physical parameters required for efficient repair are now emerging from different research groups. However, less is known about when these mechanisms come into play. This review focuses on the existence of membrane disruptions and repair mechanisms in both physiological and pathological conditions, and across multiple cell types, albeit to different degrees. Fundamentally, irrespective of the source of membrane disruption, aberrant calcium influx is the common stimulus that activates the membrane repair response. Inadequate repair responses can tip the balance between physiology and pathology, highlighting the significance of plasma membrane integrity. For example, an over-activated repair response can promote cancer invasion, while the inability to efficiently repair membrane can drive neurodegeneration and muscular dystrophies. The interdisciplinary view explored here emphasises the widespread potential of targeting plasma membrane repair mechanisms for therapeutic purposes.
Collapse
|
25
|
Porcari P, Hall MG, Clark CA, Greally E, Straub V, Blamire AM. Time-dependent diffusion MRI as a probe of microstructural changes in a mouse model of Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2020; 33:e4276. [PMID: 32101354 DOI: 10.1002/nbm.4276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/05/2020] [Accepted: 01/30/2020] [Indexed: 06/10/2023]
Abstract
Dystrophic muscles show a high variability of fibre sizes and altered sarcolemmal integrity, which are typically assessed by histology. Time-dependent diffusion MRI is sensitive to tissue microstructure and its investigation through age-related changes in dystrophic and healthy muscles may help the understanding of the onset and progression of Duchenne muscular dystrophy (DMD). We investigated the capability of time-dependent diffusion MRI to quantify age and disease-related changes in hind-limb muscle microstructure between dystrophic (mdx) and wild-type (WT) mice of three age groups (7.5, 22 and 44 weeks). Diffusion time-dependent apparent diffusion coefficients (ADCs) of the gastrocnemius and tibialis anterior muscles were determined versus age and diffusion-gradient orientation at six diffusion times (Δ; range: 25-350 ms). Mean muscle ADCs were compared between groups and ages, and correlated with T2 , using Student's t test, one-way analysis of variance and Pearson correlation, respectively. Muscle fibre sizes and sarcolemmal integrity were evaluated by histology and compared with diffusion measurements. Hind-limb muscle ADC showed characteristic restricted diffusion behaviour in both mdx and WT animals with decreasing ADC values at longer Δ. Significant differences in ADC were observed at long Δ values (≥ 250 ms; p < 0.05, comparison between groups; p < 0.01, comparison between ages) with ADC increased by 5-15% in dystrophic muscles, indicative of reduced diffusion restriction. No significant correlation was found between T2 and ADC. Additionally, muscle fibre size distributions showed higher variability and lower mean fibre size in mdx than WT animals (p < 0.001). The extensive Evans Blue Dye uptake shown in dystrophic muscles revealed substantial sarcolemmal damage, suggesting diffusion measurements as more consistent with altered permeability rather than changes in muscle fibre sizes. This study shows the potential of diffusion MRI to non-invasively discriminate between dystrophic and healthy muscles with enhanced sensitivity when using long Δ.
Collapse
Affiliation(s)
- Paola Porcari
- Institute of Genetic Medicine and Centre for In Vivo Imaging, Newcastle University, Newcastle upon Tyne, UK
| | - Matt G Hall
- Developmental Imaging and Biophysics Section, UCL GOS Institute of Child Health, London, UK
| | - Chris A Clark
- Developmental Imaging and Biophysics Section, UCL GOS Institute of Child Health, London, UK
| | - Elizabeth Greally
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew M Blamire
- Institute of Cellular Medicine and Centre for In Vivo Imaging, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
26
|
Meyers TA, Heitzman JA, Townsend D. DMD carrier model with mosaic dystrophin expression in the heart reveals complex vulnerability to myocardial injury. Hum Mol Genet 2020; 29:944-954. [PMID: 31976522 PMCID: PMC7158376 DOI: 10.1093/hmg/ddaa015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/26/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating neuromuscular disease that causes progressive muscle wasting and cardiomyopathy. This X-linked disease results from mutations of the DMD allele on the X-chromosome resulting in the loss of expression of the protein dystrophin. Dystrophin loss causes cellular dysfunction that drives the loss of healthy skeletal muscle and cardiomyocytes. As gene therapy strategies strive toward dystrophin restoration through micro-dystrophin delivery or exon skipping, preclinical models have shown that incomplete restoration in the heart results in heterogeneous dystrophin expression throughout the myocardium. This outcome prompts the question of how much dystrophin restoration is sufficient to rescue the heart from DMD-related pathology. Female DMD carrier hearts can shed light on this question, due to their mosaic cardiac dystrophin expression resulting from random X-inactivation. In this work, a dystrophinopathy carrier mouse model was derived by breeding male or female dystrophin-null mdx mice with a wild type mate. We report that these carrier hearts are significantly susceptible to injury induced by one or multiple high doses of isoproterenol, despite expressing ~57% dystrophin. Importantly, only carrier mice with dystrophic mothers showed mortality after isoproterenol. These findings indicate that dystrophin restoration in approximately half of the heart still allows for marked vulnerability to injury. Additionally, the discovery of divergent stress-induced mortality based on parental origin in mice with equivalent dystrophin expression underscores the need for better understanding of the epigenetic, developmental, and even environmental factors that may modulate vulnerability in the dystrophic heart.
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jackie A Heitzman
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
27
|
Choi JH, Jeong SY, Oh MR, Allen PD, Lee EH. TRPCs: Influential Mediators in Skeletal Muscle. Cells 2020; 9:cells9040850. [PMID: 32244622 PMCID: PMC7226745 DOI: 10.3390/cells9040850] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ itself or Ca2+-dependent signaling pathways play fundamental roles in various cellular processes from cell growth to death. The most representative example can be found in skeletal muscle cells where a well-timed and adequate supply of Ca2+ is required for coordinated Ca2+-dependent skeletal muscle functions, such as the interactions of contractile proteins during contraction. Intracellular Ca2+ movements between the cytosol and sarcoplasmic reticulum (SR) are strictly regulated to maintain the appropriate Ca2+ supply in skeletal muscle cells. Added to intracellular Ca2+ movements, the contribution of extracellular Ca2+ entry to skeletal muscle functions and its significance have been continuously studied since the early 1990s. Here, studies on the roles of channel proteins that mediate extracellular Ca2+ entry into skeletal muscle cells using skeletal myoblasts, myotubes, fibers, tissue, or skeletal muscle-originated cell lines are reviewed with special attention to the proposed functions of transient receptor potential canonical proteins (TRPCs) as store-operated Ca2+ entry (SOCE) channels under normal conditions and the potential abnormal properties of TRPCs in muscle diseases such as Duchenne muscular dystrophy (DMD).
Collapse
Affiliation(s)
- Jun Hee Choi
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Yeon Jeong
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Mi Ri Oh
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Paul D. Allen
- Leeds Institute of Biomedical & Clinical Sciences, St. James’s University Hospital, University of Leeds, Leeds LS97TF, UK
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-7279
| |
Collapse
|
28
|
Lopez JR, Uryash A, Faury G, Estève E, Adams JA. Contribution of TRPC Channels to Intracellular Ca 2 + Dyshomeostasis in Smooth Muscle From mdx Mice. Front Physiol 2020; 11:126. [PMID: 32153426 PMCID: PMC7044154 DOI: 10.3389/fphys.2020.00126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an irreversible muscle disease characterized by a progressive loss of muscle function, decreased ambulation, and ultimately death as a result of cardiac or respiratory failure. DMD is caused by the lack of dystrophin, a protein that is important for membrane stability and signaling in excitable cells. Although vascular smooth muscle cells (VSMCs) dysfunction occurs in many pathological conditions, little is known about vascular smooth muscle function in DMD. We have previously shown that striated muscle cells, as well as neurons isolated from dystrophic (mdx) mice have higher intracellular Ca2+ ([Ca2+]i) and Na+ ([Na+]i) concentrations and decreased cell viability in comparison with wild type (Wt). Experiments were carried out in isolated VSMCs from mdx (a murine model of DMD) and congenic C57BL/10SnJ Wt mice. We found elevated [Ca2+]i and [Na+]i in VSMCs from mdx mice compared to Wt. Exposure to 1-oleoyl-2-acetyl-sn-glycerol (OAG), a TRPC3 and TRPC6 channel activator, induced a greater elevation of [Ca2+]i and [Na+]i in mdx than Wt VSMCs. The OAG induced increases in [Ca2+]i could be abolished by either removal of extracellular Ca2+ or by SAR7334, a blocker of TRPC3 and TRPC 6 channels in both genotypes. Mdx and Wt VSMCs were susceptible to muscle cell stretch-induced elevations of [Ca2+]i and [Na+]i which was completely inhibited by GsMTx-4, a mechanosensitive ion channel inhibitor. Western blots showed a significant upregulation of TRPC1 -3, -6 proteins in mdx VSMCs compare to age-matched Wt. The lack of dystrophin in mdx VSMCs produced a profound alteration of [Ca2+]i and [Na+]i homeostasis that appears to be mediated by TRPC channels. Moreover, we have been able to demonstrate pharmacologically that the enhanced stretch-induced elevation of intracellular [Ca2+] and concomitant cell damage in mdx VSMCs also appears to be mediated through TRPC1, -3 and -6 channel activation.
Collapse
Affiliation(s)
- Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| | - Arkady Uryash
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - Gilles Faury
- HP2, CHU Grenoble Alpes, Inserm, University Grenoble Alpes, Grenoble, France
| | - Eric Estève
- HP2, CHU Grenoble Alpes, Inserm, University Grenoble Alpes, Grenoble, France
| | - Jose A Adams
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| |
Collapse
|
29
|
Segawa K, Sugawara N, Maruo K, Kimura K, Komaki H, Takahashi Y, Sasaki M. Left Ventricular End-Diastolic Diameter and Cardiac Mortality in Duchenne Muscular Dystrophy. Neuropsychiatr Dis Treat 2020; 16:171-178. [PMID: 32021209 PMCID: PMC6972578 DOI: 10.2147/ndt.s235166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/07/2019] [Indexed: 01/16/2023] Open
Abstract
PURPOSE This study aimed to examine weather left ventricular end-diastolic diameter (LVDd) could predict mortality from heart failure in patients with Duchenne muscular dystrophy (DMD) receiving standard cardio-protective therapies. PATIENTS AND METHODS One hundred thirty-three patients with DMD aged ≥10 years who underwent echocardiography from 2011 to 2015 were included in this study and retrospectively followed until August 2018. Patients were divided into two groups according to LVDd at initial echocardiography: ≤ 54 mm (Group 1: n=119) and ≥ 55 mm (Group 2: n=14). To identify factors other than LVDd that may affect heart failure-related mortality, Group 2 patients who developed no left atrial (LA) enlargement, moderate mitral regurgitation (MR), or pulmonary hypertension (PH) during the observation period (Group 2A: n=5) were compared with those who newly developed one or more of those complications (Group 2B: n=7). Clinical outcomes were all-cause mortality and mortality from heart failure. RESULTS Mean observation period was 5.5±1.5 years in Group 1 and 4.4±1.9 years in Group 2. A total of 14 patients (10.5%) died, including 6 of 119 (5.0%) patients in Group 1 and 8 of 14 (57.1%) patients in Group 2 (p<0.001). Among these, 1 (0.8%) patient in Group 1 and 8 (57.1%) patients in Group 2 died from heart failure (p<0.001). Group 2B patients had shorter survival compared to Group 2A patients (p=0.006). CONCLUSION LVDd ≥ 55 mm is a predictive factor for mortality from heart failure in patients with DMD. Complications including LA enlargement, moderate MR, and PH were found to be predictive factors for mortality from heart failure in a short period.
Collapse
Affiliation(s)
- Kazuhiko Segawa
- Department of Cardiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Norio Sugawara
- Department of Psychiatry, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Kazushi Maruo
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Koichi Kimura
- Department of General Medicine, The Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Hirofumi Komaki
- Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuji Takahashi
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masayuki Sasaki
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
30
|
Henríquez-Olguín C, Boronat S, Cabello-Verrugio C, Jaimovich E, Hidalgo E, Jensen TE. The Emerging Roles of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 in Skeletal Muscle Redox Signaling and Metabolism. Antioxid Redox Signal 2019; 31:1371-1410. [PMID: 31588777 PMCID: PMC6859696 DOI: 10.1089/ars.2018.7678] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Skeletal muscle is a crucial tissue to whole-body locomotion and metabolic health. Reactive oxygen species (ROS) have emerged as intracellular messengers participating in both physiological and pathological adaptations in skeletal muscle. A complex interplay between ROS-producing enzymes and antioxidant networks exists in different subcellular compartments of mature skeletal muscle. Recent evidence suggests that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a major source of contraction- and insulin-stimulated oxidants production, but they may paradoxically also contribute to muscle insulin resistance and atrophy. Recent Advances: Pharmacological and molecular biological tools, including redox-sensitive probes and transgenic mouse models, have generated novel insights into compartmentalized redox signaling and suggested that NOX2 contributes to redox control of skeletal muscle metabolism. Critical Issues: Major outstanding questions in skeletal muscle include where NOX2 activation occurs under different conditions in health and disease, how NOX2 activation is regulated, how superoxide/hydrogen peroxide generated by NOX2 reaches the cytosol, what the signaling mediators are downstream of NOX2, and the role of NOX2 for different physiological and pathophysiological processes. Future Directions: Future research should utilize and expand the current redox-signaling toolbox to clarify the NOX2-dependent mechanisms in skeletal muscle and determine whether the proposed functions of NOX2 in cells and animal models are conserved into humans.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Susanna Boronat
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Reyngoudt H, Lopez Kolkovsky AL, Carlier PG. Free intramuscular Mg 2+ concentration calculated using both 31 P and 1 H NMRS-based pH in the skeletal muscle of Duchenne muscular dystrophy patients. NMR IN BIOMEDICINE 2019; 32:e4115. [PMID: 31184793 DOI: 10.1002/nbm.4115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 06/09/2023]
Abstract
Early studies have demonstrated that (total) magnesium was decreased in skeletal muscle of Duchenne muscular dystrophy (DMD) patients. Free intramuscular Mg2+ can be derived from 31 P NMRS measurements. The value of free intramuscular magnesium concentration ([Mg2+ ]) is highly dependent on precise knowledge of intracellular pH, which is abnormally alkaline in dystrophic muscle, possibly due to an expanded interstitial space, potentially causing an underestimation of [Mg2+ ]. We have recently shown that intracellular pH can be derived using 1 H NMRS of carnosine. Our aim was to determine whether 31 P NMRS-based [Mg2+ ] is, in fact, abnormally low in DMD patients, taking advantage of the 1 H NMRS-based pH. A comparative analysis was, therefore, made between [Mg2+ ] values calculated with both 1 H and 31 P NMRS-based approaches to determine pH in 25 DMD patients, on a 3-T clinical NMR scanner. [Mg2+ ] was also assessed with 31 P NMRS only in (forearm or leg) skeletal muscle of 60 DMD patients and 63 age-matched controls. Additionally, phosphodiester levels as well as quantitative NMRI indices including water T2 , fat fraction, contractile cross-sectional area and one-year changes were evaluated. The main finding was that the significant difference in [Mg2+ ] between DMD patients and controls was preserved even when the intracellular pH determined with 1 H NMRS was similar in both groups. Consequently, we observed that [Mg2+ ] is significantly lower in DMD patients compared with controls in the larger database where only 31 P NMRS data were obtained. Significant yet weak correlations existed between [Mg2+ ] and PDE, water T2 and fat fraction. We concluded that low [Mg2+ ] is an actual finding in DMD, whether intracellular pH is normal or alkaline, and that it is a likely consequence of membrane leakiness. The response of Mg2+ to therapeutic treatment remains to be investigated in neuromuscular disorders. Free [Mg2+ ] determination with 31 P NMRS is highly dependent on a precise knowledge of intracellular pH. The pH of Duchenne muscular dystrophy (DMD) patients, as determined by 31 P NMRS, is abnormally alkaline. We have recently shown that intracellular pH could be determined using 1 H NMRS of carnosine, and that intracellular pH was alkaline in a proportion of, but not all, DMD patients with a 31 P NMRS-based alkaline pH. Taking advantage of this 1 H NMRS-based intracellular pH, we found that free intramuscular [Mg2+ ] is in fact abnormally low in DMD patients.
Collapse
Affiliation(s)
- Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
- NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Alfredo L Lopez Kolkovsky
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
- NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Pierre G Carlier
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
- NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| |
Collapse
|
32
|
Meyers TA, Townsend D. Cardiac Pathophysiology and the Future of Cardiac Therapies in Duchenne Muscular Dystrophy. Int J Mol Sci 2019; 20:E4098. [PMID: 31443395 PMCID: PMC6747383 DOI: 10.3390/ijms20174098] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/12/2019] [Accepted: 08/19/2019] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease featuring skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. Historically, respiratory failure has been the leading cause of mortality in DMD, but recent improvements in symptomatic respiratory management have extended the life expectancy of DMD patients. With increased longevity, the clinical relevance of heart disease in DMD is growing, as virtually all DMD patients over 18 year of age display signs of cardiomyopathy. This review will focus on the pathophysiological basis of DMD in the heart and discuss the therapeutic approaches currently in use and those in development to treat dystrophic cardiomyopathy. The first section will describe the aspects of the DMD that result in the loss of cardiac tissue and accumulation of fibrosis. The second section will discuss cardiac small molecule therapies currently used to treat heart disease in DMD, with a focus on the evidence supporting the use of each drug in dystrophic patients. The final section will outline the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, or repair. There are several new and promising therapeutic approaches that may protect the dystrophic heart, but their limitations suggest that future management of dystrophic cardiomyopathy may benefit from combining gene-targeted therapies with small molecule therapies. Understanding the mechanistic basis of dystrophic heart disease and the effects of current and emerging therapies will be critical for their success in the treatment of patients with DMD.
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
33
|
Niranjan N, Mareedu S, Tian Y, Kodippili K, Fefelova N, Voit A, Xie LH, Duan D, Babu GJ. Sarcolipin overexpression impairs myogenic differentiation in Duchenne muscular dystrophy. Am J Physiol Cell Physiol 2019; 317:C813-C824. [PMID: 31365291 DOI: 10.1152/ajpcell.00146.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Reduction in the expression of sarcolipin (SLN), an inhibitor of sarco(endo)plasmic reticulum (SR) Ca2+-ATPase (SERCA), ameliorates severe muscular dystrophy in mice. However, the mechanism by which SLN inhibition improves muscle structure remains unclear. Here, we describe the previously unknown function of SLN in muscle differentiation in Duchenne muscular dystrophy (DMD). Overexpression of SLN in C2C12 resulted in decreased SERCA pump activity, reduced SR Ca2+ load, and increased intracellular Ca2+ (Cai2+) concentration. In addition, SLN overexpression resulted in altered expression of myogenic markers and poor myogenic differentiation. In dystrophin-deficient dog myoblasts and myotubes, SLN expression was significantly high and associated with defective Cai2+ cycling. The dystrophic dog myotubes were less branched and associated with decreased autophagy and increased expression of mitochondrial fusion and fission proteins. Reduction in SLN expression restored these changes and enhanced dystrophic dog myoblast fusion during differentiation. In summary, our data suggest that SLN upregulation is an intrinsic secondary change in dystrophin-deficient myoblasts and could account for the Cai2+ mishandling, which subsequently contributes to poor myogenic differentiation. Accordingly, reducing SLN expression can improve the Cai2+ cycling and differentiation of dystrophic myoblasts. These findings provide cellular-level supports for targeting SLN expression as a therapeutic strategy for DMD.
Collapse
Affiliation(s)
- Nandita Niranjan
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Yimin Tian
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Kasun Kodippili
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Antanina Voit
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri.,Department of Neurology, University of Missouri, Columbia, Missouri.,Department of Biomedical, Biological & Chemical Engineering, University of Missouri, Columbia, Missouri.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
34
|
Gaglianone RB, Bloise FF, Ortiga-Carvalho TM, Quirico-Santos T, Costa ML, Mermelstein C. Comparative study of calcium and calcium-related enzymes with differentiation markers in different ages and muscle types in mdx mice. Histol Histopathol 2019; 35:203-216. [PMID: 31274171 DOI: 10.14670/hh-18-145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sarcolemma instability and increased calcium influx in muscle fibers are characteristics of the Duchenne muscular dystrophy. Excessive calcium activates calcium-dependent enzymes, such as calpains (CAPN) and matrix metalloproteases (MMP). Here, we analyzed calcium deposits, the activity of CAPN and MMP and the expression of Myh, SERCA and myogenic regulatory factors in different skeletal muscles during myonecrosis (4-weeks) and regeneration (12-weeks) phases of the mdx muscular pathology. Alizarin red staining was used to assess calcium deposits, casein and gelatin zymography were performed to evaluate CAPN and MMP activity, and qPCR was used to evaluate the expression of Myh, Capn, Atp2a1 and Atp2a2, Myod1 and Myog. We observed the following characteristics in mdx muscles: (i) calcium deposits almost exclusively in mdx muscles, (ii) lower CAPN1 activity in mdx muscles, (iii) higher CAPN2 activity in mdx muscles (only at 12 wks), (iv) autolyzed CAPN activity exclusively in mdx muscles, (v) lower expression of Capn1 and higher expression of Capn2 in mdx muscles; (vi) lower expression of Atp2a1 and Atp2a2 in mdx muscles, (vii) higher MMP (pre pro MMP2, pro MMP2, MMP2 and MMP9) activity in mdx muscles, (viii) MMP2 activity exclusively in mdx muscles at 12 wks, (ix) MMP9 activity exclusively in mdx muscles, (x) higher expression of Myog in mdx muscles at 12 wks, and (xi) lower expression of Myh (Myh7, Myh2, Myh1, Myh4) in mdx muscles, particularly Myh7 and Myh2. The collection of our results provides valuable information for a better characterization of mdx pathology phenotype.
Collapse
Affiliation(s)
- Rhayanna B Gaglianone
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Biology Institute, Fluminense Federal University, Niterói, RJ, Brazil
| | - Flavia Fonseca Bloise
- Carlos Chagas Filho Biophysical Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | - Manoel Luis Costa
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Claudia Mermelstein
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
35
|
Meyers TA, Heitzman JA, Krebsbach AM, Aufdembrink LM, Hughes R, Bartolomucci A, Townsend D. Acute AT 1R blockade prevents isoproterenol-induced injury in mdx hearts. J Mol Cell Cardiol 2019; 128:51-61. [PMID: 30664850 DOI: 10.1016/j.yjmcc.2019.01.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/31/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked disease characterized by skeletal muscle degeneration and a significant cardiomyopathy secondary to cardiomyocyte damage and myocardial loss. The molecular basis of DMD lies in the absence of the protein dystrophin, which plays critical roles in mechanical membrane integrity and protein localization at the sarcolemma. A popular mouse model of DMD is the mdx mouse, which lacks dystrophin and displays mild cardiac and skeletal pathology that can be exacerbated to advance the disease state. In clinical and pre-clinical studies of DMD, angiotensin signaling pathways have emerged as therapeutic targets due to their adverse influence on muscle remodeling and oxidative stress. Here we aim to establish a physiologically relevant cardiac injury model in the mdx mouse, and determine whether acute blockade of the angiotensin II type 1 receptor (AT1R) may be utilized for prevention of dystrophic injury. METHODS AND RESULTS A single IP injection of isoproterenol (Iso, 10 mg/kg) was used to induce cardiac stress and injury in mdx and wild type (C57Bl/10) mice. Mice were euthanized 8 h, 30 h, 1 week, or 1 month following the injection, and hearts were harvested for injury evaluation. At 8 and 30 h post-injury, mdx hearts showed 2.2-fold greater serum cTnI content and 3-fold more extensive injury than wild type hearts. Analysis of hearts 1 week and 1 month after injury revealed significantly higher fibrosis in mdx hearts, with a more robust and longer-lasting immune response compared to wild type hearts. In the 30-hour group, losartan treatment initiated 1 h before Iso injection protected dystrophic hearts from cardiac damage, reducing mdx acute injury area by 2.8-fold, without any significant effect on injury in wild type hearts. However, both wild type and dystrophic hearts showed a 2-fold reduction in the magnitude of the macrophage response to injury 30 h after Iso with losartan. CONCLUSIONS This work demonstrates that acute blockade of AT1R has the potential for robust injury prevention in a model of Iso-induced dystrophic heart injury. In addition to selectively limiting dystrophic cardiac damage, blocking AT1R may serve to limit the inflammatory nature of the immune response to injury in all hearts. Our findings strongly suggest that earlier adoption of angiotensin receptor blockers in DMD patients could limit myocardial damage and subsequent cardiomyopathy.
Collapse
MESH Headings
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Cardiomyopathies/drug therapy
- Cardiomyopathies/genetics
- Cardiomyopathies/pathology
- Dystrophin/genetics
- Heart/drug effects
- Heart/physiopathology
- Humans
- Isoproterenol/pharmacology
- Losartan/pharmacology
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Receptor, Angiotensin, Type 1/genetics
- Sarcolemma/metabolism
- Sarcolemma/pathology
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jackie A Heitzman
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Aimee M Krebsbach
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Lauren M Aufdembrink
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Robert Hughes
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
36
|
Houang EM, Sham YY, Bates FS, Metzger JM. Muscle membrane integrity in Duchenne muscular dystrophy: recent advances in copolymer-based muscle membrane stabilizers. Skelet Muscle 2018; 8:31. [PMID: 30305165 PMCID: PMC6180502 DOI: 10.1186/s13395-018-0177-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023] Open
Abstract
The scientific premise, design, and structure-function analysis of chemical-based muscle membrane stabilizing block copolymers are reviewed here for applications in striated muscle membrane injury. Synthetic block copolymers have a rich history and wide array of applications from industry to biology. Potential for discovery is enabled by a large chemical space for block copolymers, including modifications in block copolymer mass, composition, and molecular architecture. Collectively, this presents an impressive chemical landscape to leverage distinct structure-function outcomes. Of particular relevance to biology and medicine, stabilization of damaged phospholipid membranes using amphiphilic block copolymers, classified as poloxamers or pluronics, has been the subject of increasing scientific inquiry. This review focuses on implementing block copolymers to protect fragile muscle membranes against mechanical stress. The review highlights interventions in Duchenne muscular dystrophy, a fatal disease of progressive muscle deterioration owing to marked instability of the striated muscle membrane. Biophysical and chemical engineering advances are presented that delineate and expand upon current understanding of copolymer-lipid membrane interactions and the mechanism of stabilization. The studies presented here serve to underscore the utility of copolymer discovery leading toward the therapeutic application of block copolymers in Duchenne muscular dystrophy and potentially other biomedical applications in which membrane integrity is compromised.
Collapse
Affiliation(s)
- Evelyne M. Houang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| | - Yuk Y. Sham
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
- University of Minnesota Informatics Institute, MN, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, MN, USA
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, MN, USA
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| |
Collapse
|
37
|
Morgan JE, Prola A, Mariot V, Pini V, Meng J, Hourde C, Dumonceaux J, Conti F, Relaix F, Authier FJ, Tiret L, Muntoni F, Bencze M. Necroptosis mediates myofibre death in dystrophin-deficient mice. Nat Commun 2018; 9:3655. [PMID: 30194302 PMCID: PMC6128848 DOI: 10.1038/s41467-018-06057-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 08/10/2018] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe degenerative disorder caused by mutations in the dystrophin gene. Dystrophin-deficient muscles are characterised by progressive myofibre necrosis in which inflammation plays a deleterious role. However, the molecular mechanisms underlying inflammation-induced necrosis in muscle cells are unknown. Here we show that necroptosis is a mechanism underlying myofibre death in dystrophin-deficient muscle. RIPK1, RIPK3 and MLKL are upregulated in dystrophic mouse myofibres. In human DMD samples, there is strong immunoreactivity to RIPK3 and phospho-MLKL in myofibres. In vitro, TNFα can elicit necroptosis in C2C12 myoblasts, and RIPK3 overexpression sensitises myoblasts to undergo TNF-induced death. Furthermore, genetic ablation of Ripk3 in mdx mice reduces myofibre degeneration, inflammatory infiltrate, and muscle fibrosis, and eventually improves muscle function. These findings provide the first evidence of necroptotic cell death in a disease affecting skeletal muscle and identify RIPK3 as a key player in the degenerative process in dystrophin-deficient muscles.
Collapse
Affiliation(s)
- Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK.
| | - Alexandre Prola
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France
| | - Virginie Mariot
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, 30 Guilford Street, London, WC1N 1EH, UK
| | - Veronica Pini
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Jinhong Meng
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Christophe Hourde
- Inter-University Laboratory of Human Movement Biology (LIBM)-EA7424, Université Savoie Mont Blanc, Campus Scientifique Technolac, 73376, Le Bourget du Lac Cedex, France
| | - Julie Dumonceaux
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, 30 Guilford Street, London, WC1N 1EH, UK
| | - Francesco Conti
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Frederic Relaix
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France.,Nord/Est/Ile-de-France Reference Centre for Neuromuscular Diseases, Henri Mondor University Hospital (APHP), 94000, Créteil, France
| | - Francois-Jerôme Authier
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France.,Nord/Est/Ile-de-France Reference Centre for Neuromuscular Diseases, Henri Mondor University Hospital (APHP), 94000, Créteil, France
| | - Laurent Tiret
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Maximilien Bencze
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK. .,U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France.
| |
Collapse
|
38
|
Coenen-Stass AML, Sork H, Gatto S, Godfrey C, Bhomra A, Krjutškov K, Hart JR, Westholm JO, O'Donovan L, Roos A, Lochmüller H, Puri PL, El Andaloussi S, Wood MJA, Roberts TC. Comprehensive RNA-Sequencing Analysis in Serum and Muscle Reveals Novel Small RNA Signatures with Biomarker Potential for DMD. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 13:1-15. [PMID: 30219269 PMCID: PMC6140421 DOI: 10.1016/j.omtn.2018.08.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 01/03/2023]
Abstract
Extracellular small RNAs (sRNAs), including microRNAs (miRNAs), are promising biomarkers for diseases such as Duchenne muscular dystrophy (DMD), although their biological relevance is largely unknown. To investigate the relationship between intracellular and extracellular sRNA levels on a global scale, we performed sRNA sequencing in four muscle types and serum from wild-type, dystrophic mdx, and mdx mice in which dystrophin protein expression was restored by exon skipping. Differentially abundant sRNAs were identified in serum (mapping to miRNA, small nuclear RNA [snRNA], and PIWI-interacting RNA [piRNA] loci). One novel candidate biomarker, miR-483, was increased in both mdx serum and muscle, and also elevated in DMD patient sera. Dystrophin restoration induced global shifts in miRNA (including miR-483) and snRNA-fragment abundance toward wild-type levels. Specific serum piRNA-like sRNAs also responded to exon skipping therapy. Absolute miRNA expression in muscle was positively correlated with abundance in the circulation, although multiple highly expressed miRNAs in muscle were not elevated in mdx serum, suggesting that both passive and selective release mechanisms contribute to serum miRNA levels. In conclusion, this study has revealed new insights into the sRNA biology of dystrophin deficiency and identified novel DMD biomarkers.
Collapse
Affiliation(s)
- Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Helena Sork
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge 141 86, Sweden
| | - Sole Gatto
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Caroline Godfrey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Amarjit Bhomra
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Kaarel Krjutškov
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, Huddinge 141 83, Sweden; Competence Centre on Health Technologies, Tartu 50410, Estonia
| | - Jonathan R Hart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jakub O Westholm
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 17121 Solna, Sweden
| | - Liz O'Donovan
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Andreas Roos
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK; Biomedical Research Department, Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V., Otto-Hahn-Strasse 6b, 44227 Dortmund, Germany
| | - Hanns Lochmüller
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK; Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany; Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK; Department of Laboratory Medicine, Karolinska Institutet, Huddinge 141 86, Sweden
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK.
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK; Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
39
|
Nemes R, Koltai E, Taylor AW, Suzuki K, Gyori F, Radak Z. Reactive Oxygen and Nitrogen Species Regulate Key Metabolic, Anabolic, and Catabolic Pathways in Skeletal Muscle. Antioxidants (Basel) 2018; 7:antiox7070085. [PMID: 29976853 PMCID: PMC6071245 DOI: 10.3390/antiox7070085] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/15/2018] [Accepted: 06/29/2018] [Indexed: 12/29/2022] Open
Abstract
Reactive oxygen and nitrogen species (RONS) are important cellular regulators of key physiological processes in skeletal muscle. In this review, we explain how RONS regulate muscle contraction and signaling, and why they are important for membrane remodeling, protein turnover, gene expression, and epigenetic adaptation. We discuss how RONS regulate carbohydrate uptake and metabolism of skeletal muscle, and how they indirectly regulate fat metabolism through silent mating type information regulation 2 homolog 3 (SIRT3). RONS are causative/associative signaling molecules, which cause sarcopenia or muscle hypertrophy. Regular exercise influences redox biology, metabolism, and anabolic/catabolic pathways in skeletal muscle in an intensity dependent manner.
Collapse
Affiliation(s)
- Roland Nemes
- Faculty of Sports and Health Studies, Hosei University, Tokyo 194-0298, Japan.
| | - Erika Koltai
- Research Institute of Sport Science, University of Physical Education, Alkotas u. 44, H-1123 Budapest, Hungary.
| | - Albert W Taylor
- Faculty of Health Sciences, The University of Western Ontario, London, ON N6G 1H1, Canada.
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Saitama 359-1192, Japan.
| | - Ferenc Gyori
- Institute of Sport Science, University of Szeged, H-6726 Szeged, Hungary.
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, Alkotas u. 44, H-1123 Budapest, Hungary.
- Institute of Sport Science, University of Szeged, H-6726 Szeged, Hungary.
| |
Collapse
|
40
|
Allard B. From excitation to intracellular Ca 2+ movements in skeletal muscle: Basic aspects and related clinical disorders. Neuromuscul Disord 2018; 28:394-401. [DOI: 10.1016/j.nmd.2018.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/19/2018] [Accepted: 03/05/2018] [Indexed: 01/18/2023]
|
41
|
Cordova G, Negroni E, Cabello-Verrugio C, Mouly V, Trollet C. Combined Therapies for Duchenne Muscular Dystrophy to Optimize Treatment Efficacy. Front Genet 2018; 9:114. [PMID: 29692797 PMCID: PMC5902687 DOI: 10.3389/fgene.2018.00114] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/22/2018] [Indexed: 01/01/2023] Open
Abstract
Duchene Muscular Dystrophy (DMD) is the most frequent muscular dystrophy and one of the most severe due to the absence of the dystrophin protein. Typical pathological features include muscle weakness, muscle wasting, degeneration, and inflammation. At advanced stages DMD muscles present exacerbated extracellular matrix and fat accumulation. Recent progress in therapeutic approaches has allowed new strategies to be investigated, including pharmacological, gene-based and cell-based therapies. Gene and cell-based therapies are still limited by poor targeting and low efficiency in fibrotic dystrophic muscle, therefore it is increasingly evident that future treatments will have to include “combined therapies” to reach maximal efficiency. The scope of this mini-review is to provide an overview of the current literature on such combined therapies for DMD. By “combined therapies” we mean those that include both a therapy to correct the genetic defect and an additional one to address one of the secondary pathological features of the disease. In this mini-review, we will not provide a comprehensive view of the literature on therapies for DMD, since many such reviews already exist, but we will focus on the characteristics, efficiency, and potential of such combined therapeutic strategies that have been described so far for DMD.
Collapse
Affiliation(s)
- Gonzalo Cordova
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Elisa Negroni
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Claudio Cabello-Verrugio
- Laboratorio de Patologías Musculares, Fragilidad y Envejecimiento, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Vincent Mouly
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Capucine Trollet
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| |
Collapse
|
42
|
Salmaninejad A, Valilou SF, Bayat H, Ebadi N, Daraei A, Yousefi M, Nesaei A, Mojarrad M. Duchenne muscular dystrophy: an updated review of common available therapies. Int J Neurosci 2018; 128:854-864. [PMID: 29351004 DOI: 10.1080/00207454.2018.1430694] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND PURPOSE Duchenne muscular dystrophy (DMD) is a lethal progressive pediatric muscle disorder and genetically inherited as an X-linked disease that caused by mutations in the dystrophin gene. DMD leads to progressive muscle weakness, degeneration, and wasting; finally, follows with the premature demise in affected individuals due to respiratory and/or cardiac failure typically by age of 30. For decades, scientists tried massively to find an effective therapy method, but there is no absolute cure currently for patients with DMD, nevertheless, recent advanced progressions on the treatment of DMD will be hopeful in the future. Several promising gene therapies are currently under investigation. These include gene replacement, exon skipping, suppression of stop codons. More recently, a promising gene editing tool referred to as CRISPR/Cas9 offers exciting perspectives for restoring dystrophin expression in patients with DMD. This review intents to briefly describe these methods and comment on their advances. Since DMD is a genetic disorder, it should be treated by replacing the deficient DMD copy with a functional one. However, there are different types of mutations in this gene, so such therapeutic approaches are highly mutation specific and thus are personalized. Therefore, DMD has arisen as a model of genetic disorder for understanding and overcoming of the challenges of developing personalized genetic medicines, consequently, the lessons learned from these approaches will be applicable to many other disorders. CONCLUSIONS This review provides an update on the recent gene therapies for DMD that aim to compensate for dystrophin deficiency and the related clinical trials.
Collapse
Affiliation(s)
- Arash Salmaninejad
- a Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee, Department of Medical Genetics, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran.,c Medical Genetics Research Center, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Saeed Farajzadeh Valilou
- d Medical Genetics Network (MeGeNe) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Hadi Bayat
- e Department of Tissue Engineering, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Nader Ebadi
- f Department of Medical Genetics, Faculty of Medicine , Tehran University of Medical Science , Tehran , Iran
| | - Abdolreza Daraei
- g Genetic Department, Faculty of Medicine , Babol University of Medical Sciences , Babol , Iran
| | - Meysam Yousefi
- b Student Research Committee, Department of Medical Genetics, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran.,c Medical Genetics Research Center, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Abolfazl Nesaei
- h Department of Basic Sciences, Faculty of Medicine , Gonabad University of Medical Sciences , Gonabad , Iran
| | - Majid Mojarrad
- b Student Research Committee, Department of Medical Genetics, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran.,c Medical Genetics Research Center, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
43
|
Reyngoudt H, Turk S, Carlier PG. 1 H NMRS of carnosine combined with 31 P NMRS to better characterize skeletal muscle pH dysregulation in Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2018; 31:e3839. [PMID: 29130550 DOI: 10.1002/nbm.3839] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 05/18/2023]
Abstract
In recent years, quantitative nuclear magnetic resonance imaging and spectroscopy (NMRI and NMRS) have been used more systematically as outcome measures in natural history and clinical trial studies for Duchenne muscular dystrophy (DMD). Whereas most of these studies have emphasized the evaluation of the fat fraction as an assessment for disease severity, less focus has been placed on metabolic indices measured by NMRS. 31 P NMRS in DMD reveals an alkaline inorganic phosphate (Pi ) pool, originating from either leaky dystrophic myocytes or an increased interstitial space. 1 H NMRS, exploiting the pH-sensitive proton resonances of carnosine, an intracellular dipeptide, was used to distinguish between these two hypotheses. NMR data were obtained in 23 patients with DMD and 14 healthy subjects on a 3-T clinical NMR system. Both 31 P and 1 H NMRS data were acquired at the level of the gastrocnemius medialis muscle. A multi-slice multi-echo imaging acquisition was performed for the determination of water T2 and fat fraction in the same region of interest. Whereas nearly all patients with DMD showed an elevated pH compared with healthy controls when using 31 P NMRS, 1 H NMRS-determined pH was not systematically increased. As expected, the carnosine-based intracellular pH was never found to be alkaline in the absence of a concurrent Pi -based pH elevation. In addition, abnormal intracellular pH, based on carnosine, was never associated with normal water T2 values. We conclude that, in one group of patients, both 1 H and 31 P NMRS showed an alkaline pH, originating from the intracellular compartment and reflecting ionic dysregulation in dystrophic myocytes. In the other patients with DMD, intracellular pH was normal, but an alkaline Pi pool was still present, suggesting an extracellular origin, probably revealing an expanded interstitial volume fraction, often associated with fibrotic changes. The data demonstrate that 1 H NMRS could serve as a biomarker to assess the normalization of intramyocytic pH and sarcolemmal permeability following therapy inducing dystrophin expression in patients with DMD.
Collapse
Affiliation(s)
- Harmen Reyngoudt
- NMR Laboratory, Institute of Myology, Paris, France
- CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Suna Turk
- NMR Laboratory, Institute of Myology, Paris, France
- CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Pierre G Carlier
- NMR Laboratory, Institute of Myology, Paris, France
- CEA, DRF, IBFJ, MIRCen, Paris, France
| |
Collapse
|
44
|
Tempol Supplementation Restores Diaphragm Force and Metabolic Enzyme Activities in mdx Mice. Antioxidants (Basel) 2017; 6:antiox6040101. [PMID: 29210997 PMCID: PMC5745511 DOI: 10.3390/antiox6040101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/19/2017] [Accepted: 11/28/2017] [Indexed: 12/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by striated muscle weakness, cardiomyopathy, and respiratory failure. Since oxidative stress is recognized as a secondary pathology in DMD, the efficacy of antioxidant intervention, using the superoxide scavenger tempol, was examined on functional and biochemical status of dystrophin-deficient diaphragm muscle. Diaphragm muscle function was assessed, ex vivo, in adult male wild-type and dystrophin-deficient mdx mice, with and without a 14-day antioxidant intervention. The enzymatic activities of muscle citrate synthase, phosphofructokinase, and lactate dehydrogenase were assessed using spectrophotometric assays. Dystrophic diaphragm displayed mechanical dysfunction and altered biochemical status. Chronic tempol supplementation in the drinking water increased diaphragm functional capacity and citrate synthase and lactate dehydrogenase enzymatic activities, restoring all values to wild-type levels. Chronic supplementation with tempol recovers force-generating capacity and metabolic enzyme activity in mdx diaphragm. These findings may have relevance in the search for therapeutic strategies in neuromuscular disease.
Collapse
|
45
|
Jiang LG, Cheng LY, Kong SH, Yang Y, Shen YJ, Chen C, Deng XH, Liu SZ, Chao L. Toxic effects of polychlorinated biphenyls (Aroclor 1254) on human sperm motility. Asian J Androl 2017; 19:561-566. [PMID: 27624986 PMCID: PMC5566850 DOI: 10.4103/1008-682x.186876] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/31/2016] [Accepted: 06/16/2016] [Indexed: 01/08/2023] Open
Abstract
Polychlorinated biphenyls (PCBs) are common environmental contaminants that represent a considerable risk to reproductive toxicity in exposed human populations. Although some experimental studies have suggested an association between the levels of PCBs and semen quality, the direct effects of PCBs on human sperm parameters remain largely unexplored. To this aim, a short-term in vitro incubation experiment that better imitated the putative exposure of sperm to Aroclor 1254 (a commercial PCB mixture) in male reproduction tissue was conducted. Human sperm were incubated with various concentrations (0, 1, 5, or 25 mg l-1) of Aroclor 1254 for different amounts of time (3 and 6 h) in vitro. Sperm motility parameters were analyzed with computer-assisted sperm analysis (CASA). The proportion of sperm with high mitochondrial membrane potential (ΔΨm) and the levels of intracellular reactive oxygen species (ROS) were detected to explore the probable cause of sperm impairment. Human sperm exposed to continuous Aroclor 1254 exhibited: (i) reduced sperm motility and kinematic parameters, (ii) a proportion of sperm with high ΔΨm that decreased in a dose-dependent manner (P < 0.05), and (iii) increased levels of ROS compared with controls (P < 0.05). In conclusion, Aroclor 1254 can decrease sperm motility, which may culminate in increased ROS and general mitochondrial dysfunction, thus affecting the fertilization potential of sperm. Our findings suggest a broader understanding of the effect of Aroclor 1254 on human sperm.
Collapse
Affiliation(s)
- Li-Gang Jiang
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| | - Lai-Yang Cheng
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| | - Shu-Hui Kong
- College of Life Science, Shandong University, Jinan 250014, Shandong, China
| | - Yang Yang
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| | - Yan-Jun Shen
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| | - Chao Chen
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| | - Xiao-Hui Deng
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| | - Shu-Zhen Liu
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, Jinan 250014, Shandong, China
| | - Lan Chao
- Infertility Center, Qilu Hospital of Shandong University, Jinan 250014, Shandong, China
| |
Collapse
|
46
|
González-Barriga A, Nillessen B, Kranzen J, van Kessel IDG, Croes HJE, Aguilera B, de Visser PC, Datson NA, Mulders SAM, van Deutekom JCT, Wieringa B, Wansink DG. Intracellular Distribution and Nuclear Activity of Antisense Oligonucleotides After Unassisted Uptake in Myoblasts and Differentiated Myotubes In Vitro. Nucleic Acid Ther 2017; 27:144-158. [PMID: 28375678 PMCID: PMC5467152 DOI: 10.1089/nat.2016.0641] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Clinical efficacy of antisense oligonucleotides (AONs) for the treatment of neuromuscular disorders depends on efficient cellular uptake and proper intracellular routing to the target. Selection of AONs with highest in vitro efficiencies is usually based on chemical or physical methods for forced cellular delivery. Since these methods largely bypass existing natural mechanisms for membrane passage and intracellular trafficking, spontaneous uptake and distribution of AONs in cells are still poorly understood. Here, we report on the unassisted uptake of naked AONs, so-called gymnosis, in muscle cells in culture. We found that gymnosis works similarly well for proliferating myoblasts as for terminally differentiated myotubes. Cell biological analyses combined with microscopy imaging showed that a phosphorothioate backbone promotes efficient gymnosis, that uptake is clathrin mediated and mainly results in endosomal-lysosomal accumulation. Nuclear localization occurred at a low level, but the gymnotically delivered AONs effectively modulated the expression of their nuclear RNA targets. Chloroquine treatment after gymnotic delivery helped increase nuclear AON levels. In sum, we demonstrate that gymnosis is feasible in proliferating and non-proliferating muscle cells and we confirm the relevance of AON chemistry for uptake and intracellular trafficking with this method, which provides a useful means for bio-activity screening of AONs in vitro.
Collapse
Affiliation(s)
- Anchel González-Barriga
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands .,2 BioMarin Nederland B.V., Leiden, the Netherlands
| | - Bram Nillessen
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Julia Kranzen
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Ingeborg D G van Kessel
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Huib J E Croes
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | | | | | | | | | | | - Bé Wieringa
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Derick G Wansink
- 1 Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| |
Collapse
|
47
|
Johnstone VPA, Viola HM, Hool LC. Dystrophic Cardiomyopathy-Potential Role of Calcium in Pathogenesis, Treatment and Novel Therapies. Genes (Basel) 2017; 8:genes8040108. [PMID: 28338606 PMCID: PMC5406855 DOI: 10.3390/genes8040108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by defects in the DMD gene and results in progressive wasting of skeletal and cardiac muscle due to an absence of functional dystrophin. Cardiomyopathy is prominent in DMD patients, and contributes significantly to mortality. This is particularly true following respiratory interventions that reduce death rate and increase ambulation and consequently cardiac load. Cardiomyopathy shows an increasing prevalence with age and disease progression, and over 95% of patients exhibit dilated cardiomyopathy by the time they reach adulthood. Development of the myopathy is complex, and elevations in intracellular calcium, functional muscle ischemia, and mitochondrial dysfunction characterise the pathophysiology. Current therapies are limited to treating symptoms of the disease and there is therefore an urgent need to treat the underlying genetic defect. Several novel therapies are outlined here, and the unprecedented success of phosphorodiamidate morpholino oligomers (PMOs) in preclinical and clinical studies is overviewed.
Collapse
Affiliation(s)
- Victoria P A Johnstone
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia.
| | - Helena M Viola
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia.
| | - Livia C Hool
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia.
- Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia.
| |
Collapse
|
48
|
Arvanitidis A, Henriksen K, Karsdal M, Nedergaard A. Neo-epitope Peptides as Biomarkers of Disease Progression for Muscular Dystrophies and Other Myopathies. J Neuromuscul Dis 2016; 3:333-346. [PMID: 27854226 PMCID: PMC5123625 DOI: 10.3233/jnd-160150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For several decades, serological biomarkers of neuromuscular diseases as dystrophies, myopathies and myositis have been limited to routine clinical biochemistry panels. Gauging the pathological progression is a prerequisite for proper treatment and therefore identifying accessible, easy to monitor biomarkers that can predict the disease progression would be an important advancement. Most muscle diseases involve accelerated muscle fiber degradation, inflammation, fatty tissue substitution and/or fibrosis. All these pathological traits have been shown to give rise to serological peptide biomarkers in other tissues, underlining the potential application of existing biomarkers of such traits in muscle disorders. A significant quantity of tissue is involved in these pathological mechanisms alongside with qualitative changes in protein turnover in myofibrillar, extra-cellular matrix and immunological cell protein fractions accompanied by alterations in body fluids. We propose that protein and peptides can leak out of the afflicted muscles and can be of use in diagnosis, prediction of pathology trajectory and treatment efficacy. Proteolytic cleavage systems are especially modulated during a range of muscle pathologies, thereby giving rise to peptides that are differentially released during disease manifestation. Therefore, we believe that pathology-specific post-translational modifications like cleavages can give rise to neoepitope peptides that may represent a promising class of peptides for discovery of biomarkers pertaining to neuromuscular diseases.
Collapse
Affiliation(s)
- A. Arvanitidis
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - K. Henriksen
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - M.A. Karsdal
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - A. Nedergaard
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| |
Collapse
|
49
|
Wang C, Wang H, Wu D, Hu J, Wu W, Zhang Y, Peng X. A novel perspective for burn-induced myopathy: Membrane repair defect. Sci Rep 2016; 6:31409. [PMID: 27545095 PMCID: PMC4992861 DOI: 10.1038/srep31409] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 07/21/2016] [Indexed: 12/25/2022] Open
Abstract
Myopathy is a common complication of severe burn patients. One potential cause of this myopathy could be failure of the plasma membrane to undergo repair following injuries generated from toxin or exercise. The aim of this study is to assess systemic effect on muscle membrane repair deficiency in burn injury. Skeletal muscle fibers isolated from burn-injured mice were damaged with a UV laser and dye influx imaged confocally to evaluate membrane repair capacity. Membrane repair failure was also tested in burn-injured mice subjected to myotoxin or treadmill exercise. We further used C2C12 myotubules and animal models to investigate the role of MG53 in development of burn-induced membrane repair defect. We demonstrated that skeletal muscle myofibers in burn-injured mice showed significantly more dye uptake after laser damage than controls, indicating a membrane repair deficiency. Myotoxin or treadmill exercise also resulted in a higher-grade repair defect in burn-injured mice. Furthermore, we observed that burn injury induced a significant decrease in MG53 levels and its dimerization in skeletal muscles. Our findings highlight a new mechanism that implicates membrane repair failure as an underlying cause of burn-induced myopathy. And, the disorders in MG53 expression and MG53 dimerization are involved in this cellular pathology.
Collapse
Affiliation(s)
- Chao Wang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Hongyu Wang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Dan Wu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jianhong Hu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Wu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xi Peng
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
50
|
Horvath DM, Murphy RM, Mollica JP, Hayes A, Goodman CA. The effect of taurine and β-alanine supplementation on taurine transporter protein and fatigue resistance in skeletal muscle from mdx mice. Amino Acids 2016; 48:2635-2645. [PMID: 27444300 DOI: 10.1007/s00726-016-2292-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/30/2016] [Indexed: 11/29/2022]
Abstract
This study investigated the effect of taurine and β-alanine supplementation on muscle function and muscle taurine transporter (TauT) protein expression in mdx mice. Wild-type (WT) and mdx mice (5 months) were supplemented with taurine or β-alanine for 4 weeks, after which in vitro contractile properties, fatigue resistance and force recovery, and the expression of the TauT protein and proteins involved in excitation-contraction (E-C) coupling were examined in fast-twitch muscle. There was no difference in basal TauT protein expression or basal taurine content between mdx than WT muscle. Supplementation with taurine and β-alanine increased and reduced taurine content, respectively, in muscle from WT and mdx mice but had no effect of TauT protein. Taurine supplementation reduced body and muscle mass, and enhanced fatigue resistance and force recovery in mdx muscle. β-Alanine supplementation enhanced fatigue resistance in WT and mdx muscle. There was no difference in the basal expression of key E-C coupling proteins [ryanodine receptor 1 (RyR1), dihydropyridine receptor (DHPR), sarco(endo)plasmic reticulum Ca2+-ATPase 1 (SERCA1) or calsequestrin 1 (CSQ1)] between WT and mdx mice, and the expression of these proteins was not altered by taurine or β-alanine supplementation. These findings suggest that TauT protein expression is relatively insensitive to changes in muscle taurine content in WT and mdx mice, and that taurine and β-alanine supplementation may be viable therapeutic strategies to improve fatigue resistance of dystrophic skeletal muscle.
Collapse
Affiliation(s)
- Deanna M Horvath
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC, 3086, Australia.
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Janelle P Mollica
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Alan Hayes
- Centre for Chronic Disease Prevention and Management, Victoria University, Melbourne, Australia.,Institute of Sport, Exercise and Active Living, Victoria University, Melbourne, Australia.,Australian Institute for Musculoskeletal Science, Western Health, Melbourne, VIC, Australia
| | - Craig A Goodman
- Centre for Chronic Disease Prevention and Management, Victoria University, Melbourne, Australia.,Institute of Sport, Exercise and Active Living, Victoria University, Melbourne, Australia
| |
Collapse
|